1
|
Gupta H, Raghubansi A, Bharat, Sharma K, Zutshi K, Panchal P, Bhattacharya S, Ranjan P, Puri G, Saini N. Targeting GSK3β and signaling pathways in breast cancer: role of individual members of miR- 23/24/27 cluster. BMC Cancer 2025; 25:737. [PMID: 40254586 PMCID: PMC12010543 DOI: 10.1186/s12885-025-14045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/31/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND The high mortality rate of breast cancer and the difficulties associated with therapeutic resistance, especially in cases where targeted treatments are unavailable, make it a serious threat to women's health. This study examines the relationship between three mature microRNAs (miRNAs) that are clustered together, namely miR- 23a, miR- 27a, and miR- 24-2, as well as their potential correlation with breast cancer. METHODS We identified common gene targets of miR- 23a, miR- 27a, and miR- 24-2 using computational analysis. We also checked for the levels of miR- 23a, miR- 27a, and miR- 24-2 in 26 breast tumor tissues (with their matched control) as well as MCF7 and MDA-MB- 231 cell lines using qRT-PCR. Dual-luciferase reporter assay was conducted to validate the binding site of the microRNAs in their target gene. Western blot was performed to study the expression of various breast cancer related genes in the presence of the three microRNAs. In addition, the effect of microRNAs in cancer cell metastasis and cell division was carried out using invasion and cell cycle assay. RESULTS Computational analysis identified key genes, including GSK3β, NCOA1 and SP1, which are functionally linked to tumor progression and various other malignancies. All three microRNAs were found to be significantly downregulated in the breast cancer tissue samples in comparison to their respective controls. Kaplan-Meier plot analysis revealed that the expression levels of these genes and associated microRNAs correlates with breast cancer patient survival rates. Reduced SP1 and NCOA1 levels predicted a worse prognosis, but elevated levels of GSK3β were linked with decreased survival. Moreover, miR- 23a and miR- 24-2 specifically target GSK3β, potentially disrupting the Wnt/β-catenin pathway involved in breast cancer development. Functional tests showed that miR- 23a, miR- 27a and miR- 24-2 affect expression of EMT related genes, influencing cell invasion and migration, impacting ERK signaling and EMT, critical in the spread of breast cancer. CONCLUSION This study unlocks the potential of targeting the microRNA cluster as a therapeutic approach and emphasizes the complex regulatory roles of each individual members of the miR- 23a/27a/24-2 cluster in the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Harshi Gupta
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
| | - Anushka Raghubansi
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
| | - Bharat
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kritika Sharma
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
| | - Krittika Zutshi
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
| | - Partibha Panchal
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sushant Bhattacharya
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Piyush Ranjan
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Gopal Puri
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Neeru Saini
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi, 110007, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Mardi A, Ghovahi A, Abbasvandi F, Amani D. Experimental Validation of miR-4443, miR-572, and miR-150-5p in Serum and Tissue of Breast Cancer Patients as a Potential Diagnostic Biomarker: A Study Based on Bioinformatics Prediction. Biochem Genet 2025:10.1007/s10528-025-11057-8. [PMID: 40064800 DOI: 10.1007/s10528-025-11057-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/08/2025] [Indexed: 04/10/2025]
Abstract
Breast cancer is the most common invasive cancer diagnosed in females and is also the main cause of cancer-related deaths leading to more than 500,000 deaths annually. The present study aims to identify a promising panel of microRNAs (miRNAs) using bioinformatics analysis, and to clinically validate their utility for diagnosing breast cancer patients with high accuracy in a clinical setting. First, in the in silico phase of our study, using bioinformatics analysis and the data available in the GEO database, miRNAs that were increased in the interstitial fluid of the tumor tissues (differentially expressed miRNAs), were screened and their related target genes were selected. Multimir package of R software was utilized to determine the target genes of the differentially expressed miRNAs (DEMs). The biological functions of discovered genes were analyzed using Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. In order to determine the molecular mechanisms behind important signaling pathways and cellular functions, the protein-protein interaction network was built using STRING and Cytoscape software. After that, in the laboratory phase, the expression level of three candidate miRNAs on the serum samples of 26 breast cancer patients and 26 control, as well as 14 tumor tissue samples and 14 adjacent normal tissue samples, has been investigated by Real-time PCR method. Then sensitivity and specificity of candidate miRNAs were evaluated through the ROC curve analysis. After in silico analysis, we revealed that three miRNAs including miR-4443, miR-572, and miR-150-5p were highly increased in the interstitial fluid of breast cancer patients compared to breast cancer tissues. Moreover, our results revealed that the expression level of miR-4443, miR-572, and miR-150-5p were significantly decreased in the serum of breast cancer patients compare to normal controls. Also, the expression level of miR-4443 and miR-150-5p was significantly decreased in the tumor tissue compared to the adjacent non-tumor tissue. Also, ROC curve analysis showed that these three miRNAs have high sensitivity and specificity for the diagnosis of breast cancer patients. Data analysis was conducted with GraphPad Prism software. Our findings suggest the potential utility of measuring tumor-derived miRNAs in serum as an important approach for the blood-based detection of breast cancer patients. It appears that miR-4443, miR-572, and miR-150-5p may serve as promising diagnostic biomarkers with high sensitivity and specificity. However, it's important to note that further research will be needed to definitively establish the use of these miRNAs as potential biomarkers in clinical practice.
Collapse
Affiliation(s)
- Amirhossein Mardi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Ghovahi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Abbasvandi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX: 15179/64311, Tehran, Iran
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davar Amani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Halvoník A, Moravčíková N, Vostrý L, Vostra-Vydrova H, Mészáros G, Demir E, Chalupková M, Kasarda R. Heterozygosity-Rich Regions in Canine Genome: Can They Serve as Indicators of Balancing Selection? Animals (Basel) 2025; 15:612. [PMID: 40003092 PMCID: PMC11851536 DOI: 10.3390/ani15040612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Compared to the negative effect of directional selection on genetic diversity, balancing selection acts oppositely and maintains variability across the genome. This study aims to articulate whether balancing selection leads to heterozygosity-rich region islands (HRRIs) forming in the canine genome by investigating 1000 animals belonging to 50 dog breeds via 153,733 autosomal SNPs. A consecutive SNP-based approach was used to identify heterozygosity-rich regions (HRRs). Signals of balancing selection in the genome of studied breeds were then assessed with Tajima's D statistics. A total of 72,062 HRRs with an average length of 324 kb were detected to be unevenly distributed across the genome. A total of 509 and 450 genomic regions were classified as HRRIs and balancing selection signals, respectively. Although the genome-wide distributions of HRRIs varied across breeds, several HRRIs were found in the same locations across multiple breeds. A total of 109 genomic regions were classified as both HRRIs and signals of balancing selection. Even though the genomic coordinates of HRRIs and balancing selection signals did not fully overlap across all genomic regions, balancing selection may play a significant role in maintaining diversity in regions associated with various cancer diseases, immune response, and bone, skin, and cartilage tissue development.
Collapse
Affiliation(s)
- Adrián Halvoník
- Institute of Nutrition and Genomics, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 94976 Nitra, Slovakia; (M.C.); (R.K.)
| | - Nina Moravčíková
- Institute of Nutrition and Genomics, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 94976 Nitra, Slovakia; (M.C.); (R.K.)
| | - Luboš Vostrý
- Department of Genetics and Breeding, Czech University of Life Sciences Prague, Kamýcká 129, Praha-Suchdol, 165 00 Prague, Czech Republic; (L.V.); (H.V.-V.)
| | - Hana Vostra-Vydrova
- Department of Genetics and Breeding, Czech University of Life Sciences Prague, Kamýcká 129, Praha-Suchdol, 165 00 Prague, Czech Republic; (L.V.); (H.V.-V.)
| | - Gábor Mészáros
- Institute of Livestock Sciences, BOKU University, Gregor-Mendel-Straße 33, 1180 Vienna, Austria;
| | - Eymen Demir
- Department of Animal Science, Faculty of Agriculture, Akdeniz University, Antalya 07070, Türkiye;
| | - Monika Chalupková
- Institute of Nutrition and Genomics, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 94976 Nitra, Slovakia; (M.C.); (R.K.)
| | - Radovan Kasarda
- Institute of Nutrition and Genomics, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 94976 Nitra, Slovakia; (M.C.); (R.K.)
| |
Collapse
|
4
|
Vicente-Muñoz S, Davis JC, Lane A, Lane AN, Waltz SE, Wells SI. Lipid profiling of RON and DEK-dependent signaling in breast cancer guides discovery of gene networks predictive of poor outcomes. Front Oncol 2024; 14:1382986. [PMID: 39351361 PMCID: PMC11440356 DOI: 10.3389/fonc.2024.1382986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/30/2024] [Indexed: 10/04/2024] Open
Abstract
Recurrent and metastatic breast cancer is frequently treatment resistant. A wealth of evidence suggests that reprogrammed lipid metabolism supports cancer recurrence. Overexpression of the RON and DEK oncoproteins in breast cancer is associated with poor outcome. Both proteins promote cancer metastasis in laboratory models, but their influence on lipid metabolite levels remain unknown. To measure RON- and DEK-dependent steady-state lipid metabolite levels, a nuclear magnetic resonance (NMR)-based approach was utilized. The observed differences identified a lipid metabolism-related gene expression signature that is prognostic of overall survival (OS), distant metastasis-free survival (DMFS), post-progression survival (PPS), and recurrence-free survival (RFS) in patients with breast cancer. RON loss led to decreased cholesterol and sphingomyelin levels, whereas DEK loss increased total fatty acid levels and decreased free glycerol levels. Lipid-related genes were then queried to define a signature that predicts poor outcomes for patients with breast cancer patients. Taken together, RON and DEK differentially regulate lipid metabolism in a manner that predicts and may promote breast cancer metastasis and recurrence.
Collapse
Affiliation(s)
- Sara Vicente-Muñoz
- Translational Metabolomics Facility, Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
| | - James C. Davis
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Andrew N. Lane
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, United States
| | - Susan E. Waltz
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
- Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH, United States
| | - Susanne I. Wells
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, United States
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
5
|
Feng G, Zhang H, Liu H, Zhang X, Jiang H, Liao S, Luo X, Yao H, Xiang B, Liu S, Zhang J, Zhang J, Fang J. Natural Flavonoid-Derived Enzyme Mimics DHKNase Balance the Two-Edged Reactive Oxygen Species Function for Wound Healing and Inflammatory Bowel Disease Therapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0464. [PMID: 39253100 PMCID: PMC11381673 DOI: 10.34133/research.0464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Rational regulation of reactive oxygen species (ROS) plays a vital importance in maintaining homeostasis of living biological systems. For ROS-related pathologies, chemotherapy technology derived from metal nanomaterials currently occupies a pivotal position. However, they suffer from inherent issues such as complicated synthesis, batch-to-batch variability, high cost, and potential biological toxicity caused by metal elements. Here, we reported for the first time that dual-action 3,5-dihydroxy-1-ketonaphthalene-structured small-molecule enzyme imitator (DHKNase) exhibited 2-edged ROS regulation, catering to the execution of physiology-beneficial ROS destiny among diverse pathologies in living systems. Based on this, DHKNase is validated to enable remarkable therapeutic effects in 2 classic disease models, including the pathogen-infected wound-healing model and the dextran sulfate sodium (DSS)-caused inflammatory bowel disease (IBD). This work provides a guiding landmark for developing novel natural small-molecule enzyme imitator and significantly expands their application potential in the biomedical field.
Collapse
Affiliation(s)
- Guangfu Feng
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Huaizu Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Huipeng Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Xiaoyan Zhang
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Hongmei Jiang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Sijie Liao
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Xingyu Luo
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, P.R. China
| | - Hao Yao
- Changsha IMADEK Intelligent Technology Co. Ltd., Changsha, Hunan 410081, P.R. China
| | - Bo Xiang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Shiyu Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Jiali Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Jiaheng Zhang
- College of Chemistry, Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jun Fang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| |
Collapse
|
6
|
Zhou L, Liu J, Zhou M, Xu L. Serum Macrophage Colony-Stimulating Factor Relates to the Severity and the Pregnancy Outcomes in Hypertensive Disorders Complicating Pregnancy. Med Princ Pract 2024; 33:462-470. [PMID: 38830347 PMCID: PMC11460953 DOI: 10.1159/000539619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
PURPOSE This study aimed to assess the predictive value of macrophage colony-stimulating factor (M-CSF) in the first trimester for hypertensive disorders complicating pregnancy (HDCP) and its association with disease severity and adverse pregnancy outcomes. HDCP pose significant risks to both maternal health and fetal health. M-CSF is implicated in the pathogenesis of HDCP by promoting inflammation and endothelial damage. METHODS Serum levels of M-CSF were measured using an enzyme-linked immunosorbent assay, and clinical characteristics and pregnancy outcomes were compared between groups. RESULTS Pregnant women with HDCP had significantly higher levels of proteinuria, systolic blood pressure, and diastolic blood pressure compared to those with normal pregnancy. Among patients with HDCP, the severity of disease correlated positively with serum levels of M-CSF. Furthermore, M-CSF levels in the first trimester were significantly associated with adverse pregnancy outcomes. The findings suggest that M-CSF may serve as a potential biomarker for predicting HDCP and its severity, as well as adverse pregnancy outcomes. CONCLUSIONS Early detection and monitoring of M-CSF levels could aid in identifying high-risk pregnancies and implementing appropriate interventions to improve maternal and fetal outcomes.
Collapse
Affiliation(s)
- Lili Zhou
- Department of Gynecology and Obstetrics, Changchun University of Chinese Medicine Affiliated Hospital, Jilin, China
| | - Junbo Liu
- Department of Gynecology, Changchun University of Chinese Medicine Affiliated Hospital, Jilin, China
| | - Min Zhou
- Department of Gynecology, Changchun University of Chinese Medicine Affiliated Hospital, Jilin, China
| | - Lan Xu
- Department of Gynecology and Obstetrics, Changchun University of Chinese Medicine Affiliated Hospital, Jilin, China
| |
Collapse
|
7
|
Tong Z, Zhang Y, Guo P, Wang W, Chen Q, Jin J, Liu S, Yu C, Mo P, Zhang L, Huang J. Steroid receptor coactivator 1 promotes human hepatocellular carcinoma invasiveness through enhancing MMP-9. J Cell Mol Med 2024; 28:e18171. [PMID: 38506084 PMCID: PMC10951881 DOI: 10.1111/jcmm.18171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 03/21/2024] Open
Abstract
SRC-1 functions as a transcriptional coactivator for steroid receptors and various transcriptional factors. Notably, SRC-1 has been implicated in oncogenic roles in multiple cancers, including breast cancer and prostate cancer. Previous investigations from our laboratory have established the high expression of SRC-1 in human HCC specimens, where it accelerates HCC progression by enhancing Wnt/beta-catenin signalling. In this study, we uncover a previously unknown role of SRC-1 in HCC metastasis. Our findings reveal that SRC-1 promotes HCC metastasis through the augmentation of MMP-9 expression. The knockdown of SRC-1 effectively mitigated HCC cell metastasis both in vitro and in vivo by suppressing MMP-9 expression. Furthermore, we observed a positive correlation between SRC-1 mRNA levels and MMP-9 mRNA levels in limited and larger cohorts of HCC specimens from GEO database. Mechanistically, SRC-1 operates as a coactivator for NF-κB and AP-1, enhancing MMP-9 promoter activity in HCC cells. Higher levels of SRC-1 and MMP-9 expression are associated with worse overall survival in HCC patients. Treatment with Bufalin, known to inhibit SRC-1 expression, significantly decreased MMP-9 expression and inhibited HCC metastasis in both in vitro and in vivo settings. Our results demonstrated the pivotal role of SRC-1 as a critical modulator in HCC metastasis, presenting a potential therapeutic target for HCC intervention.
Collapse
Affiliation(s)
- Zhangwei Tong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
- Department of Molecular and Cellular BiologyBaylor College of MedicineHoustonTexasUSA
| | - Yong Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Peng Guo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Wei Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Qiang Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Jing Jin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Shixiao Liu
- Department of Cardiology, School of MedicineThe First Affiliated Hospital of Xiamen University, Xiamen UniversityXiamenChina
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life SciencesXiamen UniversityXiamenChina
| | - Lei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical SciencesShanxi Medical University; Shanxi Tongji Hospital, Huazhong University of Science and TechnologyTaiyuanChina
| | - Junli Huang
- Department of General SurgeryArmy 73rd Group Military Hospital of the Chinese People's Liberation Army (Chenggong Hospital of Xiamen University)XiamenChina
| |
Collapse
|
8
|
Chen Q, Guo P, Hong Y, Mo P, Yu C. The multifaceted therapeutic value of targeting steroid receptor coactivator-1 in tumorigenesis. Cell Biosci 2024; 14:41. [PMID: 38553750 PMCID: PMC10979636 DOI: 10.1186/s13578-024-01222-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024] Open
Abstract
Steroid receptor coactivator-1 (SRC-1, also known as NCOA1) frequently functions as a transcriptional coactivator by directly binding to transcription factors and recruiting to the target gene promoters to promote gene transcription by increasing chromatin accessibility and promoting the formation of transcriptional complexes. In recent decades, various biological and pathological functions of SRC-1 have been reported, especially in the context of tumorigenesis. SRC-1 is a facilitator of the progression of multiple cancers, including breast cancer, prostate cancer, gastrointestinal cancer, neurological cancer, and female genital system cancer. The emerging multiorgan oncogenic role of SRC-1 is still being studied and may not be limited to only steroid hormone-producing tissues. Growing evidence suggests that SRC-1 promotes target gene expression by directly binding to transcription factors, which may constitute a novel coactivation pattern independent of AR or ER. In addition, the antitumour effect of pharmacological inhibition of SRC-1 with agents including various small molecules or naturally active compounds has been reported, but their practical application in clinical cancer therapy is very limited. For this review, we gathered typical evidence on the oncogenic role of SRC-1, highlighted its major collaborators and regulatory genes, and mapped the potential mechanisms by which SRC-1 promotes primary tumour progression.
Collapse
Affiliation(s)
- Qiang Chen
- Zhejiang Key Laboratory of Pathophysiology, Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China.
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China.
| | - Peng Guo
- Department of Cell Biotechnology Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300308, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361104, China
| | - Yilin Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361104, China
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361104, China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361104, China.
| |
Collapse
|
9
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [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: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
10
|
Wang Y, Gao P, Hao Z, Chen L, Li X, Jiao Y, Liu J, Li J, Zhang Y, Peng X, Ning B, Zhan X. The effect of neoadjuvant chemotherapy on the tumor immune microenvironment in gastrointestinal tumors. Front Oncol 2022; 12:1054598. [DOI: 10.3389/fonc.2022.1054598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022] Open
Abstract
In recent years, numerous studies have demonstrated that the tumor immune microenvironment (TIME) is capable of regulating the growth of tumors, and tumor-infiltrating immune cells in the TIME can affect the prognosis and treatment responses of patients. Consequently, therapies targeting these immune cells have emerged as important antitumor treatments. As a crucial componet of the perioperative treatment of malignant tumors, neoadjuvant chemotherapy (NACT) can improve the surgical resection rate and prognosis of patients and is a suitable clinical model to evaluate the effect of chemotherapy on the TIME. To provide a rationale for developing valid combinational therapies, this review summarizes the impact of NACT on the TIME, the relationship between tumor-infiltrating immune cells and treatment responses of patients, and the prognostic value of these infiltrating immune cells.
Collapse
|
11
|
Yadav V, Sharma K, Bhattacharya S, Talwar P, Purohit PK, Saini N. RETRACTED: hsa-miR-23a~27a~24-2 cluster members inhibit aggressiveness of breast cancer cells by commonly targeting NCOA1, NLK and RAP1B. Life Sci 2022; 307:120906. [PMID: 36007610 DOI: 10.1016/j.lfs.2022.120906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. The corresponding author notified the journal of three examples of image duplication within the published article (two in Figure 3D and one in Figure 4A), and requested a corrigendum. As per journal policy when considering corrigendum requests, the journal requested the authors to provide source data relating to these affected figures. The editorial team noticed 12 additional suspected image duplications within the supplied source data and the corresponding author was informed. Upon submission of revised source data, the editorial team noticed two new suspected image duplications. The editorial team have concerns about the provenance of the data and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Vikas Yadav
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Kritika Sharma
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Sushant Bhattacharya
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India
| | - Puneet Talwar
- Institute of Human Behaviour & Allied Sciences (IHBAS), Delhi, India
| | - Paresh Kumar Purohit
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neeru Saini
- Functional Genomics Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, 110007, Delhi, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
12
|
Tan YD. Two-sample t α -test for testing hypotheses in small-sample experiments. Int J Biostat 2022:ijb-2021-0047. [PMID: 35749155 DOI: 10.1515/ijb-2021-0047] [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: 06/02/2021] [Accepted: 05/29/2022] [Indexed: 11/15/2022]
Abstract
It has been reported that about half of biological discoveries are irreproducible. These irreproducible discoveries were partially attributed to poor statistical power. The poor powers are majorly owned to small sample sizes. However, in molecular biology and medicine, due to the limit of biological resources and budget, most molecular biological experiments have been conducted with small samples. Two-sample t-test controls bias by using a degree of freedom. However, this also implicates that t-test has low power in small samples. A discovery found with low statistical power suggests that it has a poor reproducibility. So, promotion of statistical power is not a feasible way to enhance reproducibility in small-sample experiments. An alternative way is to reduce type I error rate. For doing so, a so-called t α -test was developed. Both theoretical analysis and simulation study demonstrate that t α -test much outperforms t-test. However, t α -test is reduced to t-test when sample sizes are over 15. Large-scale simulation studies and real experiment data show that t α -test significantly reduced type I error rate compared to t-test and Wilcoxon test in small-sample experiments. t α -test had almost the same empirical power with t-test. Null p-value density distribution explains why t α -test had so lower type I error rate than t-test. One real experimental dataset provides a typical example to show that t α -test outperforms t-test and a microarray dataset showed that t α -test had the best performance among five statistical methods. In addition, the density distribution and probability cumulative function of t α -statistic were given in mathematics and the theoretical and observed distributions are well matched.
Collapse
Affiliation(s)
- Yuan-De Tan
- Statistics core, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
13
|
Anti-Cancer Effects of Dietary Polyphenols via ROS-Mediated Pathway with Their Modulation of MicroRNAs. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27123816. [PMID: 35744941 PMCID: PMC9227902 DOI: 10.3390/molecules27123816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 12/23/2022]
Abstract
Consumption of coffee, tea, wine, curry, and soybeans has been linked to a lower risk of cancer in epidemiological studies. Several cell-based and animal studies have shown that dietary polyphenols like chlorogenic acid, curcumin, epigallocatechin-3-O-gallate, genistein, quercetin and resveratrol play a major role in these anticancer effects. Several mechanisms have been proposed to explain the anticancer effects of polyphenols. Depending on the cellular microenvironment, these polyphenols can exert double-faced actions as either an antioxidant or a prooxidant, and one of the representative anticancer mechanisms is a reactive oxygen species (ROS)-mediated mechanism. These polyphenols can also influence microRNA (miR) expression. In general, they can modulate the expression/activity of the constituent molecules in ROS-mediated anticancer pathways by increasing the expression of tumor-suppressive miRs and decreasing the expression of oncogenic miRs. Thus, miR modulation may enhance the anticancer effects of polyphenols through the ROS-mediated pathways in an additive or synergistic manner. More precise human clinical studies on the effects of dietary polyphenols on miR expression will provide convincing evidence of the preventive roles of dietary polyphenols in cancer and other diseases.
Collapse
|
14
|
Guo P, Chen Q, Peng K, Xie J, Liu J, Ren W, Tong Z, Li M, Xu J, Zhang Y, Yu C, Mo P. Nuclear receptor coactivator SRC-1 promotes colorectal cancer progression through enhancing GLI2-mediated Hedgehog signaling. Oncogene 2022; 41:2846-2859. [PMID: 35418691 DOI: 10.1038/s41388-022-02308-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 01/20/2023]
Abstract
Overexpression of nuclear coactivator steroid receptor coactivator 1 (SRC-1) and aberrant activation of the Hedgehog (Hh) signaling pathway are associated with various tumorigenesis; however, the significance of SRC-1 in colorectal cancer (CRC) and its contribution to the activation of Hh signaling are unclear. Here, we identified a conserved Hh signaling signature positively correlated with SRC-1 expression in CRC based on TCGA database; SRC-1 deficiency significantly inhibited the proliferation, survival, migration, invasion, and tumorigenesis of both human and mouse CRC cells, and SRC-1 knockout significantly suppressed azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC in mice. Mechanistically, SRC-1 promoted the expression of GLI family zinc finger 2 (GLI2), a major downstream transcription factor of Hh pathway, and cooperated with GLI2 to enhance multiple Hh-regulated oncogene expression, including Cyclin D1, Bcl-2, and Slug. Pharmacological blockages of SRC-1 and Hh signaling retarded CRC progression in human CRC cell xenograft mouse model. Together, our studies uncover an SRC-1/GLI2-regulated Hh signaling looping axis that promotes CRC tumorigenesis, offering an attractive strategy for CRC treatment.
Collapse
Affiliation(s)
- Peng Guo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qiang Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kesong Peng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.,Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200433, China
| | - Jianyuan Xie
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Junjia Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.,National Institute for Data Science in Health and Medicine Engineering, Research Center of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenjing Ren
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhangwei Tong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ming Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yongyou Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China. .,National Institute for Data Science in Health and Medicine Engineering, Research Center of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
15
|
Martinez JD, Mo Q, Xu Y, Qin L, Li Y, Xu J. Common Genomic Aberrations in Mouse and Human Breast Cancers with Concurrent P53 Deficiency and Activated PTEN-PI3K-AKT Pathway. Int J Biol Sci 2022; 18:229-241. [PMID: 34975329 PMCID: PMC8692138 DOI: 10.7150/ijbs.65763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/20/2021] [Indexed: 11/27/2022] Open
Abstract
Simultaneous P53 loss and activation of the PTEN-restricted PI3K-AKT pathway frequently occur in aggressive breast cancers. P53 loss causes genome instability, while PTEN loss and/or activating mutations of PIK3CA and AKT promote cancer cell proliferation that also increases incidences of genomic aberrations. However, the genomic alterations associated with P53 loss and activated PTEN-PI3K-AKT signaling in breast cancer have not been defined. Spatiotemporally controlled breast cancer models with inactivation of both P53 and Pten in adult mice have not been established for studying genomic alterations. Herein, we deleted both floxed Pten and Tp53 genes in the mammary gland epithelial cells in adult mice using a RCAS virus-mediated Cre-expressing system. These mice developed small tumors in 21 weeks, and poorly differentiated larger tumors in 26 weeks. In these tumors, we identified 360 genes mutated by nonsynonymous point mutations and small insertions and deletions (NSPMs/InDels), 435 genes altered by copy number amplifications (CNAs), and 450 genes inactivated by copy number deletions (CNDs). Importantly, 22.2%, 75.9% and 27.3% of these genes were also altered in human breast tumors with P53 and PTEN losses or P53 loss and activated PI3K-AKT signaling by NSPMs/InDels, CNAs and CNDs, respectively. Therefore, inactivation of P53 and Pten in adult mice causes rapid-growing breast tumors, and these tumors recapitulate a significant number of genetic aberrations in human breast tumors with inactivated P53 and activated PTEN-PI3K-AKT signaling. Further characterization of these commonly altered genes in breast cancer should help to identify novel cancer-driving genes and molecular targets for developing therapeutics.
Collapse
Affiliation(s)
- Jarrod D Martinez
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Qianxing Mo
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Yixiang Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Li Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Yi Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
16
|
Huang C, Ou R, Chen X, Zhang Y, Li J, Liang Y, Zhu X, Liu L, Li M, Lin D, Qiu J, Liu G, Zhang L, Wu Y, Tang H, Liu Y, Liang L, Ding Y, Liao W. Tumor cell-derived SPON2 promotes M2-polarized tumor-associated macrophage infiltration and cancer progression by activating PYK2 in CRC. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:304. [PMID: 34583750 PMCID: PMC8477524 DOI: 10.1186/s13046-021-02108-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023]
Abstract
Background Tumor-associated macrophages (TAMs) are key regulators of the complex interplay between cancer and the immune microenvironment. Tumor cell-derived spondin 2 (SPON2) is an extracellular matrix glycoprotein that has complicated roles in recruitment of macrophages and neutrophils during inflammation. Overexpression of SPON2 has been shown to promote tumor cell migration in colorectal cancer (CRC). However, the mechanism by which SPON2 regulates the accumulation of TAMs in the tumor microenvironment (TME) of CRC is unknown. Methods Immunohistochemistry was used to examine SPON2 expression in clinical CRC tissues. In vitro migration assays, transendothelial migration assays (iTEM), and cell adhesion assays were used to investigate the effects of SPON2 on monocyte/macrophage migration. Subcutaneous tumor formation and orthotopic implantation assays were performed in C57 BL/6 mice to confirm the effects of SPON2 on TAM infiltration in tumors. Results SPON2 expression is positively correlated with M2-TAM infiltration in clinical CRC tumors and poor prognosis of CRC patients. In addition, SPON2 promotes cytoskeletal remodeling and transendothelial migration of monocytes by activating integrin β1/PYK2 axis. SPON2 may indirectly induce M2-polarization through upregulating cytokines including IL10, CCL2 and CSF1 expression in tumor cells. Blocking M2 polarization and Macrophage depletion inhibited the SPON2-induced tumors growth and invasion. Furthermore, blocking the SPON2/integrin β1/PYK2 axis impairs the transendothelial migration of monocytes and cancer-promoting functions of TAMs in vivo. Conclusions Our findings demonstrate that SPON2-driven M2-TAM infiltration plays an important role during CRC tumor growth and metastasis. SPON2 may be a valuable biomarker guiding the use of macrophage-targeting strategies and a potential therapeutic target in advanced CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02108-0.
Collapse
Affiliation(s)
- Chengmei Huang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Ruizhang Ou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Xiaoning Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yaxin Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yihao Liang
- Department of Orthopedist, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510000, China
| | - Xiaohui Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Lei Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Mingzhou Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Dagui Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Junfeng Qiu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Guanglong Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Lingjie Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yuanyuan Wu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Huiyi Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yanmin Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China.
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China. .,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, Guangdong, China. .,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
17
|
Li J, Wang Z, Tie C. High expression of ladinin-1 (LAD1) predicts adverse outcomes: a new candidate docetaxel resistance gene for prostatic cancer (PCa). Bioengineered 2021; 12:5749-5759. [PMID: 34516317 PMCID: PMC8806705 DOI: 10.1080/21655979.2021.1968647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Docetaxel resistance is one of the major obstacles that undermine the treatment outcome of PCa. Exploring molecular mechanisms associated with docetaxel resistance could provide insights into the formulation of novel strategies enhancing the efficacy of PCa treatment. Ladinin-1 (LAD1) is an anchoring filament protein in basement membranes, which contributes to the association of the epithelial cells with the underlying mesenchyme. LAD1 has been implicated in the progression of different cancers. However, its role in PCa remains to be investigated. In the present study, we found that LAD1 was highly expressed in docetaxel-resistant PCa cells, while its expression was significantly suppressed in tumor samples after docetaxel treatment. Moreover, the expression level of LAD1 in PCa tissues was significantly higher than that of normal tissue, and high expression level of LAD1 was significantly associated with adverse outcomes of PCa patients. Finally, high expression of LAD1 in PCa tissue was also correlated with the expression level of genes involving in tumor cell proliferation and invasive behaviors. Collectively, our data suggest that LAD1 may serve as a potential prognostic factor in PCa patients.
Collapse
Affiliation(s)
- Jianping Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Ziming Wang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Chong Tie
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
18
|
Qin L, Chen J, Lu D, Jain P, Yu Y, Cardenas D, Peng X, Yu X, Xu J, Wang J, O’ Malley BW, Lonard DM. Development of improved SRC-3 inhibitors as breast cancer therapeutic agents. Endocr Relat Cancer 2021; 28:657-670. [PMID: 34310341 PMCID: PMC8404148 DOI: 10.1530/erc-20-0402] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 07/26/2021] [Indexed: 11/08/2022]
Abstract
Steroid receptor coactivators (SRCs) possess specific and distinct oncogenic roles in the initiation of cancer and in its progression to a more aggressive disease. These coactivators interact with nuclear receptors and other transcription factors to boost transcription of multiple genes, which potentiate cancer cell proliferation, migration, invasion, tumor angiogenesis and epithelial-mesenchymal transition (EMT). Targeting SRCs using small molecule inhibitors (SMIs) is a promising approach to control cancer progression and metastasis. By high-throughput screening analysis, we recently identified SI-2 as a potent SRC SMI. To develop therapeutic agents, SI-10 and SI-12, the SI-2 analogs are synthesized that incorporate the addition of F atoms to the SI-2 chemical structure. As a result, these analogs exhibit a significantly prolonged plasma half-life, minimal toxicity and improved hERG activity. Biological functional analysis showed that SI-10 and SI-12 treatment (5-50 nM) can significantly inhibit viability, migration and invasion of breast cancer cells in vitro and repress the growth of breast cancer PDX organoids. Treatment of mice with 10 mg/kg/day of either SI-10 or SI-12 was sufficient to repress the growth of xenograft tumors derived from MDA-MB-231 and LM2 cells. Furthermore, in spontaneous and experimental metastasis mouse models developed from MDA-MB-231 and LM2 cells, respectively, SI-10 and SI-12 effectively inhibited the progression of breast cancer lung metastasis. These results demonstrate that SI-10 and SI-12 are promising therapeutic agents and are specifically effective in blocking tumor metastasis, a key point in tumor progression to a more lethal state that results in patient mortality in the majority of cases.
Collapse
Affiliation(s)
- Li Qin
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - Jianwei Chen
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030 USA
| | - Dong Lu
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030 USA
| | - Prashi Jain
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - Yang Yu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - David Cardenas
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - Xiaohui Peng
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - Xiaobin Yu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - Jin Wang
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030 USA
| | - Bert W. O’ Malley
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
| | - David M. Lonard
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030 USA
- Corresponding Author: David M. Lonard ()
| |
Collapse
|
19
|
Tu Y, Hu Y. MiRNA-34c-5p protects against cerebral ischemia/reperfusion injury: involvement of anti-apoptotic and anti-inflammatory activities. Metab Brain Dis 2021; 36:1341-1351. [PMID: 33842985 DOI: 10.1007/s11011-021-00724-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) are known as important regulators of gene expression and play important roles in diverse biological activities. However, the involvement of miRNAs in cerebral ischemia remains elusive. In the present study, using the middle cerebral artery occlusion (MCAO) model and oxygen-glucose deprivation/reperfusion (OGD/RP)-induced cell injury model, we found that the expression levels of miR-34c-5p were significantly reduced in MCAO rats and OGD/RP cells. Overexpression of miR-34c-5p could improve the increased brain infarction, brain water content and neurological scores in MCAO rats, as well as the abnormal expression of inflammatory cytokines (TNF-α, IL-6, COX-2, iNOS, IL-10) in OGD/RP cells. Moreover, overexpression of miR-34c-5p was found to inhibit the activity of nuclear factor-kappa B (NF-κB) by regulating the expression of nuclear receptor coactivator 1 (NCOA1), and increase the apoptotic rate of cortical neurons by inhibiting the expression of Caspase-3 and Bax and upregulating the expression of Bcl-2. Taken together, our findings demonstrated that miR-34c-5p plays an important role in cerebral ischemia/reperfusion injury, which may be mediated through inflammatory and apoptotic signaling pathways.
Collapse
Affiliation(s)
- Yaoran Tu
- Trauma Center, Third Affiliated Hospital of Nanchang University, No. 739 Qingshan South Road, Nanchang City, Jiangxi Province, 330000, People's Republic of China
| | - Yong Hu
- Trauma Center, Third Affiliated Hospital of Nanchang University, No. 739 Qingshan South Road, Nanchang City, Jiangxi Province, 330000, People's Republic of China.
| |
Collapse
|
20
|
Watters RJ, Verdelis K, Lucas PC, Jiang S, Chen Y, Lu F, Martin BM, Lukashova L, Pecar G, Morales-Restrepo A, Hankins M, Zhu L, Mittwede P, Hartmaier RJ, Alexander PG, Tseng GC, Weiss KR, Galson DL, Lee AV, Lee B, Oesterreich S. A Novel Mouse Model for SNP in Steroid Receptor Co-Activator-1 Reveals Role in Bone Density and Breast Cancer Metastasis. Endocrinology 2021; 162:6272285. [PMID: 33963375 PMCID: PMC8248588 DOI: 10.1210/endocr/bqab094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Indexed: 02/07/2023]
Abstract
The steroid receptor coactivator-1 (SRC-1) is a nuclear receptor co-activator, known to play key roles in both estrogen response in bone and in breast cancer metastases. We previously demonstrated that the P1272S single nucleotide polymorphism (SNP; P1272S; rs1804645) in SRC-1 decreases the activity of estrogen receptor in the presence of selective estrogen receptor modulators (SERMs) and that it is associated with a decrease in bone mineral density (BMD) after tamoxifen therapy, suggesting it may disrupt the agonist action of tamoxifen. Given such dual roles of SRC-1 in the bone microenvironment and in tumor cell-intrinsic phenotypes, we hypothesized that SRC-1 and a naturally occurring genetic variant, P1272S, may promote breast cancer bone metastases. We developed a syngeneic, knock-in mouse model to study if the SRC-1 SNP is critical for normal bone homeostasis and bone metastasis. Our data surprisingly reveal that the homozygous SRC-1 SNP knock-in increases tamoxifen-induced bone protection after ovariectomy. The presence of the SRC-1 SNP in mammary glands resulted in decreased expression levels of SRC-1 and reduced tumor burden after orthotopic injection of breast cancer cells not bearing the SRC-1 SNP, but increased metastases to the lungs in our syngeneic mouse model. Interestingly, the P1272S SNP identified in a small, exploratory cohort of bone metastases from breast cancer patients was significantly associated with earlier development of bone metastasis. This study demonstrates the importance of the P1272S SNP in both the effect of SERMs on BMD and the development of tumor in the bone.
Collapse
Affiliation(s)
- Rebecca J Watters
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Correspondence: Rebecca J. Watters, PhD, Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219, USA.
| | - Kostas Verdelis
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Peter C Lucas
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Shiming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Feiqi Lu
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Benjamin M Martin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Lyuda Lukashova
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Geoffrey Pecar
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Alejandro Morales-Restrepo
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Margaret Hankins
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Li Zhu
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Peter Mittwede
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Ryan J Hartmaier
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Peter G Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - George C Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Deborah L Galson
- Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232, USA
| | - Adrian V Lee
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Steffi Oesterreich
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
21
|
Choi SR, Wang HM, Shin MH, Lim HS. Hydrophobic Tagging-Mediated Degradation of Transcription Coactivator SRC-1. Int J Mol Sci 2021; 22:6407. [PMID: 34203850 PMCID: PMC8232704 DOI: 10.3390/ijms22126407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 01/19/2023] Open
Abstract
Steroid receptor coactivator-1 (SRC-1) is a transcription coactivator playing a pivotal role in mediating a wide range of signaling pathways by interacting with related transcription factors and nuclear receptors. Aberrantly elevated SRC-1 activity is associated with cancer metastasis and progression, and therefore, suppression of SRC-1 is emerging as a promising therapeutic strategy. In this study, we developed a novel SRC-1 degrader for targeted degradation of cellular SRC-1. This molecule consists of a selective ligand for SRC-1 and a bulky hydrophobic group. Since the hydrophobic moiety on the protein surface could mimic a partially denatured hydrophobic region of a protein, SRC-1 could be recognized as an unfolded protein and experience the chaperone-mediated degradation in the cells through the ubiquitin-proteasome system (UPS). Our results demonstrate that a hydrophobic-tagged chimeric molecule is shown to significantly reduce cellular levels of SRC-1 and suppress cancer cell migration and invasion. Together, these results highlight that our SRC-1 degrader represents a novel class of therapeutic candidates for targeting cancer metastasis. Moreover, we believe that the hydrophobic tagging strategy would be widely applicable to develop peptide-based protein degraders with enhanced cellular activity.
Collapse
Affiliation(s)
- So Ra Choi
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
| | - Hee Myeong Wang
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
| | - Min Hyeon Shin
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
- POSTECH Biotech Center, Pohang 37673, Korea
| | - Hyun-Suk Lim
- Department of Chemistry and Division of Advanced Material Science, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.R.C.); (H.M.W.)
| |
Collapse
|
22
|
Altaf R, Nadeem H, Babar MM, Ilyas U, Muhammad SA. Genome-scale meta-analysis of breast cancer datasets identifies promising targets for drug development. JOURNAL OF BIOLOGICAL RESEARCH (THESSALONIKE, GREECE) 2021; 28:5. [PMID: 33593445 PMCID: PMC7885587 DOI: 10.1186/s40709-021-00136-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/05/2021] [Indexed: 01/19/2023]
Abstract
Background Because of the highly heterogeneous nature of breast cancer, each subtype differs in response to several treatment regimens. This has limited the therapeutic options for metastatic breast cancer disease requiring exploration of diverse therapeutic models to target tumor specific biomarkers. Methods Differentially expressed breast cancer genes identified through extensive data mapping were studied for their interaction with other target proteins involved in breast cancer progression. The molecular mechanisms by which these signature genes are involved in breast cancer metastasis were also studied through pathway analysis. The potential drug targets for these genes were also identified. Results From 50 DEGs, 20 genes were identified based on fold change and p-value and the data curation of these genes helped in shortlisting 8 potential gene signatures that can be used as potential candidates for breast cancer. Their network and pathway analysis clarified the role of these genes in breast cancer and their interaction with other signaling pathways involved in the progression of disease metastasis. The miRNA targets identified through miRDB predictor provided potential miRNA targets for these genes that can be involved in breast cancer progression. Several FDA approved drug targets were identified for the signature genes easing the therapeutic options for breast cancer treatment. Conclusion The study provides a more clarified role of signature genes, their interaction with other genes as well as signaling pathways. The miRNA prediction and the potential drugs identified will aid in assessing the role of these targets in breast cancer.
Collapse
Affiliation(s)
- Reem Altaf
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan.
| | - Humaira Nadeem
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Mustafeez Mujtaba Babar
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-E-Millat University, Islamabad, 44000, Pakistan
| | - Umair Ilyas
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Riphah International University, Islamabad, 44000, Pakistan
| | - Syed Aun Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, 66000, Pakistan
| |
Collapse
|
23
|
Exploring the mechanism of aidi injection for lung cancer by network pharmacology approach and molecular docking validation. Biosci Rep 2021; 41:227696. [PMID: 33506873 PMCID: PMC7881165 DOI: 10.1042/bsr20204062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Background. Aidi injection (ADI) is an effective Traditional Chinese medicine preparation widely used for lung cancer. However, the pharmacological mechanisms of ADI on lung cancer remain to be elucidated. Methods. A network pharmacology (NP)-based approach and the molecular docking validation were conducted to explore underlying mechanisms of ADI on lung cancer. The compounds and target genes were screened by Traditional Chinese Medicine Systems Pharmacology (TCMSP) database and Bioinformatics Analysis Tool for Molecular mechANism of Traditional Chinese Medicine (Batman-TCM) database. The STRING database was utilized for protein interaction network construction. The R package clusterProfiler was used for bioinformatics annotation of hub target genes. The gene expression analysis and survival analysis were performed based on The Cancer Genome Atlas (TCGA) database. The Autodock Vina was used for molecular docking validation. Results. A total of five key compounds with 324 putative target genes were screened out, and 14 hub target genes were identified for treating lung cancer. Six hub genes could influence the survival of non-small cell lung cancer (NSCLC) patients. Of these hub genes, the expression pattern of EGFR, MYC, PIK3CA, and SMAD3 were significantly higher in the LUSC, while PIK3CA and RELA expressed lower in the LUAD group and LUSC group, respectively. These six hub genes had good docking affinity with the key compounds of ADI. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that ADI may exert therapeutic effects on lung cancer by regulating critical pathways including the thyroid hormone signaling pathway, MAPK signaling pathway, and PI3K-Akt signaling pathway. Conclusions. The present study explored the potential pharmacological mechanisms of ADI on lung cancer, promoting the clinical application of ADI in treating lung cancer, and providing references for advanced researches.
Collapse
|
24
|
Lee Y, Heo J, Jeong H, Hong KT, Kwon DH, Shin MH, Oh M, Sable GA, Ahn GO, Lee JS, Song HK, Lim HS. Targeted Degradation of Transcription Coactivator SRC-1 through the N-Degron Pathway. Angew Chem Int Ed Engl 2020; 59:17548-17555. [PMID: 33026161 DOI: 10.1002/anie.202005004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/01/2020] [Indexed: 01/12/2023]
Abstract
Aberrantly elevated steroid receptor coactivator-1 (SRC-1) expression and activity are strongly correlated with cancer progression and metastasis. Here we report, for the first time, the development of a proteolysis targeting chimera (PROTAC) that is composed of a selective SRC-1 binder linked to a specific ligand for UBR box, a unique class of E3 ligases recognizing N-degrons. We showed that the bifunctional molecule efficiently and selectively induced the degradation of SRC-1 in cells through the N-degron pathway. Importantly, given the ubiquitous expression of the UBR protein in most cells, PROTACs targeting the UBR box could degrade a protein of interest regardless of cell types. We also showed that the SRC-1 degrader significantly suppressed cancer cell invasion and migration in vitro and in vivo. Together, these results demonstrate that the SRC-1 degrader can be an invaluable chemical tool in the studies of SRC-1 functions. Moreover, our findings suggest PROTACs based on the N-degron pathway as a widely useful strategy to degrade disease-relevant proteins.
Collapse
Affiliation(s)
- Yeongju Lee
- Department of Chemistry and Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Jiwon Heo
- School of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Hoibin Jeong
- Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Kyung Tae Hong
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), 5. Hwarang-ro, 14-gil, Seongbuk-gu, Seoul, 02792, South Korea
| | - Do Hoon Kwon
- School of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Min Hyeon Shin
- Department of Chemistry and Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Misook Oh
- Department of Chemistry and Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - Ganesh A Sable
- Department of Chemistry and Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| | - G-One Ahn
- Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Jun-Seok Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), 5. Hwarang-ro, 14-gil, Seongbuk-gu, Seoul, 02792, South Korea
| | - Hyun Kyu Song
- School of Life Sciences and Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, South Korea
| | - Hyun-Suk Lim
- Department of Chemistry and Division of Advanced Materials Science, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, 37673, South Korea
| |
Collapse
|
25
|
Lee Y, Heo J, Jeong H, Hong KT, Kwon DH, Shin MH, Oh M, Sable GA, Ahn G, Lee J, Song HK, Lim H. Targeted Degradation of Transcription Coactivator SRC‐1 through the N‐Degron Pathway. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Yeongju Lee
- Department of Chemistry and Division of Advanced Materials Science Pohang University of Science and Technology (POSTECH) 77 Cheongam-Ro, Nam-Gu Pohang 37673 South Korea
| | - Jiwon Heo
- School of Life Sciences and Biotechnology Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 South Korea
| | - Hoibin Jeong
- Research Institute for Veterinary Science and College of Veterinary Medicine Seoul National University 1 Gwanak-ro Gwanak-gu Seoul 08826 South Korea
| | - Kyung Tae Hong
- Molecular Recognition Research Center Korea Institute of Science and Technology (KIST) 5. Hwarang-ro, 14-gil, Seongbuk-gu Seoul 02792 South Korea
| | - Do Hoon Kwon
- School of Life Sciences and Biotechnology Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 South Korea
| | - Min Hyeon Shin
- Department of Chemistry and Division of Advanced Materials Science Pohang University of Science and Technology (POSTECH) 77 Cheongam-Ro, Nam-Gu Pohang 37673 South Korea
| | - Misook Oh
- Department of Chemistry and Division of Advanced Materials Science Pohang University of Science and Technology (POSTECH) 77 Cheongam-Ro, Nam-Gu Pohang 37673 South Korea
| | - Ganesh A. Sable
- Department of Chemistry and Division of Advanced Materials Science Pohang University of Science and Technology (POSTECH) 77 Cheongam-Ro, Nam-Gu Pohang 37673 South Korea
| | - G‐One Ahn
- Research Institute for Veterinary Science and College of Veterinary Medicine Seoul National University 1 Gwanak-ro Gwanak-gu Seoul 08826 South Korea
| | - Jun‐Seok Lee
- Molecular Recognition Research Center Korea Institute of Science and Technology (KIST) 5. Hwarang-ro, 14-gil, Seongbuk-gu Seoul 02792 South Korea
| | - Hyun Kyu Song
- School of Life Sciences and Biotechnology Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 South Korea
| | - Hyun‐Suk Lim
- Department of Chemistry and Division of Advanced Materials Science Pohang University of Science and Technology (POSTECH) 77 Cheongam-Ro, Nam-Gu Pohang 37673 South Korea
| |
Collapse
|
26
|
Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J, Wei X. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther 2019; 4:62. [PMID: 31871779 PMCID: PMC6915746 DOI: 10.1038/s41392-019-0095-0] [Citation(s) in RCA: 662] [Impact Index Per Article: 110.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/16/2019] [Accepted: 10/24/2019] [Indexed: 02/05/2023] Open
Abstract
Epigenetic alternations concern heritable yet reversible changes in histone or DNA modifications that regulate gene activity beyond the underlying sequence. Epigenetic dysregulation is often linked to human disease, notably cancer. With the development of various drugs targeting epigenetic regulators, epigenetic-targeted therapy has been applied in the treatment of hematological malignancies and has exhibited viable therapeutic potential for solid tumors in preclinical and clinical trials. In this review, we summarize the aberrant functions of enzymes in DNA methylation, histone acetylation and histone methylation during tumor progression and highlight the development of inhibitors of or drugs targeted at epigenetic enzymes.
Collapse
Affiliation(s)
- Yuan Cheng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Mo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shengyong Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Junhong Han
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Hinshaw DC, Shevde LA. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res 2019; 79:4557-4566. [PMID: 31350295 DOI: 10.1158/0008-5472.can-18-3962] [Citation(s) in RCA: 2043] [Impact Index Per Article: 340.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/28/2019] [Accepted: 05/21/2019] [Indexed: 12/14/2022]
Abstract
Cancer development and progression occurs in concert with alterations in the surrounding stroma. Cancer cells can functionally sculpt their microenvironment through the secretion of various cytokines, chemokines, and other factors. This results in a reprogramming of the surrounding cells, enabling them to play a determinative role in tumor survival and progression. Immune cells are important constituents of the tumor stroma and critically take part in this process. Growing evidence suggests that the innate immune cells (macrophages, neutrophils, dendritic cells, innate lymphoid cells, myeloid-derived suppressor cells, and natural killer cells) as well as adaptive immune cells (T cells and B cells) contribute to tumor progression when present in the tumor microenvironment (TME). Cross-talk between cancer cells and the proximal immune cells ultimately results in an environment that fosters tumor growth and metastasis. Understanding the nature of this dialog will allow for improved therapeutics that simultaneously target multiple components of the TME, increasing the likelihood of favorable patient outcomes.
Collapse
Affiliation(s)
- Dominique C Hinshaw
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Lalita A Shevde
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama. .,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
28
|
Meng X, Zhao Y, Liu J, Wang L, Dong Z, Zhang T, Gu X, Zheng Z. Comprehensive analysis of histone modification-associated genes on differential gene expression and prognosis in gastric cancer. Exp Ther Med 2019; 18:2219-2230. [PMID: 31452712 DOI: 10.3892/etm.2019.7808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests that the epigenetic alterations caused by histone modifications have important roles in the genesis of gastric cancer (GC), particularly the well-studied acetylation and methylation modifications. In the present study, a Bioinformatics analysis of the expression of histone modification-associated genes in GC and normal tissues was performed by using datasets from Oncomine, the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). The clinical data of GC patients were downloaded from TCGA to determine the association between histone modification-associated gene expression and clinicopathological parameters or survival of GC. Finally, lysine acetyltransferase 2A (KAT2A), nuclear receptor coactivator 1 (NCOA1), SMYD family member 5 (SMYD5), protein arginine methyltransferase 1 (PRMT1) and PRDF1-RIZ (PR)/Su(var)3-9, enhancer-of-zeste and trithorax (SET) domain 16 (PRDM16) were screened; KAT2A, SMYD5 and PRMT1 were upregulated, while PRDM16 expression was downregulated in GC. Analysis of the GEO and Oncomine datasets revealed that NCOA1 was upregulated, which was contrary to the result obtained with the TCGA stomach adenocarcinoma dataset. Aberrant expression of KAT2A, NCOA1, SMYD5 and PRMT1 was more obvious in gastric intestinal-type adenocarcinoma; low NCOA1 expression was associated with better overall survival of GC patients [hazard ratio (HR)=0.690, 95% CI=0.570-0.840, P<0.001] and was an independent predictor for patients diagnosed with GC (HR=0.639, 95% CI=0.437-0.933, P=0.020). Correlation analysis and protein-protein interaction network analysis indicated a close association between ATAD2 and estrogen receptor 1 (ESR1), PRMT1, NCOA1 and KAT2A. In conclusion, differential expression of KAT2A, NCOA1, SMYD5, PRMT1 and PRDM16 was identified in GC vs. normal tissues, low NCOA1 expression was associated with poor survival of GC and ATAD2 may interact with ESR1 to regulate NCOA1 and PRMT1 in GC.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Jingwei Liu
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lu Wang
- Department of Ultrasonography, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Zhe Dong
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Tao Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Xiaohu Gu
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Zhichao Zheng
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
29
|
NEMP1 Promotes Tamoxifen Resistance in Breast Cancer Cells. Biochem Genet 2019; 57:813-826. [PMID: 31079234 DOI: 10.1007/s10528-019-09926-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
Abstract
Breast cancer (BC) is a worldwide malignant and a leading death cancer in women. Studies have shown that adjuvant tamoxifen reduces the recurrence rate and metastasis in BC. Even though tamoxifen has been used for the therapy of BC for decades, the resistance of it on BC cells could not be ignored. In this study, we first established a tamoxifen-resistant BC cell line and then demonstrated the overexpression of nuclear envelope integral membrane protein 1 (NEMP1) in the tamoxifen-resistant BC cells. Moreover, through a cell viability assay combined with depletion or overexpression technology, we addressed the important role of NEMP1 for the tamoxifen resistance in BC cells. Importantly, we further revealed that NEMP1 modulated tamoxifen resistance by regulating nuclear receptor coactivator 1 (NCOA1). In general, NEMP1 shows responsibility for the resistance of tamoxifen through regulating NCOA1 in BC cells. These results broaden the understanding of the tamoxifen resistance during the chemotherapy in BC and may provide new therapy method for BC.
Collapse
|
30
|
Francipane MG, Bulanin D, Lagasse E. Establishment and Characterization of 5-Fluorouracil-Resistant Human Colorectal Cancer Stem-Like Cells: Tumor Dynamics under Selection Pressure. Int J Mol Sci 2019; 20:ijms20081817. [PMID: 31013771 PMCID: PMC6515384 DOI: 10.3390/ijms20081817] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/21/2019] [Accepted: 04/09/2019] [Indexed: 12/20/2022] Open
Abstract
5-Fluorouracil (5-FU) remains the gold standard of first-line treatment for colorectal cancer (CRC). Although it may initially debulk the tumor mass, relapses frequently occur, indicating the existence of cancer cells that are therapy-resistant and are capable of refueling tumor growth. To identify mechanisms of drug resistance, CRC stem-like cells were subjected to long-term 5-FU selection using either intermittent treatment regimen with the IC50 drug dose or continuous treatment regimen with escalating drug doses. Parental cancer cells were cultivated in parallel. Real-time PCR arrays and bioinformatic tools were used to investigate gene expression changes. We found the first method selected for cancer cells with more aggressive features. We therefore transplanted these cancer cells or parental cells in mice, and again, found that not only did the 5-FU-selected cancer cells generate more aggressive tumors with respect to their parental counterpart, but they also showed a different gene expression pattern as compared to what we had observed in vitro, with ID1 the top upregulated gene. We propose ID1 as a stemness marker pervasively expressed in secondary lesions emerging after completion of chemotherapy.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine and Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
- Ri.MED Foundation, 90133 Palermo, Italy.
| | - Denis Bulanin
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan.
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine and Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
31
|
Angiotensin II induced CSF1 transcription is mediated by a crosstalk between different epigenetic factors in vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:1-11. [PMID: 30317027 DOI: 10.1016/j.bbagrm.2018.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 01/15/2023]
Abstract
Endothelium-derived colony stimulating factor (CSF1) plays a key role in a range of human pathologies. Angiotensin II (Ang II) has been documented to stimulate CSF1 transcription although the underlying epigenetic mechanism remains unclear. Here we report that induction of CSF1 transcription by Ang II in vascular endothelial cells paralleled alterations of signature histone modifications surrounding the CSF1 promoter. Specifically, ChIP assays indicated that there was a simultaneous up-regulation of both acetylated H3 and trimethylated H3K4, indicative of transcriptional activation, and down-regulation of dimethyl H3K9, implicated in transcriptional repression, surrounding the proximal CSF1 promoter. Further analysis revealed that silencing of brahma related gene 1 (BRG1), a chromatin remodeling protein, abrogated CSF1 induction by Ang II. In the meantime, BRG1 silencing erased H3 acetylation and H3K4 trimethylation and restored H3K9 dimethylation. Mechanistically, BRG1 interacted with and recruited SET1A, a histone H3K4 methyltransferase, and JMJD1A, a histone H3K9 demethylase, to the CSF1 promoter to alter chromatin structure thereby promoting CSF1 trans-activation in response to Ang II stimulation. Knockdown of either SET1A or JMJD1A blocked CSF1 induction by Ang II. Finally, we demonstrate that the crosstalk between BRG1 and histone modifying enzymes was mediated by the transcription factor AP-1. In conclusion, our data unveil a novel epigenetic mechanism whereby a BRG1-centered complex mediates transcriptional activation of CSF1 by Ang II in vascular endothelial cells.
Collapse
|
32
|
Wang L, Li W, Li K, Guo Y, Liu D, Yao Z, Lin X, Li S, Jiang Z, Liu Q, Jiang Y, Zhang B, Chen L, Zhou F, Ren H, Lin D, Zhang D, Yeung SJ, Zhang H. The oncogenic roles of nuclear receptor coactivator 1 in human esophageal carcinoma. Cancer Med 2018; 7:5205-5216. [PMID: 30270520 PMCID: PMC6198200 DOI: 10.1002/cam4.1786] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/05/2018] [Accepted: 08/16/2018] [Indexed: 02/05/2023] Open
Abstract
Nuclear receptor coactivator 1 (NCOA1) plays crucial roles in the regulation of gene expression mediated by a wide spectrum of steroid receptors such as androgen receptor (AR), estrogen receptor α (ER α), and estrogen receptor β (ER β). Therefore, dysregulations of NCOA1 have been found in a variety of cancer types. However, the clinical relevance and the functional roles of NCOA1 in human esophageal squamous cell carcinoma (ESCC) are less known. We found in this study that elevated levels of NCOA1 protein and/or mRNA as well as amplification of the NCOA1 gene occur in human ESCC. Elevated levels of NCOA1 due to these dysregulations were not only associated with more aggressive clinic-pathologic parameters but also poorer survival. Results from multiple cohorts of ESCC patients strongly suggest that the levels of NCOA1 could serve as an independent predictor of overall survival. In addition, silencing NCOA1 in ESCC cells remarkably decreased proliferation, migration, and invasion. These findings not only indicate that NCOA1 plays important roles in human ESCC but the levels of NCOA1 also could serve as a potential prognostic biomarker of ESCC and targeting NCOA1 could be an efficacious strategy in ESCC treatment.
Collapse
Affiliation(s)
- Lu Wang
- Department of Immunotherapy and Gastrointestinal OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Weiwei Li
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Kai Li
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Yi Guo
- Endoscopy CentreAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Ditian Liu
- Department of Thoracic SurgeryAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Zhimeng Yao
- Department of Immunotherapy and Gastrointestinal OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Xianjie Lin
- Department of Immunotherapy and Gastrointestinal OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Shujun Li
- Department of Thoracic SurgerySecond Affiliated Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Zuojie Jiang
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Qing Liu
- Department of PathologyThe First People's Hospital of FoshanFoshanGuangdongChina
| | - Yi Jiang
- Department of Immunotherapy and Gastrointestinal OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Beien Zhang
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
- Department of Science and EducationAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Lei Chen
- Department of Immunotherapy and Gastrointestinal OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Fuyou Zhou
- Department of PathologyAnyang Tumour HospitalAnyangHenanChina
| | - Hongzheng Ren
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
| | - Danxia Lin
- Department of Breast OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| | - Dianzheng Zhang
- Department of Bio‐Medical SciencesPhiladelphia College of Osteopathic MedicinePhiladelphiaPennsylvania
| | - Sai‐Ching Jim Yeung
- Department of Emergency Medicine, Department of Endocrine Neoplasia and Hormonal DisordersThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Hao Zhang
- Department of Immunotherapy and Gastrointestinal OncologyAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
- Cancer Research CentreShantou University Medical CollegeShantouGuangdongChina
- Institute of Precision Cancer Medicine and Pathology and Department of PathologyJinan University Medical CollegeGuangzhouChina
- Tumor Tissue BankAffiliated Cancer Hospital of Shantou University Medical CollegeShantouGuangdongChina
| |
Collapse
|
33
|
Cheng M, Michalski S, Kommagani R. Role for Growth Regulation by Estrogen in Breast Cancer 1 (GREB1) in Hormone-Dependent Cancers. Int J Mol Sci 2018; 19:ijms19092543. [PMID: 30154312 PMCID: PMC6163654 DOI: 10.3390/ijms19092543] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
Sex hormones play important roles in the onset and progression of several cancers, such as breast, ovarian, and prostate cancer. Although drugs targeting sex hormone function are useful in treating cancer, tumors often develop resistance. Thus, we need to define the downstream effectors of sex hormones in order to develop new treatment strategies for these cancers. Recent studies unearthed one potential mediator of steroid hormone action in tumors: growth regulation by estrogen in breast cancer 1 (GREB1). GREB1 is an early estrogen-responsive gene, and its expression is correlated with estrogen levels in breast cancer patients. Additionally, GREB1 responds to androgen in prostate cancer cells, and can stimulate the proliferation of breast, ovarian, and prostate cancer cells. Recent studies have shown that GREB1 also responds to progesterone in human endometrial cells, suggesting that GREB1 is a pan steroid-responsive gene. This mini-review examines evidence that GREB1 participates in several hormone-dependent cancers and could be targeted to treat these cancers.
Collapse
Affiliation(s)
- Meng Cheng
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Stephanie Michalski
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Ramakrishna Kommagani
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
34
|
Jia Z, Wan F, Zhu Y, Shi G, Zhang H, Dai B, Ye D. Forkhead-box series expression network is associated with outcome of clear-cell renal cell carcinoma. Oncol Lett 2018; 15:8669-8680. [PMID: 29805604 PMCID: PMC5950509 DOI: 10.3892/ol.2018.8405] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/17/2017] [Indexed: 12/21/2022] Open
Abstract
Previous studies have demonstrated that several members of the Forkhead-box (FOX) family of genes are associated with tumor progression and metastasis. The objective of the current study was to screen candidate FOX family genes identified from analysis of molecular networks in clear cell renal cell carcinoma (ccRCC). The expression of FOX family genes as well as FOX family-associated genes was examined, and Kaplan-Meier survival analysis was performed in The Cancer Genome Atlas (TCGA) cohort (n=525). Patient characteristics, including sex, age, tumor diameter, laterality, tumor-node-metastasis, tumor grade, stage, white blood cell count, platelet count, the levels of hemoglobin, overall survival (OS) and disease-free survival (DFS), were collected for univariate and multivariate Cox proportional hazards ratio analyses. A total of seven candidate FOX family genes were selected from the TCGA database subsequent to univariate and multivariate Cox proportional hazards ratio analyses. FOXA1, FOXA2, FOXD1, FOXD4L2, FOXK2 and FOXL1 were associated with poor OS time, while FOXA1, FOXA2, FOXD1 and FOXK2 were associated with poor DFS time (P<0.05). FOXN2 was associated with favorable outcomes for overall and disease-free survival (P<0.05). In the gene cluster network analysis, the expression of FOX family-associated genes, including nuclear receptor coactivator (NCOA)1, NADH-ubiquinone oxidoreductase flavoprotein 3 (NDUFV3), phosphatidylserine decarboxylase (PISD) and pyruvate kinase liver and red blood cell (PKLR), were independent prognostic factors for OS in patients with ccRCC. Results of the present study revealed that the expression of FOX family genes, including FOXA1, FOXA2, FOXD1, FOXD4L2, FOXK2 and FOXL1, and FOX family-associated genes, including NCOA1, NDUFV3, PISD and PKLR, are independent prognostic factors for patients with ccRCC.
Collapse
Affiliation(s)
- Zhongwei Jia
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| | - Fangning Wan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai 200032, P.R. China
| |
Collapse
|
35
|
Browne AL, Charmsaz S, Varešlija D, Fagan A, Cosgrove N, Cocchiglia S, Purcell S, Ward E, Bane F, Hudson L, Hill AD, Carroll JS, Redmond AM, Young LS. Network analysis of SRC-1 reveals a novel transcription factor hub which regulates endocrine resistant breast cancer. Oncogene 2018; 37:2008-2021. [PMID: 29367763 PMCID: PMC5895607 DOI: 10.1038/s41388-017-0042-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 01/15/2023]
Abstract
Steroid receptor coactivator 1 (SRC-1) interacts with nuclear receptors and other transcription factors (TFs) to initiate transcriptional networks and regulate downstream genes which enable the cancer cell to evade therapy and metastasise. Here we took a top-down discovery approach to map out the SRC-1 transcriptional network in endocrine resistant breast cancer. First, rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) was employed to uncover new SRC-1 TF partners. Next, RNA sequencing (RNAseq) was undertaken to investigate SRC-1 TF target genes. Molecular and patient-derived xenograft studies confirmed STAT1 as a new SRC-1 TF partner, important in the regulation of a cadre of four SRC-1 transcription targets, NFIA, SMAD2, E2F7 and ASCL1. Extended network analysis identified a downstream 79 gene network, the clinical relevance of which was investigated in RNAseq studies from matched primary and local-recurrence tumours from endocrine resistant patients. We propose that SRC-1 can partner with STAT1 independently of the estrogen receptor to initiate a transcriptional cascade and control regulation of key endocrine resistant genes.
Collapse
Affiliation(s)
- Alacoque L Browne
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Sara Charmsaz
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Ailis Fagan
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Nicola Cosgrove
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Sinéad Cocchiglia
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Siobhan Purcell
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Elspeth Ward
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Fiona Bane
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Lance Hudson
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Arnold D Hill
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland
| | - Jason S Carroll
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Aisling M Redmond
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons, Dublin, Ireland.
| |
Collapse
|
36
|
Qin L, Xu Y, Yu X, Toneff MJ, Li D, Liao L, Martinez JD, Li Y, Xu J. The histone demethylase Kdm3a is required for normal epithelial proliferation, ductal elongation and tumor growth in the mouse mammary gland. Oncotarget 2017; 8:84761-84775. [PMID: 29156681 PMCID: PMC5689571 DOI: 10.18632/oncotarget.21380] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/31/2017] [Indexed: 01/08/2023] Open
Abstract
Histone modification alters chromatin architecture to regulate gene transcription. KDM3A is a histone demethylase in the JmjC domain-containing protein family. It removes di- and mono- methyl residues from di- or mono-methylated lysine 9 of histone H3 (H3K9me2/me1). Recent studies have shown that Kdm3a plays an important role in self-renewal of embryonic stem cells, spermatogenesis, metabolism, sex determination and tumor angiogenesis. However, its role in mammary gland development and breast carcinogenesis remains unclear. In this study, we found that Kdm3a is expressed in the mouse mammary gland epithelial cells. Knockout of Kdm3a significantly increased H3K9me2/me1 levels in these epithelial cells, which correlated with markedly decreased mammary gland ductal elongation and branching in the intact knockout virgin mice. Furthermore, estrogen replacement in the ovariectomized Kdm3a knockout mice couldn’t rescue the retarded ductal growth. Moreover, transplantation of KO mammary gland pieces to wild type recipient mice showed slower ductal growth compared with that of WT gland pieces. Consistently, knockout of Kdm3a also reduced the proliferation rates and cyclin D1 expression in the mammary gland epithelial cells. In addition, Kdm3a knockout did not significantly change the latency of the polyoma middle T oncogene-induced mammary gland tumorigenesis. Tumor growth, however, was slowed which might be due to the decrease in cyclin D1 expression and tumor cell proliferation. We also found that Kdm3a binds and activates the cyclin D1 promoter. These results demonstrate that Kdm3a plays an important intrinsic role in promoting mammary gland ductal growth and tumor growth probably through enhancing cyclin D1 expression and cell proliferation.
Collapse
Affiliation(s)
- Li Qin
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Yixiang Xu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xiaobin Yu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Michael J Toneff
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Dabing Li
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Institute for Cancer Medicine and College of Basic Biomedical Sciences, Southwest Medical University, Sichuan, China
| | - Lan Liao
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jarrod D Martinez
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Yi Li
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Institute for Cancer Medicine and College of Basic Biomedical Sciences, Southwest Medical University, Sichuan, China
| |
Collapse
|
37
|
Rohira AD, Lonard DM. Steroid receptor coactivators present a unique opportunity for drug development in hormone-dependent cancers. Biochem Pharmacol 2017; 140:1-7. [DOI: 10.1016/j.bcp.2017.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/04/2017] [Indexed: 01/17/2023]
|
38
|
Wu H, Bi J, Peng Y, Huo L, Yu X, Yang Z, Zhou Y, Qin L, Xu Y, Liao L, Xie Y, Conneely OM, Jonkers J, Xu J. Nuclear receptor NR4A1 is a tumor suppressor down-regulated in triple-negative breast cancer. Oncotarget 2017; 8:54364-54377. [PMID: 28903348 PMCID: PMC5589587 DOI: 10.18632/oncotarget.17532] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/15/2017] [Indexed: 12/11/2022] Open
Abstract
The nuclear receptor (NR) superfamily contains hormone-inducible transcription factors that regulate many physiological and pathological processes through regulating gene expression. NR4A1 is an NR family member that still does not have an identified endogenous ligand, and its role in cancer is also currently unclear and controversial. In this study, we aimed to define the expression profiles and specific role of NR4A1 in the highly malignant triple-negative breast cancer (TNBC), which still lacks available targeted therapies. Bioinformatic analysis revealed a decrease of NR4A1 mRNA expression in human TNBC samples. Semi-quantitative analysis of NR4A1 protein expression by immunohistochemistry also identified a progressive NR4A1 reduction during the development of mouse basal-like mammary tumors and a significant NR4A1 downregulation in human TNBC samples. Furthermore, the expression levels of NR4A1 in human TNBC were negatively associated with tumor stage, lymph node metastasis and disease recurrence. Moreover, ectopic expression of NR4A1 in MDA-MB-231, a TNBC cell line with little endogenous NR4A1, inhibited the proliferation, viability, migration and invasion of these cells, and these inhibitions were associated with an attenuated JNK1–AP-1–cyclin D1 pathway. NR4A1 expression also largely suppressed the growth and metastasis of these cell-derived tumors in mice. These results demonstrate that NR4A1 is downregulated in TNBC and restoration of NR4A1 expression inhibits TNBC growth and metastasis, suggesting that NR4A1 is a tumor suppressor in TNBC.
Collapse
Affiliation(s)
- Hongmei Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Current address: College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jiong Bi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Current address: Departments of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yan Peng
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lei Huo
- Department of Pathology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaobin Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhihui Yang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Institute for Cancer Medicine, School of Basic Medical Sciences, and Department of Pathology, Xinan Medical University, Luzhou, Sichuan 646000, China
| | - Yunyun Zhou
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA.,Current address: Department of Data Science, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Li Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yixiang Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang Xie
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Orla M Conneely
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jos Jonkers
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, 1066 CX Amsterdam, Netherlands
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.,Institute for Cancer Medicine, School of Basic Medical Sciences, and Department of Pathology, Xinan Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
39
|
Frycz BA, Murawa D, Borejsza-Wysocki M, Wichtowski M, Spychała A, Marciniak R, Murawa P, Drews M, Jagodziński PP. mRNA expression of steroidogenic enzymes, steroid hormone receptors and their coregulators in gastric cancer. Oncol Lett 2017; 13:3369-3378. [PMID: 28521442 PMCID: PMC5431337 DOI: 10.3892/ol.2017.5881] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023] Open
Abstract
Epidemiological and experimental findings suggest that the development of gastric cancer (GC) is regulated by steroid hormones. In postmenopausal women and older men, the majority of steroid hormones are produced locally in peripheral tissue through the enzymatic conversion of steroid precursors. Therefore, using reverse transcription-quantitative polymerase chain reaction analysis, the mRNA expression of genes encoding steroidogenic enzymes, including steroid sulfatase (STS), hydroxy-delta-5-steroid dehydrogenase 3 beta- and steroid delta-isomerase 1 (HSD3B1), 17β-hydroxysteroid dehydrogenase type 7 and aromatase (CYP19A1), was investigated in primary tumoral and adjacent healthy gastric mucosa from 60 patients with GC. Furthermore, the mRNA levels for estrogen receptor α, estrogen receptor β (ESR2) and androgen receptor (AR), along with their coregulators, including proline, glutamate and leucine rich protein 1, CREB binding protein, nuclear receptor coactivator 1 (NCOA1), nuclear receptor corepressor 1 (NCOR1) and nuclear receptor subfamily 2 group F member 1 (NR2F1), were investigated. Additionally, the association between the mRNA expression of these genes and the clinicopathological features of patients with GC was examined. Significantly decreased levels of STS, HSD3B1, ESR2, AR, NCOA1 and NCOR1 mRNA, in addition to significantly increased levels of CYP19A1 mRNA were demonstrated in tumoral tissue samples compared with adjacent healthy gastric tissue samples. Deregulated expression of these genes in the analyzed tissue samples was associated with certain clinicopathological features of GC, such as age and localization of the tumor. The results of the current study suggest that all of the genes analyzed are expressed in tumoral and adjacent healthy gastric mucosa. In addition, the results indicate that abnormal expression of STS, ESR2, AR, NCOA1 and NCOR1 may serve a role in the development and progression of GC, and may be associated with specific clinicopathological features in patients with GC.
Collapse
Affiliation(s)
- Bartosz Adam Frycz
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 60-781 Poznań, Poland
| | - Dawid Murawa
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Research and Development Centre, Regional Specialist Hospital of Wrocław, 51-124 Wrocław, Poland
| | - Maciej Borejsza-Wysocki
- Department of General and Endocrine Surgery and Gastroenterological Oncology, Heliodor Święcicki Clinical Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
| | - Mateusz Wichtowski
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Arkadiusz Spychała
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Ryszard Marciniak
- Department of General and Endocrine Surgery and Gastroenterological Oncology, Heliodor Święcicki Clinical Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
| | - Paweł Murawa
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Michał Drews
- Department of General and Endocrine Surgery and Gastroenterological Oncology, Heliodor Święcicki Clinical Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 60-781 Poznań, Poland
| |
Collapse
|
40
|
Guo J, Cao R, Yu X, Xiao Z, Chen Z. MicroRNA-223-3p inhibits human bladder cancer cell migration and invasion. Tumour Biol 2017; 39:1010428317691678. [PMID: 28222670 DOI: 10.1177/1010428317691678] [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] [Indexed: 12/30/2022] Open
Abstract
The regulation of initiation and progression during carcinogenesis of bladder carcinoma is not completely elucidated. Dysregulation of microRNAs has been detected to play critical roles in the development of various cancers, including bladder carcinoma, whereas the involvement of miR-223-3p in the tumorigenesis of bladder carcinoma has not been studied. Here, we show that significantly higher levels of nuclear receptor coactivator 1 and significantly lower levels of miR-223-3p were detected in bladder carcinoma tissue, compared to the adjacent non-tumor tissue. In addition, the levels of nuclear receptor coactivator 1 and miR-223-3p were inversely correlated. Moreover, low miR-223-3p levels in bladder carcinoma specimens were associated with poor prognosis. In vitro, depletion of miR-223-3p increased bladder carcinoma cell invasion, which was abolished by overexpression of nuclear receptor coactivator 1. Bioinformatics studies demonstrate that miR-223-3p may bind to the 3'-UTR of nuclear receptor coactivator 1 messenger RNA to inhibit its protein translation in bladder carcinoma cells. Together, our study highlights miR-223-3p as a previously unrecognized microRNA that inhibits bladder carcinoma invasiveness via nuclear receptor coactivator 1, and this finding may be important for developing innovative therapeutic targets in treating bladder carcinoma.
Collapse
Affiliation(s)
- Ju Guo
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Runfu Cao
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xingwei Yu
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zewen Xiao
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiwen Chen
- Institute of Urology, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
41
|
Ma YS, Wu TM, Lv ZW, Lu GX, Cong XL, Xie RT, Yang HQ, Chang ZY, Sun R, Chai L, Cai MX, Zhong XJ, Zhu J, Fu D. High expression of miR-105-1 positively correlates with clinical prognosis of hepatocellular carcinoma by targeting oncogene NCOA1. Oncotarget 2017; 8:11896-11905. [PMID: 28060733 PMCID: PMC5355313 DOI: 10.18632/oncotarget.14435] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence supports that microRNA (miRNA) plays a significant functional role in cancer progression by directly regulating respective targets. In this study, the expression levels of miR-105-1 and its target gene were analyzed using genes microarray and hierarchical clustering analysis followed by validation with quantitative RT-PCR in hepatocellular carcinoma (HCC) and normal liver tissues. We examined the expression of nuclear receptor coactivator 1 (NCOA1), the potential target gene of miR-105-1, following the transfection of miR-105-1 mimics or inhibitors. Our results showed that miR-105-1 was downregulated in HCC tissues when compared with normal liver tissues and patients with lower miR-105-1 expression had shorter overall survival (OS) and progression free survival (PFS). Moreover, NCOA1 was confirmed to be a direct target of miR-105-1. Furthermore, concomitant high expression of NCOA1 and low expression of miR-105-1 correlated with a shorter median OS and PFS in HCC patients. In conclusion, our results provide the first evidence that NCOA1 is a direct target of miR-105-1 suggesting that NCOA1 and miR-105-1 may have potential prognostic value and may be useful as tumor biomarkers for the diagnosis of HCC patients.
Collapse
Affiliation(s)
- Yu-Shui Ma
- 1 Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
- 2 Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, College of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Ting-Miao Wu
- 1 Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
- 3 Department of Radiology, the Fourth Affiliated Hospital, Medical University of Anhui, Hefei 230601, China
| | - Zhong-Wei Lv
- 1 Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Gai-Xia Lu
- 1 Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xian-Ling Cong
- 4 Tissue Bank, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Ru-Ting Xie
- 5 Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Hui-Qiong Yang
- 5 Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Zheng-Yan Chang
- 5 Department of Pathology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ran Sun
- 4 Tissue Bank, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Li Chai
- 1 Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ming-Xiang Cai
- 1 Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiao-Jun Zhong
- 6 Department of Medical Oncology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jian Zhu
- 7 Department of Digestive Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Da Fu
- 8 Central Laboratory for Medical Research, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
42
|
Meerson A, Yehuda H. Leptin and insulin up-regulate miR-4443 to suppress NCOA1 and TRAF4, and decrease the invasiveness of human colon cancer cells. BMC Cancer 2016; 16:882. [PMID: 27842582 PMCID: PMC5109693 DOI: 10.1186/s12885-016-2938-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/27/2016] [Indexed: 01/17/2023] Open
Abstract
Background Obesity is a risk factor for colorectal cancer (CRC). Normal and tumor cells respond to metabolic hormones, such as leptin and insulin. Thus, obesity-associated resistance to these hormones likely leads to changes in gene expression and behavior of tumor cells. However, the mechanisms affected by leptin and insulin signaling in CRC cells remain mostly unknown. Methods We hypothesized that microRNAs (miRNAs) are involved in the regulation of tumorigenesis-related gene expression in CRC cells by leptin and insulin. To test this hypothesis, miRNA levels in the CRC-derived cell lines HCT-116, HT-29 and DLD-1 were profiled, following leptin and insulin treatment. Candidate miRNAs were validated by RT-qPCR. Predicted miRNA targets with known roles in cancer, were validated by immunoblots and reporter assays in HCT-116 cells. Transfection of HCT-116 cells with candidate miRNA mimic was used to test in vitro effects on proliferation and invasion. Results Of ~800 miRNAs profiled, miR-4443 was consistently up-regulated by leptin and insulin in HCT-116 and HT-29, but not in DLD-1, which lacked normal leptin receptor expression. Dose response experiments showed that leptin at 100 ng/ml consistently up-regulated miR-4443 in HCT-116 cells, concomitantly with a significant decrease in cell invasion ability. Transfection with miR-4443 mimic decreased invasion and proliferation of HCT-116 cells. Moreover, leptin and miR-4443 transfection significantly down-regulated endogenous NCOA1 and TRAF4, both predicted targets of miR-4443 with known roles in cancer metastasis. miR-4443 was found to directly regulate TRAF4 and NCOA1, as validated by a reporter assay. The up-regulation of miR-4443 by leptin or insulin was attenuated by the inhibition of MEK1/2. Conclusions Our findings suggest that miR-4443 acts in a tumor-suppressive manner by down-regulating TRAF4 and NCOA1 downstream of MEK-C/EBP-mediated leptin and insulin signaling, and that insulin and/or leptin resistance (e.g. in obesity) may suppress this pathway and increase the risk of metastatic CRC. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2938-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ari Meerson
- Molecular Biology of Chronic Diseases, MIGAL Galilee Research Institute, PO Box 831, Kiryat Shmona, 11016, Israel. .,Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark.
| | - Hila Yehuda
- Molecular Biology of Chronic Diseases, MIGAL Galilee Research Institute, PO Box 831, Kiryat Shmona, 11016, Israel
| |
Collapse
|
43
|
Xu Y, Qin L, Sun T, Wu H, He T, Yang Z, Mo Q, Liao L, Xu J. Twist1 promotes breast cancer invasion and metastasis by silencing Foxa1 expression. Oncogene 2016; 36:1157-1166. [PMID: 27524420 PMCID: PMC5311074 DOI: 10.1038/onc.2016.286] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/12/2016] [Accepted: 07/05/2016] [Indexed: 12/31/2022]
Abstract
The heterogeneous breast cancers can be classified into different subtypes according to their histopathological characteristics and molecular signatures. Foxa1 expression is linked with luminal breast cancer (LBC) with good prognosis, whereas Twist1 expression is associated with basal-like breast cancer (BLBC) with poor prognosis owing to its role in promoting epithelial-to-mesenchymal transition (EMT), invasiveness and metastasis. However, the regulatory and functional relationships between Twist1 and Foxa1 in breast cancer progression are unknown. In this study, we demonstrate that in the estrogen receptor (ERα)-positive LBC cells Twist1 silences Foxa1 expression, which has an essential role in relieving Foxa1-arrested migration, invasion and metastasis of breast cancer cells. Mechanistically, Twist1 binds to Foxa1 proximal promoter and recruits the NuRD transcriptional repressor complex to de-acetylate H3K9 and repress RNA polymerase II recruitment. Twist1 also silences Foxa1 promoter by inhibiting AP-1 recruitment. Twist1 expression in MCF7 cells silenced Foxa1 expression, which was concurrent with the induction of EMT, migration, invasion and metastasis of these cells. Importantly, restored Foxa1 expression in these cells largely inhibited Twist1-promoted migration, invasion and metastasis. Restored Foxa1 expression did not change the Twist1-induced mesenchymal cellular morphology and the expression of Twist1-regulated E-cadherin, β-catenin, vimentin and Slug, but it partially rescued Twist1-silenced ERα and cytokeratin 8 expression and reduced Twist1-induced integrin α5, integrin β1 and MMP9 expression. In a xenografted mouse model, restored Foxa1 also increased Twist1-repressed LBC markers and decreased Twist1-induced BLBC markers. Furthermore, Twist1 expression is negatively correlated with Foxa1 in the human breast tumors. The tumors with high Twist1 and low Foxa1 expressions are associated with poor distant metastasis-free survival. These results demonstrate that Twist1's silencing effect on Foxa1 expression is largely responsible for Twist1-induced migration, invasion and metastasis, but less responsible for Twist1-induced mesenchymal morphogenesis and expression of certain EMT markers.
Collapse
Affiliation(s)
- Y Xu
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - L Qin
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - T Sun
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - H Wu
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - T He
- Institution for Cancer Medicine and College of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Z Yang
- Institution for Cancer Medicine and College of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Q Mo
- Department of Medicine-Hematology/Oncology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - L Liao
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - J Xu
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA.,Institution for Cancer Medicine and College of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
44
|
Qin L, Xu Y, Xu Y, Ma G, Liao L, Wu Y, Li Y, Wang X, Wang X, Jiang J, Wang J, Xu J. NCOA1 promotes angiogenesis in breast tumors by simultaneously enhancing both HIF1α- and AP-1-mediated VEGFa transcription. Oncotarget 2016; 6:23890-904. [PMID: 26287601 PMCID: PMC4695159 DOI: 10.18632/oncotarget.4341] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/09/2015] [Indexed: 01/03/2023] Open
Abstract
Nuclear receptor coactivator 1 (NCOA1) is overexpressed in a subset of breast cancer and its increased expression positively correlates with disease recurrence and metastasis. Although NCOA1 is known to promote breast cancer metastasis through working with multiple transcription factors to upregulate the expression of Twist1, ITGA5, CSF-1, SDF1 and CXCR4, the role of NCOA1 in breast tumor angiogenesis has not been investigated. In this study, we found that the microvascular density (MVD) was significantly decreased and increased in Ncoa1-knockout and NCOA1-overexpressing mammary tumors, respectively, in several breast cancer mouse models. Knockout or knockdown of NCOA1 in breast cancer cell lines also markedly compromised their capability to induce angiogenesis in Matrigel plugs embedded subcutaneously in mice, while this compromised capability could be rescued by VEGFa treatment. At the molecular level, NCOA1 upregulates VEGFa expression in both mouse mammary tumors and cultured breast cancer cells, and it does so by associating with both c-Fos, which is recruited to the AP-1 site at bp −938 of the VEGFa promoter, and HIF1α, which is recruited to the HIF1α-binding element at bp −979 of the VEGFa promoter, to enhance VEGFa transcription. In 140 human breast tumors, high NCOA1 protein correlates with high MVD and patients with both high NCOA1 and high MVD showed significantly shorter survival time. In summary, this study revealed a novel mechanism that NCOA1 potentiates breast cancer angiogenesis through upregulating HIF1α and AP-1-mediated VEGFa expression, which reinforces the rational of targeting NCOA1 in controlling breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Li Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yan Xu
- Department of Breast and Thyroid Surgery, Daping Hospital, Third Military Medical University, Chongqing, China.,Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yixiang Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Gang Ma
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Lan Liao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yelin Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Xian Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Xiaosong Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Jun Jiang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jin Wang
- Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Institute for Cancer Medicine and College of Basic Medical Sciences, Sichuan Medical University, Luzhou, Sichuan, China
| |
Collapse
|
45
|
Polyphenol-rich strawberry extract (PRSE) shows in vitro and in vivo biological activity against invasive breast cancer cells. Sci Rep 2016; 6:30917. [PMID: 27498973 PMCID: PMC4976366 DOI: 10.1038/srep30917] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 07/11/2016] [Indexed: 12/20/2022] Open
Abstract
We describe the biological effects of a polyphenol-rich strawberry extract (PRSE), obtained from the “Alba” variety, on the highly aggressive and invasive basal-like breast cancer cell line A17. Dose-response and time-course experiments showed that PRSE is able to decrease the cellular viability of A17 cells in a time- and dose-dependent manner. PRSE effect on cell survival was investigated in other tumor and normal cell lines of both mouse and human origin, demonstrating that PRSE is more active against breast cancer cells. Cytofluorimetric analysis of A17 cells demonstrated that sub-lethal doses of PRSE reduce the number of cells in S phase, inducing the accumulation of cells in G1 phase of cell cycle. In addition, the migration of A17 cells was studied monitoring the ability of PRSE to inhibit cellular mobility. Gene expression analysis revealed the modulation of 12 genes playing different roles in the cellular migration, adhesion and invasion processes. Finally, in vivo experiments showed the growth inhibition of A17 cells orthotopically transplanted into FVB syngeneic mice fed with PRSE. Overall, we demonstrated that PRSE exerts important biological activities against a highly invasive breast cancer cell line both in vitro and in vivo suggesting the strawberry extracts as preventive/curative food strategy.
Collapse
|
46
|
Dikshit A, Gao C, Small C, Hales K, Hales DB. Flaxseed and its components differentially affect estrogen targets in pre-neoplastic hen ovaries. J Steroid Biochem Mol Biol 2016; 159:73-85. [PMID: 26925929 PMCID: PMC4821676 DOI: 10.1016/j.jsbmb.2016.02.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/12/2016] [Accepted: 02/24/2016] [Indexed: 02/07/2023]
Abstract
Flaxseed has been studied for decades for its health benefits that include anti-cancer, cardio-protective, anti-diabetic, anti-inflammatory properties. The biologically active components that mediate these effects are the omega-3 fatty acids and the lignan, secoisolariciresinol diglucoside. We have previously shown that whole flaxseed supplemented diet decreases the severity and incidence of ovarian cancer while a 15% dose of flaxseed is most protective against inflammation and estrogen-induced chemical and genotoxicity. The objective of this study was to dissect the independent effects of the two flaxseed components on estrogen signaling and metabolism. Two and half year old hens were fed either a control diet, 15% whole flaxseed diet, defatted flax meal diet or 5% flax oil diet for 3 months after which the animals were sacrificed and blood and tissues were harvested. Whole flaxseed diet caused a decrease in expression of ERα. ERα target gene expression was assessed using RT(2) profiler PCR array. Some targets involved in the IGF/insulin signaling pathway (IRS1, IGFBP4, IGFBP5) were downregulated by flaxseed and its components. Flaxseed diet also downregulated AKT expression. A number of targets related to NF-kB signaling were altered by flaxseed diet including a series of targets implicated in cancer. Whole flaxseed diet also affected E2 metabolism by increasing CYP1A1 expression with a corresponding increase in the onco-protective E2 metabolite, 2-methoxyestradiol. The weak anti-estrogens, enterolactone, enterodiol and 2-methoxyestradiol, might be working synergistically to generate a protective effect on the ovaries from hens on whole flaxseed diet by altering the estrogen signaling and metabolism.
Collapse
Affiliation(s)
- Anushka Dikshit
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Chunqi Gao
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Carrie Small
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA
| | - Karen Hales
- Department of Obstetrics and Gynecology, Southern Illinois University School of Medicine, Carbondale, Illinois, USA
| | - Dale Buchanan Hales
- Department of Physiology, Southern Illinois University School of Medicine, 1125 Lincoln Drive, Life Science II, Room 245B, Carbondale, Illinois 62901, USA.
| |
Collapse
|
47
|
Liu X, Li H, Rajurkar M, Li Q, Cotton JL, Ou J, Zhu LJ, Goel HL, Mercurio AM, Park JS, Davis RJ, Mao J. Tead and AP1 Coordinate Transcription and Motility. Cell Rep 2016; 14:1169-1180. [PMID: 26832411 DOI: 10.1016/j.celrep.2015.12.104] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 11/11/2015] [Accepted: 12/23/2015] [Indexed: 11/25/2022] Open
Abstract
The Tead family transcription factors are the major intracellular mediators of the Hippo-Yap pathway. Despite the importance of Hippo signaling in tumorigenesis, Tead-dependent downstream oncogenic programs and target genes in cancer cells remain poorly understood. Here, we characterize Tead4-mediated transcriptional networks in a diverse range of cancer cells, including neuroblastoma, colorectal, lung, and endometrial carcinomas. By intersecting genome-wide chromatin occupancy analyses of Tead4, JunD, and Fra1/2, we find that Tead4 cooperates with AP1 transcription factors to coordinate target gene transcription. We find that Tead-AP1 interaction is JNK independent but engages the SRC1-3 co-activators to promote downstream transcription. Furthermore, we show that Tead-AP1 cooperation regulates the activity of the Dock-Rac/CDC42 module and drives the expression of a unique core set of target genes, thereby directing cell migration and invasion. Together, our data unveil a critical regulatory mechanism underlying Tead- and AP1-controlled transcriptional and functional outputs in cancer cells.
Collapse
Affiliation(s)
- Xiangfan Liu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huapeng Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Mihir Rajurkar
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Qi Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jennifer L Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jianhong Ou
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lihua J Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hira L Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Arthur M Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Joo-Seop Park
- Divisions of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
48
|
Wang T, Mu L, Jin H, Zhang P, Wang Y, Ma X, Pan J, Miao J, Yuan Y. The effects of bufadienolides on HER2 overexpressing breast cancer cells. Tumour Biol 2015; 37:7155-63. [DOI: 10.1007/s13277-015-4381-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/04/2015] [Indexed: 10/22/2022] Open
|
49
|
Transformer2 proteins protect breast cancer cells from accumulating replication stress by ensuring productive splicing of checkpoint kinase 1. Front Chem Sci Eng 2015. [DOI: 10.1007/s11705-015-1540-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Interaction between smoking history and gene expression levels impacts survival of breast cancer patients. Breast Cancer Res Treat 2015. [DOI: 10.1007/s10549-015-3507-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|