51 results on '"Zhikun Ma"'
Search Results
2. Ganetespib targets multiple levels of the receptor tyrosine kinase signaling cascade and preferentially inhibits ErbB2-overexpressing breast cancer cells
- Author
-
Harry Lee, Nipun Saini, Erin W. Howard, Amanda B. Parris, Zhikun Ma, Qingxia Zhao, Ming Zhao, Bolin Liu, Susan M. Edgerton, Ann D. Thor, and Xiaohe Yang
- Subjects
Medicine ,Science - Abstract
Abstract Although ErbB2-targeted therapeutics have significantly improved ErbB2+ breast cancer patient outcomes, therapeutic resistance remains a significant challenge. Therefore, the development of novel ErbB2-targeting strategies is necessary. Importantly, ErbB2 is a sensitive client protein of heat shock protein 90 (HSP90), which regulates client protein folding, maturation, and stabilization. HSP90 inhibition provides an alternative therapeutic strategy for ErbB2-targeted degradation. In particular, ganetespib, a novel HSP90 inhibitor, is a promising agent for ErbB2+ cancers. Nevertheless, the anti-cancer efficacy and clinical application of ganetespib for ErbB2+ breast cancer is largely unknown. In our study, we examined the anti-cancer effects of ganetespib on ErbB2+ BT474 and SKBR3 breast cancer cells, and isogenic paired cancer cell lines with lentivirus-mediated ErbB2 overexpression. Ganetespib potently inhibited cell proliferation, cell cycle progression, survival, and activation/phosphorylation of ErbB2 and key downstream effectors in ErbB2+ breast cancer cells. Moreover, ganetespib decreased the total protein levels of HSP90 client proteins and reduced ErbB2 protein half-life. ErbB2-overexpressing cancer cells were also more sensitive to ganetespib-mediated growth inhibition than parental cells. Ganetespib also strikingly potentiated the inhibitory effects of lapatinib in BT474 and SKBR3 cells. Ultimately, our results support the application of ganetespib-mediated HSP90 inhibition as a promising therapeutic strategy for ErbB2+ breast cancer.
- Published
- 2018
- Full Text
- View/download PDF
3. Bisphenol AF promotes estrogen receptor-positive breast cancer cell proliferation through amphiregulin-mediated crosstalk with receptor tyrosine kinase signaling.
- Author
-
Qingxia Zhao, Erin W Howard, Amanda B Parris, Zhikun Ma, Ying Xing, and Xiaohe Yang
- Subjects
Medicine ,Science - Abstract
Exposure to bisphenol A (BPA), an endocrine-disrupting compound, is associated with increased risk of estrogen-related diseases, including estrogen receptor-positive (ER+) breast cancer. Although bisphenol analogs, i.e. bisphenol AF (BPAF), have replaced BPA in industrial settings, increasing data indicate that these alternatives may have similar or even more potent estrogenic effects. As such, BPAF exhibits increased ER binding affinities than BPA in biochemical assays. However, preclinical studies exploring the effects of BPAF on ER+ breast cancer are missing mechanistic data. Thus, we aimed to characterize the effects of BPAF on MCF-7 and T47D ER+ breast cancer cells with mechanistic insight. We found that BPAF promoted cell growth and cell cycle progression concurrently with BPAF-induced ERα transcriptional activity and ER-RTK signaling activation. ER signaling blockage revealed that BPAF-induced cell proliferation and ER-RTK crosstalk were ER-dependent. Gene expression data demonstrated that AREG is a sensitive target of BPAF in our in vitro models. Importantly, we determined that AREG upregulation is necessary for BPAF-induced cellular responses. Ultimately, our novel finding that AREG mediates BPAF-induced ER-RTK crosstalk in ER+ breast cancer cells supports future studies to characterize the impact of BPAF on human ER+ breast cancer risk and to assess the safety profile of BPAF.
- Published
- 2019
- Full Text
- View/download PDF
4. Multiple indicators of rice remains and the process of rice domestication: A case study in the lower Yangtze River region, China.
- Author
-
Yongchao Ma, Xiaoyan Yang, Xiujia Huan, Yu Gao, Weiwei Wang, Zhao Li, Zhikun Ma, Linda Perry, Guoping Sun, Leping Jiang, Guiyun Jin, and Houyuan Lu
- Subjects
Medicine ,Science - Abstract
The process of rice domestication has been studied for decades based on changing morphological characteristics in assemblages of both macroremains, such as charred seeds and spikelet bases, and microremains, such as phytoliths, esp. bulliform and double-peaked phytoliths. The applicability of these indicators in determining if a specific assemblage is wild or domesticated, however, is rarely discussed. To understand the significance of these indicators in the determination of domestication, we collected 38 archaeological samples from eight Neolithic sites, dating from 10-2ka BP, in the lower Yangtze River region to analyze and compare the changes of these different indicators over eight thousand years. The data demonstrate that the comprehensive analysis of multiple indicators may be the best method to study the process of rice domestication developed thus far. An assemblage of rice remains can be identified as domesticated forms if they meet the following criteria simultaneously: 1) the proportion of domesticated-type bulliform phytoliths is more than 73%; and 2) the proportion of domesticated-type rice spikelet bases is higher than 75%. Furthermore, we found that each indicator tends to change steadily and gradually over time, and each stabilized at a different time, suggesting that the characteristics of domesticated rice developed slowly and successively. Changes of multiple indicators during the period between 10,000-2,000 yr BP indicate that the process of rice domestication in the lower Yangtze River region lasted as long as ca. 6,000 years during the Neolithic, and can be divided into three stages with the turning points in the middle Hemudu-late Majiabang culture (6,500-5,800yr BP) and the late Liangzhu culture (4,600-4,300yr BP).
- Published
- 2018
- Full Text
- View/download PDF
5. Sago-type palms were an important plant food prior to rice in southern subtropical China.
- Author
-
Xiaoyan Yang, Huw J Barton, Zhiwei Wan, Quan Li, Zhikun Ma, Mingqi Li, Dan Zhang, and Jun Wei
- Subjects
Medicine ,Science - Abstract
Poor preservation of plant macroremains in the acid soils of southern subtropical China has hampered understanding of prehistoric diets in the region and of the spread of domesticated rice southwards from the Yangtze River region. According to records in ancient books and archaeological discoveries from historical sites, it is presumed that roots and tubers were the staple plant foods in this region before rice agriculture was widely practiced. But no direct evidences provided to test the hypothesis. Here we present evidence from starch and phytolith analyses of samples obtained during systematic excavations at the site of Xincun on the southern coast of China, demonstrating that during 3,350-2,470 aBC humans exploited sago palms, bananas, freshwater roots and tubers, fern roots, acorns, Job's-tears as well as wild rice. A dominance of starches and phytoliths from palms suggest that the sago-type palms were an important plant food prior to the rice in south subtropical China. We also believe that because of their reliance on a wide range of starch-rich plant foods, the transition towards labour intensive rice agriculture was a slow process.
- Published
- 2013
- Full Text
- View/download PDF
6. p53 inactivation upregulates p73 expression through E2F-1 mediated transcription.
- Author
-
Chaitali Tophkhane, Shi-He Yang, Yunbo Jiang, Zhikun Ma, Dharmalingam Subramaniam, Shrikant Anant, Shingo Yogosawa, Toshiyuki Sakai, Wan-Guo Liu, Susan Edgerton, Ann Thor, and Xiaohe Yang
- Subjects
Medicine ,Science - Abstract
While p73 overexpression has been associated with increased apoptosis in cancer tissues, p73 overexpressing tumors appear to be of high grade malignancy. Why this putative tumor suppressor is overexpressed in cancer cells and what the function of overexpressed p73 is in breast cancers are critical questions to be addressed. By investigating the effect of p53 inactivation on p73 expression, we found that both protein and mRNA levels of TAp73 were increased in MCF-7/p53siRNA cells, MCF-7/p53mt135 cells and HCT-116/p53-/- cells, as compared to wild type control, suggesting that p53 inactivation by various forms upregulates p73. We showed that p53 knockdown induced p73 was mainly regulated at the transcriptional level. However, although p53 has a putative binding site in the TAp73 promoter, deletion of this binding site did not affect p53 knockdown mediated activation of TAp73 promoter. Chromatin immuno-precipitation (ChIP) data demonstrated that loss of p53 results in enhanced occupancy of E2F-1 in the TAp73 promoter. The responsive sequence of p53 inactivation mediated p73 upregulation was mapped to the proximal promoter region of the TAp73 gene. To test the role of E2F-1 in p53 inactivation mediated regulation of p73 transcription, we found that p53 knockdown enhanced E2F-1 dependent p73 transcription, and mutations in E2F-1 binding sites in the TAp73 promoter abrogated p53 knockdown mediated activation of TAp73 promoter. Moreover, we demonstrated that p21 is a mediator of p53-E2F crosstalk in the regulation of p73 transcription. We concluded that p53 knockdown/inactivation may upregulate TAp73 expression through E2F-1 mediated transcriptional regulation. p53 inactivation mediated upregulation of p73 suggests an intrinsic rescuing mechanism in response to p53 mutation/inactivation. These findings support further analysis of the correlation between p53 status and p73 expression and its prognostic/predictive significance in human cancers.
- Published
- 2012
- Full Text
- View/download PDF
7. A Super-Enhancer Driven by FOSL1 Controls miR-21-5p Expression in Head and Neck Squamous Cell Carcinoma
- Author
-
Yuehan Wan, Rosalie G. Hoyle, Nan Xie, Wenjin Wang, Hongshi Cai, Ming Zhang, Zhikun Ma, Gan Xiong, Xiuyun Xu, Zhengxian Huang, Xiqiang Liu, Jiong Li, and Cheng Wang
- Subjects
Cancer Research ,super enhancer ,Cell growth ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biology ,FOSL1 ,medicine.disease ,head and neck squamous cell carcinoma ,Head and neck squamous-cell carcinoma ,Super-enhancer ,Downregulation and upregulation ,Oncology ,microRNA ,Cancer research ,medicine ,metastasis ,miR-21-5p ,Gene ,Transcription factor ,RC254-282 ,Original Research - Abstract
MiR-21-5p is one of the most common oncogenic miRNAs that is upregulated in many solid cancers by inhibiting its target genes at the posttranscriptional level. However, the upstream regulatory mechanisms of miR-21-5p are still not well documented in cancers. Here, we identify a super-enhancer associated with the MIR21 gene (MIR21-SE) by analyzing the MIR21 genomic regulatory landscape in head and neck squamous cell carcinoma (HNSCC). We show that the MIR21-SE regulates miR-21-5p expression in different HNSCC cell lines and disruption of MIR21-SE inhibits miR-21-5p expression. We also identified that a key transcription factor, FOSL1 directly controls miR-21-5p expression by interacting with the MIR21-SE in HNSCC. Moreover, functional studies indicate that restoration of miR-21-5p partially abrogates FOSL1 depletion-mediated inhibition of cell proliferation and invasion. Clinical studies confirmed that miR-21-5p expression is positively correlated with FOSL1 expression. These findings suggest that FOSL1-SE drives miR-21-5p expression to promote malignant progression of HNSCC
- Published
- 2021
- Full Text
- View/download PDF
8. FOSL1 promotes metastasis of head and neck squamous cell carcinoma through super-enhancer-driven transcription program
- Author
-
Jinsong Hou, Cheng Wang, Nan Xie, Rosalie G. Hoyle, Xiqiang Liu, Glen E. Kellogg, Hongshi Cai, Jiong Li, Zhikun Ma, Bo Sun, Ming Zhang, Weixin Cai, Yadong Zhang, Demeng Chen, Hisashi Harada, and Yue Sun
- Subjects
Epithelial-Mesenchymal Transition ,Tumor initiation ,Biology ,Metastasis ,03 medical and health sciences ,Retinoids ,0302 clinical medicine ,Super-enhancer ,Cancer stem cell ,Cell Line, Tumor ,Drug Discovery ,Genetics ,medicine ,Humans ,Neoplasm Metastasis ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Squamous Cell Carcinoma of Head and Neck ,Cancer ,FOSL1 ,medicine.disease ,Head and neck squamous-cell carcinoma ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,stomatognathic diseases ,SNAI2 ,Enhancer Elements, Genetic ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Original Article ,Snail Family Transcription Factors ,Proto-Oncogene Proteins c-fos - Abstract
Previously, we discovered that FOSL1 facilitates the metastasis of head and neck squamous cell carcinoma (HNSCC) cancer stem cells in a spontaneous mouse model. However, the molecular mechanisms remained unclear. Here, we demonstrated that FOSL1 serves as the dominant activating protein 1 (AP1) family member and is significantly upregulated in HNSCC tumor tissues and correlated with metastasis of HNSCC. Mechanistically, FOSL1 exerts its function in promoting tumorigenicity and metastasis predominantly via selective association with Mediators to establish super-enhancers (SEs) at a cohort of cancer stemness and pro-metastatic genes, such as SNAI2 and FOSL1 itself. Depletion of FOSL1 led to disruption of SEs and expression inhibition of these key oncogenes, which resulted in the suppression of tumor initiation and metastasis. We also revealed that the abundance of FOSL1 is positively associated with the abundance of SNAI2 in HNSCC and the high expression levels of FOSL1 and SNAI2 are associated with short overall disease-free survival. Finally, the administration of the FOSL1 inhibitor SR11302 significantly suppressed tumor growth and lymph node metastasis of HNSCC in a patient-derived xenograft model. These findings indicate that FOSL1 is a master regulator that promotes the metastasis of HNSCC through a SE-driven transcription program that may represent an attractive target for therapeutic interventions.
- Published
- 2021
9. Transcriptional super-enhancers control cancer stemness and metastasis genes in squamous cell carcinoma
- Author
-
Jiong Li, Yongxin Yu, Yang Li, Jiaqiang Dong, Zhikun Ma, and Cun-Yu Wang
- Subjects
0301 basic medicine ,BRD4 ,Science ,General Physics and Astronomy ,Antineoplastic Agents ,Cell Cycle Proteins ,Mice, SCID ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Metastasis ,03 medical and health sciences ,Mediator Complex Subunit 1 ,0302 clinical medicine ,Cancer stem cell ,Cell Line, Tumor ,TP63 ,medicine ,Animals ,Humans ,Enhancer ,Head and neck cancer ,Gene ,Polycomb Repressive Complex 1 ,Multidisciplinary ,Squamous Cell Carcinoma of Head and Neck ,Cancer stem cells ,NF-kappa B ,Cancer ,General Chemistry ,Azepines ,FOSL1 ,Triazoles ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,030104 developmental biology ,Enhancer Elements, Genetic ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Lymphatic Metastasis ,Cancer research ,Neoplastic Stem Cells ,Transcription Factors - Abstract
Cancer stem cells (CSCs) play a critical role in invasive growth and metastasis of human head and neck squamous cell carcinoma (HNSCC). Although significant progress has been made in understanding the self-renewal and pro-tumorigenic potentials of CSCs, a key challenge remains on how to eliminate CSCs and halt metastasis effectively. Here we show that super-enhancers (SEs) play a critical role in the transcription of cancer stemness genes as well as pro-metastatic genes, thereby controlling their tumorigenic potential and metastasis. Mechanistically, we find that bromodomain-containing protein 4 (BRD4) recruits Mediators and NF-κB p65 to form SEs at cancer stemness genes such as TP63, MET and FOSL1, in addition to oncogenic transcripts. In vivo lineage tracing reveals that disrupting SEs by BET inhibitors potently inhibited CSC self-renewal and eliminated CSCs in addition to elimination of proliferating non-stem tumor cells in a mouse model of HNSCC. Moreover, disrupting SEs also inhibits the invasive growth and lymph node metastasis of human CSCs isolated from human HNSCC. Taken together, our results suggest that targeting SEs may serve as an effective therapy for HNSCC by eliminating CSCs., Little is known on the transcriptional mechanisms that control cancer stem cell (CSC) self-renewal and stemness. Here the authors show superenhancers (SEs) play a role in the transcription of cancer stemness genes as well as prometastatic genes, thereby controlling their tumorigenic potential; disrupting SEs inhibited CSC self-renewal and eliminated CSCs.
- Published
- 2020
10. Ganetespib targets multiple levels of the receptor tyrosine kinase signaling cascade and preferentially inhibits ErbB2-overexpressing breast cancer cells
- Author
-
Qingxia Zhao, Nipun Saini, Ann D. Thor, Harry Lee, Zhikun Ma, Xiaohe Yang, Bolin Liu, Erin W. Howard, Susan M. Edgerton, Amanda B. Parris, and Ming Zhao
- Subjects
0301 basic medicine ,Cell Survival ,Receptor, ErbB-2 ,Science ,Ganetespib ,Antineoplastic Agents ,Apoptosis ,Breast Neoplasms ,Mice, Transgenic ,Lapatinib ,Receptor tyrosine kinase ,Article ,Hsp90 inhibitor ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Breast cancer ,medicine ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,skin and connective tissue diseases ,neoplasms ,Cell Proliferation ,Multidisciplinary ,biology ,Cell growth ,Chemistry ,Receptor Protein-Tyrosine Kinases ,Drug Synergism ,Triazoles ,medicine.disease ,3. Good health ,G2 Phase Cell Cycle Checkpoints ,030104 developmental biology ,SKBR3 ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,MCF-7 Cells ,Medicine ,Female ,medicine.drug ,Half-Life ,Signal Transduction - Abstract
Although ErbB2-targeted therapeutics have significantly improved ErbB2+ breast cancer patient outcomes, therapeutic resistance remains a significant challenge. Therefore, the development of novel ErbB2-targeting strategies is necessary. Importantly, ErbB2 is a sensitive client protein of heat shock protein 90 (HSP90), which regulates client protein folding, maturation, and stabilization. HSP90 inhibition provides an alternative therapeutic strategy for ErbB2-targeted degradation. In particular, ganetespib, a novel HSP90 inhibitor, is a promising agent for ErbB2+ cancers. Nevertheless, the anti-cancer efficacy and clinical application of ganetespib for ErbB2+ breast cancer is largely unknown. In our study, we examined the anti-cancer effects of ganetespib on ErbB2+ BT474 and SKBR3 breast cancer cells, and isogenic paired cancer cell lines with lentivirus-mediated ErbB2 overexpression. Ganetespib potently inhibited cell proliferation, cell cycle progression, survival, and activation/phosphorylation of ErbB2 and key downstream effectors in ErbB2+ breast cancer cells. Moreover, ganetespib decreased the total protein levels of HSP90 client proteins and reduced ErbB2 protein half-life. ErbB2-overexpressing cancer cells were also more sensitive to ganetespib-mediated growth inhibition than parental cells. Ganetespib also strikingly potentiated the inhibitory effects of lapatinib in BT474 and SKBR3 cells. Ultimately, our results support the application of ganetespib-mediated HSP90 inhibition as a promising therapeutic strategy for ErbB2+ breast cancer.
- Published
- 2018
11. Phenformin inhibits growth and epithelial-mesenchymal transition of ErbB2-overexpressing breast cancer cells through targeting the IGF1R pathway
- Author
-
Zhikun Ma, Amanda B. Parris, Erin W. Howard, Xiaohe Yang, Qingxia Zhao, Zhiying Guo, and Ming Zhao
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.drug_class ,epithelial-mesenchymal transition ,Phenformin ,Receptor tyrosine kinase ,03 medical and health sciences ,chemistry.chemical_compound ,breast cancer ,0302 clinical medicine ,Breast cancer ,Growth factor receptor ,Internal medicine ,MMTV-ErbB2 transgenic mice ,medicine ,skin and connective tissue diseases ,phenformin ,Insulin-like growth factor 1 receptor ,biology ,Biguanide ,business.industry ,medicine.disease ,3. Good health ,IRS1 ,Metformin ,030104 developmental biology ,Endocrinology ,Oncology ,chemistry ,insulin-like growth factor 1 receptor ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,business ,Research Paper ,medicine.drug - Abstract
// Zhiying Guo 1 , Ming Zhao 1 , Erin W. Howard 1 , Qingxia Zhao 1 , Amanda B. Parris 1 , Zhikun Ma 1 and Xiaohe Yang 1 1 Julius L. Chambers Biomedical/Biotechnology Research Institute, Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA Correspondence to: Xiaohe Yang, email: xyang@nccu.edu Keywords: phenformin, breast cancer, epithelial-mesenchymal transition, insulin-like growth factor 1 receptor, MMTV-ErbB2 transgenic mice Received: November 04, 2016 Accepted: June 19, 2017 Published: July 22, 2017 ABSTRACT Reports suggest that metformin, a popular anti-diabetes drug, prevents breast cancer through various systemic effects, including insulin-like growth factor receptor (IGFR) regulation. Although the anti-cancer properties of metformin have been well-studied, reports on a more bioavailable/potent biguanide, phenformin, remain sparse. Phenformin exerts similar functional activity to metformin and has been reported to impede mammary carcinogenesis in rats. Since the effects of phenformin on specific breast cancer subtypes have not been fully explored, we used ErbB2-overexpressing breast cancer cell and animal models to test the anti-cancer potential of phenformin. We report that phenformin (25–75 μM) decreased cell proliferation and impaired cell cycle progression in SKBR3 and 78617 breast cancer cells. Reduced tumor size after phenformin treatment (30 mg/kg/day) was demonstrated in an MMTV-ErbB2 transgenic mouse syngeneic tumor model. Phenformin also blocked epithelial-mesenchymal transition, decreased the invasive phenotype, and suppressed receptor tyrosine kinase signaling, including insulin receptor substrate 1 and IGF1R, in ErbB2-overexpressing breast cancer cells and mouse mammary tumor-derived tissues. Moreover, phenformin suppressed IGF1-stimulated proliferation, receptor tyrosine kinase signaling, and epithelial-mesenchymal transition markers in vitro . Together, our study implicates phenformin-mediated IGF1/IGF1R regulation as a potential anti-cancer mechanism and supports the development of phenformin and other biguanides as breast cancer therapeutics.
- Published
- 2017
- Full Text
- View/download PDF
12. Activation of cancerous inhibitor of PP2A (CIP2A) contributes to lapatinib resistance through induction of CIP2A-Akt feedback loop in ErbB2-positive breast cancer cells
- Author
-
Erin W. Howard, Ming Zhao, Zhiying Guo, Zhikun Ma, Qingxia Zhao, Amanda B. Parris, Susan M. Edgerton, Bolin Liu, Ann D. Thor, Ying Xing, and Xiaohe Yang
- Subjects
0301 basic medicine ,Lapatinib ,CIP2A ,03 medical and health sciences ,chemistry.chemical_compound ,breast cancer ,0302 clinical medicine ,Breast cancer ,ErbB2 ,Downregulation and upregulation ,Medicine ,LY294002 ,skin and connective tissue diseases ,Protein kinase B ,EGFR inhibitors ,business.industry ,lapatinib resistance ,medicine.disease ,PP2A ,3. Good health ,030104 developmental biology ,Oncology ,chemistry ,SKBR3 ,030220 oncology & carcinogenesis ,Cancer research ,Growth inhibition ,business ,Research Paper ,medicine.drug - Abstract
// Ming Zhao 1 , Erin W. Howard 1 , Amanda B. Parris 1 , Zhiying Guo 1 , Qingxia Zhao 1, 2 , Zhikun Ma 1 , Ying Xing 2 , Bolin Liu 3 , Susan M. Edgerton 3 , Ann D. Thor 3 and Xiaohe Yang 1, 4 1 Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biological and Biomedical Sciences, North Carolina Central University, Kannapolis, North Carolina, USA 2 Basic Medical College of Zhengzhou University, Zhengzhou, Henan, P.R. China 3 Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA 4 College of Medicine, Henan University of Sciences and Technology, Luoyang, Henan, P.R. China Correspondence to: Xiaohe Yang, email: xyang@nccu.edu Keywords: CIP2A, lapatinib resistance, ErbB2, breast cancer, PP2A Received: November 05, 2016 Accepted: July 11, 2017 Published: July 19, 2017 ABSTRACT Lapatinib, a small molecule ErbB2/EGFR inhibitor, is FDA-approved for the treatment of metastatic ErbB2-overexpressing breast cancer; however, lapatinib resistance is an emerging clinical challenge. Understanding the molecular mechanisms of lapatinib-mediated anti-cancer activities and identifying relevant resistance factors are of pivotal significance. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified oncoprotein that is overexpressed in breast cancer. Our study investigated the role of CIP2A in the anti-cancer efficacy of lapatinib in ErbB2-overexpressing breast cancer cells. We found that lapatinib concurrently downregulated CIP2A and receptor tyrosine kinase signaling in ErbB2-overexpressing SKBR3 and 78617 cells; however, these effects were attenuated in lapatinib-resistant (LR) cells. CIP2A overexpression rendered SKBR3 and 78617 cells resistant to lapatinib-induced apoptosis and growth inhibition. Conversely, CIP2A knockdown via lentiviral shRNA enhanced cell sensitivity to lapatinib-induced growth inhibition and apoptosis. Results also suggested that lapatinib downregulated CIP2A through regulation of protein stability. We further demonstrated that lapatinib-induced CIP2A downregulation can be recapitulated by LY294002, suggesting that Akt mediates CIP2A upregulation. Importantly, lapatinib induced differential CIP2A downregulation between parental BT474 and BT474/LR cell lines. Moreover, CIP2A shRNA knockdown significantly sensitized the BT474/LR cells to lapatinib. Collectively, our results demonstrate that CIP2A is a molecular target and resistance factor of lapatinib with a critical role in lapatinib-induced cellular responses, including the inhibition of the CIP2A-Akt feedback loop. Further investigation of lapatinib-mediated CIP2A regulation will advance our understanding of lapatinib-associated anti-tumor activities and drug resistance.
- Published
- 2017
- Full Text
- View/download PDF
13. GASC1-Adapted Neoadjuvant Chemotherapy for Resectable Esophageal Squamous Cell Carcinoma: A Prospective Clinical Biomarker Trial
- Author
-
Qiangjian Mi, Ruonan Li, Zhikun Ma, Wanying Li, Shegan Gao, You-Jia Mi, Ya-Fei Zhu, Xiang Yuan, Dejiu Kong, Jinyu Kong, Ruinuo Jia, Na Li, and Bingbing Wang
- Subjects
0301 basic medicine ,Oncology ,Tumor Regression Grade ,Chemotherapy ,medicine.medical_specialty ,Article Subject ,business.industry ,medicine.medical_treatment ,Significant difference ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Esophageal squamous cell carcinoma ,Clinical biomarker ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cancer stem cell ,030220 oncology & carcinogenesis ,Internal medicine ,Potential biomarkers ,medicine ,Immunohistochemistry ,business ,RC254-282 ,Research Article - Abstract
Neoadjuvant chemotherapy (NCT) is a standard care for esophageal squamous cell carcinoma (ESCC), but the efficacy is unsatisfactory. Cancer stem cells (CSCs) play key roles in chemotherapy resistance. Gene amplified in squamous cell carcinoma 1 (GASC1) is a neoteric gene in stemness maintaining of ESCC. We aimed to reveal whether GASC1 could be a predictive biomarker for NCT in ESCC. ESCC patients (T2-4N0-2M0) were evaluated for GASC1 expression using immunohistochemical staining and classified as GASC1-low group (GLG) and GASC1-high group (GHG). NCT was delivered in two cycles and then the surgery was completed. Primary endpoints were tumor regression grade (TRG) and objective response rate (ORR); secondary endpoints were radical surgical resection (R0) rate and three-year overall survival (OS). 60 patients were eligible with evaluable outcomes: 24 in GHG and 36 in GLG. Between GHG and GLG, TRG1, TRG2, TRG3, and TRG4 were 0 : 16.7%, 20.8% : 41.7%, 58.3% : 36.1%, and 20.8% : 5.6%, respectively (P=0.006); ORR and R0 rate were 33.3% : 69.4% (P=0.006) and 75% : 94.4% (P=0.046), respectively; the median OS was 20 : 32 (months) (P=0.0356). No significant difference in the three-year OS was observed between GHG and GLG: 29.2% : 41.7% (P=0.24). Furthermore, the GASC1 expression level was associated with poor OS independent of other factors by univariate and multivariate analyses. Therefore, GASC1 might be a potential biomarker to predict NCT efficacy for ESCC.
- Published
- 2020
14. Bisphenol AF promotes estrogen receptor-positive breast cancer cell proliferation through amphiregulin-mediated crosstalk with receptor tyrosine kinase signaling
- Author
-
Ying Xing, Xiaohe Yang, Amanda B. Parris, Qingxia Zhao, Zhikun Ma, and Erin W. Howard
- Subjects
Bisphenol ,Physiology ,Estrogen receptor ,010501 environmental sciences ,01 natural sciences ,Biochemistry ,Receptor tyrosine kinase ,Endocrinology ,Cell Signaling ,Endocrine Signaling ,Breast Tumors ,Medicine and Health Sciences ,Enzyme assays ,Colorimetric assays ,Bioassays and physiological analysis ,0303 health sciences ,Multidisciplinary ,MTT assay ,biology ,Chemistry ,3. Good health ,Enzymes ,Oncology ,Cellular Crosstalk ,Cell Processes ,Physical Sciences ,MCF-7 Cells ,Medicine ,Female ,Oxidoreductases ,Luciferase ,Research Article ,Signal Transduction ,Proto-Oncogene Proteins B-raf ,medicine.drug_class ,Science ,Breast Neoplasms ,Amphiregulin ,Cell Growth ,03 medical and health sciences ,Breast cancer ,Downregulation and upregulation ,Phenols ,Breast Cancer ,medicine ,Humans ,Benzhydryl Compounds ,030304 developmental biology ,0105 earth and related environmental sciences ,Cell Proliferation ,Endocrine Physiology ,Cell growth ,Chemical Compounds ,Estrogen Receptor alpha ,Cancers and Neoplasms ,Biology and Life Sciences ,Proteins ,Cell Biology ,medicine.disease ,Research and analysis methods ,Estrogen ,Biochemical analysis ,Cancer research ,biology.protein ,Enzymology - Abstract
Exposure to bisphenol A (BPA), an endocrine-disrupting compound, is associated with increased risk of estrogen-related diseases, including estrogen receptor-positive (ER+) breast cancer. Although bisphenol analogs, i.e. bisphenol AF (BPAF), have replaced BPA in industrial settings, increasing data indicate that these alternatives may have similar or even more potent estrogenic effects. As such, BPAF exhibits increased ER binding affinities than BPA in biochemical assays. However, preclinical studies exploring the effects of BPAF on ER+ breast cancer are missing mechanistic data. Thus, we aimed to characterize the effects of BPAF on MCF-7 and T47D ER+ breast cancer cells with mechanistic insight. We found that BPAF promoted cell growth and cell cycle progression concurrently with BPAF-induced ERα transcriptional activity and ER-RTK signaling activation. ER signaling blockage revealed that BPAF-induced cell proliferation and ER-RTK crosstalk were ER-dependent. Gene expression data demonstrated that AREG is a sensitive target of BPAF in our in vitro models. Importantly, we determined that AREG upregulation is necessary for BPAF-induced cellular responses. Ultimately, our novel finding that AREG mediates BPAF-induced ER-RTK crosstalk in ER+ breast cancer cells supports future studies to characterize the impact of BPAF on human ER+ breast cancer risk and to assess the safety profile of BPAF.
- Published
- 2019
15. Effect of perioperative administration of dexmedetomidine on delirium after cardiac surgery in elderly patients: a double-blinded, multi-center, randomized study
- Author
-
Jin Jin, Zhikun Ma, Cunxian Shi, Leyan Qiao, Tao Li, and Jiahai Ma
- Subjects
Male ,medicine.medical_specialty ,Time Factors ,Randomization ,Anesthesia, General ,anesthesia ,elderly patients ,law.invention ,postoperative delirium ,03 medical and health sciences ,0302 clinical medicine ,Double-Blind Method ,Randomized controlled trial ,law ,Intensive care ,medicine ,Humans ,Hypnotics and Sedatives ,Postoperative Period ,030212 general & internal medicine ,Cardiac Surgical Procedures ,Dexmedetomidine ,Propofol ,Aged ,Analgesics ,business.industry ,dexmedetomidine ,Delirium ,General Medicine ,Perioperative ,Cardiac surgery ,Analgesics, Opioid ,Clinical Trial Report ,Anesthesia ,Clinical Interventions in Aging ,Female ,Geriatrics and Gerontology ,medicine.symptom ,business ,cardiac surgery ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Cunxian Shi,* Jin Jin,* Leyan Qiao, Tao Li, Jiahai Ma, Zhikun Ma Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, Shandong, China*These authors contributed equally to this workObjective: Postoperative delirium (POD) is a serious complication in elderly patients undergoing cardiac surgery. This study was aimed at investigating the effect of perioperative administration of dexmedetomidine for general anesthesia maintenance on occurrence and duration of POD in elderly patients after cardiac surgery.Methods: One hundred and sixty-four patients were enrolled after cardiac surgery between June 2009 and December 2016. Patients were assigned by a computer-generated randomization sequence in a 1:1 ratio to receive dexmedetomidine general anesthesia maintenance or propofol general anesthesia maintenance. POD was assessed every day with confusion assessment method for intensive care units (ICU) during the first 5 postoperative days.Results: There was no significance in incidence of POD between the dexmedetomidine group and the propofol group (P=0.0758). In patients treated with dexmedetomidine, the median onset time of delirium was delayed (second day vs first day) and the duration of delirium reduced (2 days vs 3 days) when compared with propofol-treated patients. The dexmedetomidine-treated patients also displayed a lower VAS score and less opiate analgesic consumption. No difference was observed in respect to other postoperative outcomes.Conclusion: For elderly patients, perioperative administration of dexmedetomidine reduced incidence, delayed onset and shortened duration of POD after cardiac surgery.Keywords: dexmedetomidine, postoperative delirium, anesthesia, cardiac surgery, elderly patients
- Published
- 2019
16. Plant utilization at the Jiangxigou site during the middle Holocene
- Author
-
Weng Zhang, E. Chongyi, Zhikun Ma, GuangLiang Hou, and Haicheng Wei
- Subjects
Starch grain ,010506 paleontology ,Archeology ,Setaria ,Plateau ,geography.geographical_feature_category ,060102 archaeology ,biology ,Ecology ,06 humanities and the arts ,medicine.disease_cause ,biology.organism_classification ,01 natural sciences ,Prehistory ,Geography ,Pollen ,Botany ,Foxtail ,medicine ,0601 history and archaeology ,Domestication ,Holocene ,0105 earth and related environmental sciences - Abstract
Agricultural activities on the Qinghai–Tibetan Plateau (QTP) are difficult due to the hostile environment. Despite the difficulties, recently early agriculture has been documented by Holocene plant remains on the QTP. The details of the time, place, and mechanisms of agricultural origin as well as the evolution of agriculture on the QTP are still lacking. The Jiangxigou site (JXG2) contains artifacts dominated by microliths from the early to middle Holocene and provides important information on agricultural origins and evolution of prehistoric culture on the QTP. Here we report ancient starch grains and pollen extracted from ceramics and deposits excavated from each layer. The starch remains include 21 grains identical to starches from millets ( Setaria spp. and Panicum spp.), probably including 24% domesticated foxtail millet ( Setaria italica ), indicating that humans at 3000 m above sea level (masl) in the northeastern margin of the Qinghai–Tibetan Plateau (QTP) began to use millets ca. 5600 cal BP. Considering archeological evidence, the reasons for millet utilization by plateau residents may be associated with the expansion and influence of the Yangshao culture via the Loess Plateau.
- Published
- 2016
- Full Text
- View/download PDF
17. FGFR1 overexpression renders breast cancer cells resistant to metformin through activation of IRS1/ERK signaling
- Author
-
Amanda B. Parris, Yudan Wu, Zhikun Ma, Qiong Cheng, Xiaohe Yang, Lingfei Kong, and Yujie Shi
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,endocrine system diseases ,MAP Kinase Signaling System ,Breast Neoplasms ,Pyrogallol ,Receptor, IGF Type 1 ,03 medical and health sciences ,0302 clinical medicine ,Mediator ,Breast cancer ,medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Molecular Biology ,Protein kinase B ,Insulin-like growth factor 1 receptor ,Sulfonamides ,business.industry ,TOR Serine-Threonine Kinases ,Fibroblast growth factor receptor 1 ,digestive, oral, and skin physiology ,nutritional and metabolic diseases ,Cell Biology ,medicine.disease ,Metformin ,IRS1 ,Gene Expression Regulation, Neoplastic ,stomatognathic diseases ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Insulin Receptor Substrate Proteins ,MCF-7 Cells ,Cancer research ,Female ,business ,medicine.drug - Abstract
Metformin has been suggested as an anti-cancer agent. However, increasing reports show that some tumors are resistant to metformin. Identification of factors affecting metformin mediated cancer therapy is of great significance. FGFR1 is a receptor-tyrosine-kinase that is frequently overexpressed in breast cancer, which is associated with poor-prognosis. To investigate the effect of FGFR1 overexpression on metformin-induced inhibition of breast cancer cells, we demonstrated that FGFR1 overexpression rendered MCF-7 and T47D cells resistant to metformin. In particular, we found that, in addition to AKT and ERK1/2 activation, FGFR1-induced activation of IRS1 and IGF1R, key regulators connecting metabolism and cancer, was associated with metformin resistance. Targeting IRS with IRS1 KO or IRS inhibitor NT157 significantly sensitized FGFR1 overexpressing cells to metformin. Combination of NT157 with metformin induced enhanced inhibition of p-IGF1R, p-ERK1/2 and p-mTOR. Moreover, we demonstrated that IRS1 functions as a critical mediator of the crosstalk between FGFR1 and IGF1R pathways, which involves a feedback loop between IRS1 and MAPK/ERK. Our study highlights the significance of FGFR1 status and IRS1 activation in metformin-resistance, which will facilitate the development of strategies targeting FGFR overexpression-associated metformin resistance.
- Published
- 2021
- Full Text
- View/download PDF
18. DMBA promotes ErbB2‑mediated carcinogenesis via ErbB2 and estrogen receptor pathway activation and genomic instability
- Author
-
Stanley D. Kosanke, Shibo Li, Xia Cao, Yunbo Jiang, Zhikun Ma, Erin W. Howard, Xiaoshan Feng, Young Mi Kim, Amanda B. Parris, Shihe Yang, and Xiaohe Yang
- Subjects
0301 basic medicine ,Cancer Research ,Receptor, ErbB-2 ,9,10-Dimethyl-1,2-benzanthracene ,DMBA ,Estrogen receptor ,Apoptosis ,Mice, Transgenic ,Biology ,medicine.disease_cause ,estrogen receptor pathway ,Mice ,03 medical and health sciences ,breast cancer ,0302 clinical medicine ,Chromosome instability ,MMTV-ErbB2 transgenic mice ,Tumor Cells, Cultured ,medicine ,Animals ,7,12-dimethylbenz[a]anthracene ,Epidermal growth factor receptor ,skin and connective tissue diseases ,neoplasms ,Cell Proliferation ,Mammary tumor ,receptor tyrosine kinase pathway ,Mammary Neoplasms, Experimental ,Cancer ,Articles ,General Medicine ,genomic instability ,medicine.disease ,3. Good health ,Cell Transformation, Neoplastic ,030104 developmental biology ,Receptors, Estrogen ,Oncology ,030220 oncology & carcinogenesis ,Carcinogens ,Cancer research ,biology.protein ,Female ,Tumor promotion ,Carcinogenesis - Abstract
Environmental factors, including 7,12-dimethylbenz [a]anthracene (DMBA) exposure, and genetic predisposition, including ErbB2 overexpression/amplification, have been demonstrated to increase breast cancer susceptibility. Although DMBA- and ErbB2-mediated breast cancers are well-studied in their respective models, key interactions between environmental and genetic factors on breast cancer risk remain unclear. Therefore, the present study aimed to investigate the effect of DMBA exposure on ErbB2-mediated mammary tumorigenesis. MMTV-ErbB2 transgenic mice exposed to DMBA (1 mg) via weekly oral gavage for 6 weeks exhibited significantly enhanced mammary tumor development, as indicated by reduced tumor latency and increased tumor multiplicity compared with control mice. Whole mount analysis of premalignant mammary tissues from 15-week-old mice revealed increased ductal elongation and proliferative index in DMBA-exposed mice. Molecular analyses of premalignant mammary tissues further indicated that DMBA exposure enhanced epidermal growth factor receptor (EGFR)/ErbB2 and estrogen receptor (ER) signaling, which was associated with increased mRNA levels of EGFR/ErbB2 family members and ER-targeted genes. Furthermore, analysis of tumor karyotypes revealed that DMBA-exposed tumors displayed more chromosomal alterations compared with control tumors, implicating DMBA-induced chromosomal instability in tumor promotion in this model. Together, the data suggested that DMBA-induced deregulation of EGFR/ErbB2-ER pathways plays a critical role in the enhanced chromosomal instability and promotion of ErbB2-mediated mammary tumorigenesis. The study highlighted gene-environment interactions that may increase risk of breast cancer, which is a critical clinical issue.
- Published
- 2018
- Full Text
- View/download PDF
19. Down-regulation of C/EBP homologous protein (CHOP) expression in gastric cardia adenocarcinoma: Their relationship with clinicopathological parameters and prognostic significance
- Author
-
Shegan Gao, Zhikun Ma, Zhenguo Shi, Xiaojuan Zhu, Xiaoshan Feng, Li Sanqiang, and Zhu Shanshan
- Subjects
Male ,Pathology ,medicine.medical_specialty ,genetic structures ,Down-Regulation ,Adenocarcinoma ,CHOP ,Downregulation and upregulation ,Western blot ,Stomach Neoplasms ,immune system diseases ,hemic and lymphatic diseases ,Humans ,Medicine ,skin and connective tissue diseases ,Survival rate ,Hepatology ,medicine.diagnostic_test ,business.industry ,Gastroenterology ,Cancer ,Middle Aged ,Prognosis ,Gastric Cardia Adenocarcinoma ,medicine.disease ,Survival Rate ,cardiovascular system ,Cancer research ,Immunohistochemistry ,Female ,Gastritis ,medicine.symptom ,business ,Transcription Factor CHOP - Abstract
C/EBP homologous protein (CHOP) is a multi-functional protein involved in the apoptosis pathway of endoplasmic reticulum stress (ERS) and is related to cancer progression. The purpose of this study was to assess CHOP expression as a prognostic biomarker in gastric cardia adenocarcinoma (GCA).The levels of CHOP mRNA and protein in GCA and matched adjacent non-cancerous tissues were evaluated by quantitative real-time-polymerase chain reaction (qRT-PCR) and western blot. Furthermore, the CHOP protein expression and localization were examined by immunohistochemistry in GCA and corresponding adjacent non-cancerous tissues, gastritis and normal cardiac tissues. The association of CHOP expression with clinical pathological parameters and prognosis of GCA patients was statistically analyzed.Compared with adjacent non-cancerous tissues, the CHOP was down-regulated at mRNA and protein levels in GCA (P0.01). In addition, immunohistochemistry analysis showed that CHOP positivity was lower in GCA than that in paired adjacent non-cancerous tissues, gastritis and normal tissues (P0.01). CHOP expression rate gradually decreased with an increase in clinical stage, tumor differentiation and lymph node metastasis of GCA (P0.05). Kaplan-Meier survival analysis revealed that low expression of CHOP correlated with poor prognosis of GCA patients. Moreover, univariate and multivariate analyses showed that CHOP was an independent prognostic marker for overall survival of GCA patients.Our results suggest that low CHOP expression predicts poor prognosis of GCA patients, and CHOP may be potentially a prognostic biomarker for GCA.
- Published
- 2015
- Full Text
- View/download PDF
20. Preoperative serum immunoglobulin G and A antibodies to Porphyromonas gingivalis are potential serum biomarkers for the diagnosis and prognosis of esophageal squamous cell carcinoma
- Author
-
Xiang Yuan, Xiaoshan Feng, Shegan Gao, Guang-Chao Wang, Hua Wei, Yijun Qi, Zhikun Ma, Jun-Qiang Yang, and Chen Zhao
- Subjects
Male ,0301 basic medicine ,Immunoglobulin A ,Cancer Research ,Esophageal Neoplasms ,Gastroenterology ,Immunoglobulin G ,Metastasis ,0302 clinical medicine ,Diagnosis ,Bacteroidaceae Infections ,Lymph node ,biology ,Middle Aged ,Prognosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Antibodies, Bacterial ,Survival Rate ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Immunoglobulin G/A ,Female ,Antibody ,Porphyromonas gingivalis ,Research Article ,medicine.medical_specialty ,Enzyme-Linked Immunosorbent Assay ,lcsh:RC254-282 ,03 medical and health sciences ,Esophageal squamous cell carcinoma ,Internal medicine ,Preoperative Care ,Biomarkers, Tumor ,Genetics ,medicine ,Humans ,neoplasms ,Survival rate ,Antigens, Bacterial ,business.industry ,Cancer ,medicine.disease ,biology.organism_classification ,digestive system diseases ,030104 developmental biology ,biology.protein ,business ,Follow-Up Studies - Abstract
Background The key-stone-pathogen, Porphyromonas gingivalis associates not only with periodontal diseases but with a variety of other chronic diseases such as cancer. We previously reported an association between the presence of Porphyromonas gingivalis in esophageal squamous cell carcinoma (ESCC) and its progression. We now report the diagnostic and prognostic potential of serum immunoglobulin G and A antibodies (IgG/A) against Porphyromonas gingivalis for ESCC. Methods An enzyme-linked immunosorbent assay (ELISA) was used to determine the serum levels of Porphyromonas gingivalis IgG and IgA in 96 cases with ESCC, 50 cases with esophagitis and 80 healthy controls. Results The median serum levels of IgG and IgA for P. gingivalis were significantly higher in ESCC patients than non-ESCC controls. P. gingivalis IgG and IgA in serum demonstrated sensitivities/specificities of 29.17%/96.90% and 52.10%/70.81%, respectively, and combination of IgG and IgA produced a sensitivity/specificity of 68.75%/68.46%. The diagnostic performance of serum P. gingivalis IgA for early ESCC was superior to that of IgG (54.54% vs. 20.45%). Furthermore, high serum levels of P. gingivalis IgG or IgA were associated with worse prognosis of ESCC patients, in particular for patients with stage 0-IIor negative lymphnode metastasis, and ESCC patients with high levels of both IgG and IgA had the worst prognosis. Multivariate analysis revealed that lymph node status, IgG and IgA were independent prognostic factors. Conclusions The IgG and IgA for P. gingivalis are potential serum biomarkers for ESCC and combination of IgG and IgA improves the diagnostic and prognostic performance. Furthermore, serum P. gingivalis IgG and IgA can detect early stage ESCC. Electronic supplementary material The online version of this article (10.1186/s12885-017-3905-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
21. Multiple indicators of rice remains and the process of rice domestication: A case study in the lower Yangtze River region, China
- Author
-
Weiwei Wang, Guiyun Jin, Xiujia Huan, Yu Gao, Guoping Sun, Leping Jiang, Zhikun Ma, Yong Chao Ma, Zhao Li, Linda Perry, Houyuan Lu, and Xiaoyan Yang
- Subjects
Leaves ,Social Sciences ,Stone Age ,Plant Science ,010502 geochemistry & geophysics ,01 natural sciences ,Domestication ,Assemblage (archaeology) ,History, Ancient ,Multidisciplinary ,biology ,Fossils ,Plant Anatomy ,Eukaryota ,Geology ,Plants ,Freshwater Fish ,Archaeology ,Experimental Organism Systems ,Neolithic Period ,Seeds ,Vertebrates ,Freshwater fish ,Yangtze river ,Medicine ,Research Article ,China ,010506 paleontology ,Animal Types ,Science ,Research and Analysis Methods ,Rivers ,Plant and Algal Models ,Animals ,Domestic Animals ,Grasses ,0105 earth and related environmental sciences ,Organisms ,Biology and Life Sciences ,Oryza ,Geologic Time ,biology.organism_classification ,Fish ,Agronomy ,Archaeological Dating ,Animal Studies ,Earth Sciences ,Period (geology) ,Rice ,Edible Grain ,Zoology - Abstract
The process of rice domestication has been studied for decades based on changing morphological characteristics in assemblages of both macroremains, such as charred seeds and spikelet bases, and microremains, such as phytoliths, esp. bulliform and double-peaked phytoliths. The applicability of these indicators in determining if a specific assemblage is wild or domesticated, however, is rarely discussed. To understand the significance of these indicators in the determination of domestication, we collected 38 archaeological samples from eight Neolithic sites, dating from 10-2ka BP, in the lower Yangtze River region to analyze and compare the changes of these different indicators over eight thousand years. The data demonstrate that the comprehensive analysis of multiple indicators may be the best method to study the process of rice domestication developed thus far. An assemblage of rice remains can be identified as domesticated forms if they meet the following criteria simultaneously: 1) the proportion of domesticated-type bulliform phytoliths is more than 73%; and 2) the proportion of domesticated-type rice spikelet bases is higher than 75%. Furthermore, we found that each indicator tends to change steadily and gradually over time, and each stabilized at a different time, suggesting that the characteristics of domesticated rice developed slowly and successively. Changes of multiple indicators during the period between 10,000–2,000 yr BP indicate that the process of rice domestication in the lower Yangtze River region lasted as long as ca. 6,000 years during the Neolithic, and can be divided into three stages with the turning points in the middle Hemudu-late Majiabang culture (6,500–5,800yr BP) and the late Liangzhu culture (4,600–4,300yr BP).
- Published
- 2018
22. Caloric restriction inhibits mammary tumorigenesis in MMTV-ErbB2 transgenic mice through the suppression of ER and ErbB2 pathways and inhibition of epithelial cell stemness in premalignant mammary tissues
- Author
-
Lingfei Kong, Yunbo Jiang, Amanda B. Parris, Zhikun Ma, Yujie Shi, Shihe Yang, Erin W. Howard, and Xiaohe Yang
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,Receptor, ErbB-2 ,Mammary gland ,Cell ,Blotting, Western ,Estrogen receptor ,Mice, Transgenic ,medicine.disease_cause ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Mice ,medicine ,Animals ,Humans ,Progenitor cell ,skin and connective tissue diseases ,Mammary Glands, Human ,Caloric Restriction ,Cell Proliferation ,Mammary tumor ,Cell growth ,Chemistry ,Mammary Neoplasms, Experimental ,Epithelial Cells ,General Medicine ,Flow Cytometry ,Immunohistochemistry ,030104 developmental biology ,medicine.anatomical_structure ,Receptors, Estrogen ,Cancer research ,Female ,Stem cell ,Inflammation, Microenvironment and Prevention ,Signal Transduction - Abstract
Caloric intake influences the onset of many diseases, including cancer. In particular, caloric restriction (CR) has been reported to suppress mammary tumorigenesis in various models. However, the underlying cancer preventive mechanisms have not been fully explored. To this end, we aimed to characterize the anticancer mechanisms of CR using MMTV-ErbB2 transgenic mice, a well-established spontaneous ErbB2-overexpressing mammary tumor model, by focusing on cellular and molecular changes in premalignant tissues. In MMTV-ErbB2 mice with 30% CR beginning at 8 weeks of age, mammary tumor development was dramatically inhibited, as exhibited by reduced tumor incidence and increased tumor latency. Morphogenic mammary gland analyses in 15- and 20-week-old mice indicated that CR significantly decreased mammary epithelial cell (MEC) density and proliferative index. To understand the underlying mechanisms, we analyzed the effects of CR on mammary stem/progenitor cells. Results from fluorescence-activated cell sorting analyses showed that CR modified mammary tissue hierarchy dynamics, as evidenced by decreased luminal cells (CD24(high)CD49f(low)), putative mammary reconstituting unit subpopulation (CD24(high)CD49f(high)) and luminal progenitor cells (CD61(high)CD49f(high)). Mammosphere and colony-forming cell assays demonstrated that CR significantly inhibited mammary stem cell self-renewal and progenitor cell numbers. Molecular analyses indicated that CR concurrently inhibited estrogen receptor (ER) and ErbB2 signaling. These molecular changes were accompanied by decreased mRNA levels of ER-targeted genes and epidermal growth factor receptor/ErbB2 family members and ligands, suggesting ER-ErbB2 signaling cross-talk. Collectively, our data demonstrate that CR significantly impacts ER and ErbB2 signaling, which induces profound changes in MEC reprogramming, and mammary stem/progenitor cell inhibition is a critical mechanism of CR-mediated breast cancer prevention.
- Published
- 2017
23. FGFR inhibitor, AZD4547, impedes the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice
- Author
-
Qingxia Zhao, Amanda B. Parris, Zhiying Guo, Erin W. Howard, Zhikun Ma, Xiaohe Yang, Ming Zhao, and Ying Xing
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cell Survival ,Receptor, ErbB-2 ,lcsh:Medicine ,Gene Expression ,Antineoplastic Agents ,Mammary Neoplasms, Animal ,Mice, Transgenic ,Biology ,Receptor tyrosine kinase ,Article ,Piperazines ,03 medical and health sciences ,Mice ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Cell Self Renewal ,lcsh:Science ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Mammary tumor ,Multidisciplinary ,Stem Cells ,lcsh:R ,Cell Cycle ,Wnt signaling pathway ,FGFR Inhibitor AZD4547 ,Mammary Neoplasms, Experimental ,Epithelial Cells ,Receptors, Fibroblast Growth Factor ,3. Good health ,030104 developmental biology ,Endocrinology ,Fibroblast growth factor receptor ,Cancer cell ,Benzamides ,biology.protein ,Cancer research ,Pyrazoles ,lcsh:Q ,Female ,Signal transduction ,Precancerous Conditions ,Signal Transduction - Abstract
The fibroblast growth factor receptor (FGFR) family of receptor tyrosine kinases (RTKs) regulates signaling pathways involved in cell proliferation and differentiation. Currently, the anti-tumor properties of FGFR inhibitors are being tested in preclinical and clinical studies. Nevertheless, reports on FGFR inhibitor-mediated breast cancer prevention are sparse. In this study, we investigated the anti-cancer benefits of AZD4547, an FGFR1-3 inhibitor, in ErbB2-overexpressing breast cancer models. AZD4547 (1–5 µM) demonstrated potent anti-proliferative effects, inhibition of stemness, and suppression of FGFR/RTK signaling in ErbB2-overexpressing human breast cancer cells. To study the in vivo effects of AZD4547 on mammary development, mammary epithelial cell (MEC) populations, and oncogenic signaling, MMTV-ErbB2 transgenic mice were administered AZD4547 (2–6 mg/kg/day) for 10 weeks during the ‘risk window’ for mammary tumor development. AZD4547 significantly inhibited ductal branching and MEC proliferation in vivo, which corroborated the in vitro anti-proliferative properties. AZD4547 also depleted CD24/CD49f-sorted MEC populations, as well as the CD61highCD49fhigh tumor-initiating cell-enriched population. Importantly, AZD4547 impaired stem cell-like characteristics in primary MECs and spontaneous tumor cells. Moreover, AZD4547 downregulated RTK, mTOR, and Wnt/β-catenin signaling pathways in premalignant mammary tissues. Collectively, our data provide critical preclinical evidence for AZD4547 as a potential breast cancer preventative and therapeutic agent.
- Published
- 2017
24. Inhibition of Glycogen Synthase Kinase 3 beta (GSK3β) Suppresses the Progression of Esophageal Squamous Cell Carcinoma by Modifying STAT3 Activity
- Author
-
Xiaoshan Feng, Shuoguo Li, Shegan Gao, Xiang Yuan, Huizhi Wang, Zhen Gu, Xiaoxian Duan, and Zhikun Ma
- Subjects
0301 basic medicine ,Male ,STAT3 Transcription Factor ,Cancer Research ,Esophageal Neoplasms ,Cell Survival ,Pyridines ,Biology ,Article ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Phosphorylation ,STAT3 ,Molecular Biology ,GSK3B ,neoplasms ,Glycogen Synthase Kinase 3 beta ,Cancer ,Cell migration ,Tyrphostins ,medicine.disease ,Prognosis ,Survival Analysis ,In vitro ,digestive system diseases ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Carcinoma, Squamous Cell ,Disease Progression ,Female ,Esophageal Squamous Cell Carcinoma ,Lithium Chloride ,Signal Transduction - Abstract
Although GSK3β has been reported to have contrasting effects on the progression of different tumors, it's possible functions in esophageal squamous cell carcinoma (ESCC) and the related molecular mechanisms remain unknown. Here, we investigated the expression, function, and molecular mechanism of GSK3β in the development of ESCC in vitro and in vivo. Though the expression of total GSK3β was significantly increased, the phosphorylated (inactivated) form of GSK3β (Ser9) was concurrently decreased in the cancerous tissues of patients with ESCC compared with controls, suggesting that GSK3β activity was enhanced in cancerous tissues. Further pathological data analysis revealed that higher GSK3β expression was associated with poorer differentiation, higher metastasis rates, and worse prognosis of ESCC. These results were confirmed in different ESCC cell lines using a pharmacological inhibitor and specific siRNA to block GSK3β. Using a cancer phospho-antibody array, we found that STAT3 is a target of GSK3β. GSK3 inhibition reduced STAT3 phosphorylation, and overexpression of constitutively active GSK3β had the opposite effect. Moreover, STAT3 inhibition mimicked the effects of GSK3β inhibition on ESCC cell migration and viability, while overexpression of a plasmid encoding mutant STAT3 (Y705F) abrogated these effects, and these results were further substantiated by clinicopathological data. In addition, a GSK3 inhibitor (LiCl) and/or STAT3 inhibitor (WP-1066) efficiently suppressed the growth of ESCC cells in a xenograft tumor model. Altogether, these results reveal that higher GSK3β expression promotes ESCC progression through STAT3 in vitro and in vivo, and GSK3β-STAT3 signaling could be a potential therapeutic target for ESCC treatment.
- Published
- 2017
25. Abstract 3009: FGFR1 overexpression ER positive breast cancer cells confer the cells resistant to Palbociclib via upregulation of RTK-ER signaling
- Author
-
Xiaohe Yang, Amanda B. Parris, Lingfei Kong, Qiong Cheng, Zhikun Ma, Yujie Shi, and Yudan Wu
- Subjects
Cancer Research ,Chemistry ,Cancer ,Estrogen receptor ,Palbociclib ,medicine.disease ,stomatognathic diseases ,Oncology ,Cell culture ,Cancer cell ,Cancer research ,medicine ,skin and connective tissue diseases ,Cyclin D3 ,Cyclin B1 ,Clonogenic assay - Abstract
Palbociclib is the first CDK4/6 inhibitor approved by the FDA for treating ER+/HER2- breast cancer. Palbociclib resistance is emerging as a clinical challenge. Identification of the factors that determine the sensitivity to Palbociclib is of pivotal significance. Fibroblast Growth Factor Receptor (FGFR) overexpression is frequently detected in breast cancer, which has been associated with poor prognosis. To investigate the impact of FGFR overexpression on Palbociclib resistance, we first established stable sublines of MCF7 and T47D cells that overexpress FGFR1, and examined their responses to Palbociclib. Data from MTT and clonogenic assays demonstrated that overexpression of FGFR1 rendered ER+ MCF7 and T47D cells resistant to Palbociclib significantly. Cell cycle analysis showed that the percentage of cells in S phase in Palbociclib treated MCF7/FGFR1 and T47D/FGFR1 cells was significantly higher than that of the control. FGFR overexpression also significantly attenuated Palbociclib-associated inhibition of cancer cell invasion. We further found that Palbociclib-induced inhibition of cancer cell stemness, as indicated by colony formation in soft agar and mammosphere assays, was significantly reduced in MCF7/FGFR1 and T47D/FGFR1 cells, as compared to control. The data indicate that FGFR1 overexpression in ER+ breast cancer cells are resistant to Palbociclib. We next examined the signaling of the key pathways in the paired cell lines treated with Palbociclib. We found that Palbociclib-induced inhibition of phospho-Rb, E2F, cyclin D3, cyclin B1, cdc-2, c-myc, and cdc-25 was significantly attenuated in MCF-7/FGFR1 and T47D/FGFR1 cells. The protein levels of p-ERK, p-AKT, and p-p38 in MCF-7/FGFR1 and T47D/FGFR1 cells were significantly higher than the control cells with/without Palbociclib treatment. The results also showed that ER alpha phosphorylation, estrogen response element (ERE)-luciferase activity and mRNA levels of ER target genes, including PS2, c-myc, E2F, FGF2 and FGFR2, in MCF-7/FGFR1 and T47D/FGFR1 cells were maintained at higher levels post Palbociclib, as compared to control. We then treated control and FGFR1 overexpressing MCF-7 and T47D cells with Palbociclib in combination with FGFR inhibitor, AZD 4547. The combined drug treatment resulted in remarkably synergistic effect on MCF-7/FGFR1 and T47D/FGFR1 cells, as indicated by MTT and clonogenic assays. Key markers in cell cycle regulation, RTK signaling and ER pathway were significantly inhibited in the cells treated with both Palbociclib and AZD 4547. Taken together, our results demonstrated that FGFR1 overexpression is a critical factor affecting Palbociclib sensitivity. The underlying mechanisms involve FGFR1 overexpression-associated RTK and ER signaling. Combination of Palbociclib with FGFR targeting agents may overcome FGFR-associated Palbociclib resistance. Citation Format: Yujie Shi, Zhikun Ma, Yudan Wu, Amanda B. Parris, Qiong Cheng, Lingfei Kong, Xiaohe Yang. FGFR1 overexpression ER positive breast cancer cells confer the cells resistant to Palbociclib via upregulation of RTK-ER signaling [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3009.
- Published
- 2019
- Full Text
- View/download PDF
26. Abstract 1168: Characterization of tumor initiation cells in mammary tissues of C3-1-TAg transgenic mice
- Author
-
Yujie Shi, Amanda B. Parris, Xiaohe Yang, Qiong Cheng, Zhikun Ma, and Lingfei Kong
- Subjects
Cancer Research ,education.field_of_study ,Mammary tumor ,Stromal cell ,Population ,Tumor initiation ,Biology ,medicine.disease_cause ,Stem cell marker ,medicine.disease ,Primary tumor ,Oncology ,Cancer stem cell ,medicine ,Cancer research ,Carcinogenesis ,education - Abstract
Animal models are powerful tools for the understanding of tumor development and cancer stem cell (CSC) origin. C3-1-TAg transgenic mice are a model for human triple negative breast cancer. Although general patterns of tumor development in this model have been well characterized, the tumor initiation cells of this model have not been defined. The objective of this study was to characterize the stemness and growth properties of mammary tumor cells in context with mammary epithelial cell (MEC) subpopulations of C3-1-TAg transgenic mice to identify the origin of tumor initiation cells. We first isolated MECs from mammary tissues of C3-1-TAg mice at various ages (8, 17, and 23 weeks old) and primary tumor cells from tumors of this model. Flow cytometry analysis with CD24 and CD49f as cell markers showed that cells from mammary tissues were separated into three subpopulations, including luminal, basal, and stromal populations. We found that the percentage of luminal subpopulation increased with age. Analysis of tumor derived cells using the same markers indicated that the majority of the cells in the plots overlapped with the position of luminal epithelial cells, suggesting their mutual connections. Next, we performed FACS sorting of mammary epithelial cells from the glands based on CD24 and CD49f and obtained four subpopulations of cells, including P1(stromal), P2(luminal), P3(CD24high/CD49fhigh, top of basal population on the plot), and P4(CD24mid/CD49fhigh, lower part of basal population), followed by the characterization of each subpopulations. Colony formation cell (CFC) assay and mammosphere assay were used for evaluation of luminal progenitor cells and self-renewal potential, respectively. Data from CFC assays showed that CFC colony numbers were the highest from the P2 population, followed by a few colonies from P3 population. Mammosphere assay showed that cells from P1 population formed typical mammospheres, while as cells from P2 and P3 formed cellular clusters different from typical mammospheres. FACS sorting with cells from C3-1-TAg mammary tumors obtained P2 and P3 subpopulations. Cells from P2, not P3, subpopulation were efficient in mammosphere formation. Because cells in P2 population are luminal mammary epithelial cells and are enriched with luminal progenitor cells, integration of data above suggest that C3-1-TAg mammary tumor cells are mainly derived from luminal progenitor cells. Using CD61, which is a luminal progenitor cell marker, we found that the percentage of luminal progenitor cells (CD61+/CD49fhigh) in the mammary glands was increased with age, and most tumor derived cells were CD61+/CD49fhigh. Taken together, our data demonstrate that the tumor initiation cells in the C3-1-TAg mammary tumors were derived from luminal progenitor cells. These findings lay a foundation for further characterization of cellular basis and molecular analysis of tumorigenesis in C3-1-TAg transgenic models. Citation Format: Qiong Cheng, Amanda B. Parris, Zhikun Ma, Yujie Shi, Lingfei Kong, Xiaohe Yang. Characterization of tumor initiation cells in mammary tissues of C3-1-TAg transgenic mice [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 1168.
- Published
- 2019
- Full Text
- View/download PDF
27. Buformin inhibits the stemness of erbB-2-overexpressing breast cancer cells and premalignant mammary tissues of MMTV-erbB-2 transgenic mice
- Author
-
Qingxia Zhao, Ming Zhao, Erin W. Howard, Xiaohe Yang, Zhikun Ma, and Amanda B. Parris
- Subjects
0301 basic medicine ,Cancer Research ,Cell Survival ,Receptor, ErbB-2 ,Antineoplastic Agents ,Breast Neoplasms ,Mice, Transgenic ,Mice ,03 medical and health sciences ,ErbB-2 ,Breast cancer ,0302 clinical medicine ,Cancer stem cell ,ErbB ,Cell Line, Tumor ,Animals ,Humans ,Medicine ,skin and connective tissue diseases ,Clonogenic assay ,Cell Proliferation ,Buformin ,Mammary tumor ,Cancer stem cells ,business.industry ,Cell growth ,TOR Serine-Threonine Kinases ,Research ,Drug Repositioning ,Protein-Tyrosine Kinases ,Cell cycle ,Xenograft Model Antitumor Assays ,Up-Regulation ,3. Good health ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,Neoplastic Stem Cells ,Cancer research ,Female ,Stem cell ,business ,Signal Transduction ,Mammary epithelial cells ,medicine.drug - Abstract
Background Metformin, an FDA-approved drug for the treatment of Type II diabetes, has emerged as a promising anti-cancer agent. Other biguanide analogs, including buformin and phenformin, are suggested to have similar properties. Although buformin was shown to reduce mammary tumor burden in carcinogen models, the anti-cancer effects of buformin on different breast cancer subtypes and the underlying mechanisms remain unclear. Therefore, we aimed to investigate the effects of buformin on erbB-2-overexpressing breast cancer with in vitro and in vivo models. Methods MTT, cell cycle, clonogenic/CFC, ALDEFLUOR, tumorsphere, and Western blot analyses were used to determine the effects of buformin on cell growth, stem cell populations, stem cell-like properties, and signaling pathways in SKBR3 and BT474 erbB-2-overexpressing breast cancer cell lines. A syngeneic tumor cell transplantation model inoculating MMTV-erbB-2 mice with 78617 mouse mammary tumor cells was used to study the effects of buformin (1.2 g buformin/kg chow) on tumor growth in vivo. MMTV-erbB-2 mice were also fed buformin for 10 weeks, followed by analysis of premalignant mammary tissues for changes in morphological development, mammary epithelial cell (MEC) populations, and signaling pathways. Results Buformin significantly inhibited SKBR3 and BT474 cell growth, and in vivo activity was demonstrated by considerable growth inhibition of syngeneic tumors derived from MMTV-erbB-2 mice. In particular, buformin suppressed stem cell populations and self-renewal in vitro, which was associated with inhibited receptor tyrosine kinase (RTK) and mTOR signaling. Consistent with in vitro data, buformin suppressed mammary morphogenesis and reduced cell proliferation in MMTV-erbB-2 mice. Importantly, buformin decreased MEC populations enriched with mammary reconstitution units (MRUs) and tumor-initiating cells (TICs) from MMTV-erbB-2 mice, as supported by impaired clonogenic and mammosphere formation in primary MECs. We further demonstrated that buformin-mediated in vivo inhibition of MEC stemness is associated with suppressed activation of mTOR, RTK, ER, and β-catenin signaling pathways. Conclusions Overall, our results provide evidence for buformin as an effective anti-cancer drug that selectively targets TICs, and present a novel prevention and/or treatment strategy for patients who are genetically predisposed to erbB-2-overexpressing breast cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0498-0) contains supplementary material, which is available to authorized users.
- Published
- 2017
- Full Text
- View/download PDF
28. Expression of ERO1L in gastric cancer and its association with patient prognosis
- Author
-
Shegan Gao, Zengfang Wang, Bo Zhou, Wei Zhang, Canhui Jin, Yan-Tong Yang, Xiaoshan Feng, Zhikun Ma, Gongping Wang, and Ye Chen
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Oncogene ,business.industry ,Cancer ,General Medicine ,Articles ,Cell cycle ,medicine.disease_cause ,medicine.disease ,Molecular medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immunology and Microbiology (miscellaneous) ,Apoptosis ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Carcinoma ,Immunohistochemistry ,business ,Carcinogenesis - Abstract
The present study aimed to assess the expression of endoplasmic reticulum oxidoreductin-1-like (ERO1L) in gastric cancer and determine its association with patient prognosis. A total of 105 patients with gastric cancer undergoing radical gastrectomy were selected for the current study. Gastric cancer tissues (the observation group) and normal gastric tissue adjacent to the carcinoma (the control group) were resected from patients. Levels of ERO1L mRNA and protein in tumor tissues and adjacent tissues were detected using reverse transcription-quantitative polymerase chain reaction, western blotting and immunohistochemistry. Patients were divided into two groups: A positive group and negative group, according to the expression of ERO1. The expression of ERO1L in gastric cancer and its association with patient prognosis was analyzed. Levels of ERO1 mRNA and protein in gastric cancer were significantly higher than those of adjacent tissues (P
- Published
- 2016
29. Abstract P6-07-04: Rapamycin inhibits the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice
- Author
-
Zhiying Guo, Qingxia Zhao, Ying Xing, EW Howard, AB Parris, Xiaohe Yang, Zhikun Ma, and Ming Zhao
- Subjects
Cancer Research ,Mammary tumor ,education.field_of_study ,Population ,Myoepithelial cell ,Wnt signaling pathway ,Estrogen receptor ,Biology ,medicine.disease ,Metastasis ,Malignant transformation ,Oncology ,medicine ,Cancer research ,Stem cell ,skin and connective tissue diseases ,education - Abstract
Rapamycin, a well-studied mTOR inhibitor, has been demonstrated to inhibit mammary carcinogenesis at multiple stages, including initiation, invasion, and metastasis, in preclinical animal models. Nevertheless, the cancer preventative potential and underlying mechanisms remain unclear, especially in individual breast cancer subtypes like ErbB2/Her2-positive breast cancers. ErbB2 amplification/overexpression is a particular clinical concern because it occurs in approximately one-third of human breast cancers and is associated with poor prognosis. Therefore, we used MMTV-ErbB2 transgenic mice as our model system to test the efficacy of rapamycin in the prevention of ErbB2-mediated mammary tumor development. Our initial data provided proof of concept regarding the anti-cancer effects of rapamycin in vivo. Indeed, rapamycin (1.5 mg/kg/day for 12 days) significantly reduced the volume and weight of syngeneic 78617 cell-derived mammary tumors in MMTV-ErbB2 mice, despite observed decreases in CD4+ and CD8+ immune cells. Since advanced mammary gland development can serve as an indicator of breast cancer risk, we investigated the effects of rapamycin on mammary gland development in MMTV-ErbB2 mice that were treated with low-dose rapamycin (1 mg/kg/day) between weeks 10 and 20 of age. As such, rapamycin significantly attenuated mammary morphogenesis at 20 weeks of age, as indicated by decreased branching density, ductal elongation, and proliferative index of the premalignant mammary glands. Flow cytometric analysis of isolated primary mammary epithelial cells (MECs) was performed using CD24 and CD49f markers to identify MEC populations. We found that rapamycin has a significant impact on MEC stemness based on changes in luminal (CD24highCD49flow), mammary stem cell (MaSC)-enriched (CD24highCD49fhigh), and myoepithelial/basal (CD24low/highCD49fhigh) MEC populations. We also used CD61 and CD49f markers to identify a population enriched with luminal progenitor cells (CD61highCD49fhigh) that was selectively inhibited by rapamycin. Consistent with our flow cytometric analyses, rapamycin inhibited the luminal progenitor cell-enriched population, self-renewal, and anchorage-independent cell growth of primary MECs, as demonstrated by colony-forming cell, mammosphere, and 3D culture assays, respectively. These functional stem cell assays further corroborate that rapamycin suppresses the stemness of primary MECs. Molecular analysis of MECs demonstrated that rapamycin inhibited mTOR signaling, as expected. Importantly, rapamycin also significantly suppressed the receptor tyrosine kinase/ErbB2, estrogen receptor, Wnt/β-catenin, and TGFβ/Smad3 signaling pathways prior to malignant transformation. Collectively, our study provides evidence that rapamycin has potential cancer preventative effects in the mammary glands of MMTV-ErbB2 mice during the premalignant risk window. These rapamycin-induced anti-cancer effects ultimately highlight the promising clinical significance of rapamycin for the prevention and treatment of human ErbB2-overexpressing breast cancers. Citation Format: Yang X, Zhao Q, Parris AB, Howard EW, Zhao M, Guo Z, Xing Y, Ma Z. Rapamycin inhibits the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr P6-07-04.
- Published
- 2018
- Full Text
- View/download PDF
30. Trisomy chromosome 5 is a recurrent cytogenetic lesion in mammary tumors from parous MMTV-erbB-2 transgenic mice
- Author
-
Shibo Li, Young Mi Kim, Seungjik Lee, Zhikun Ma, Xiaohe Yang, and Jiyun Lee
- Subjects
Genetically modified mouse ,Cancer Research ,Mammary tumor ,Pathology ,medicine.medical_specialty ,animal structures ,Cancer ,Articles ,Biology ,medicine.disease ,Oncology ,Mammary tumor virus ,ErbB ,medicine ,Immunohistochemistry ,Trisomy ,X chromosome - Abstract
erbB-2 is amplified or overexpressed in approximately 30% of human breast cancers, and has been associated with poor prognosis and therapeutic resistance. Previous studies have suggested that erbB-2 overexpression in transgenic mice induces genomic instability; however, the patterns of genetic lesions vary with individual model systems. The development of mammary tumors in multiparous murine mammary tumor virus (MMTV)-erbB-2 transgenic mice is accelerated due to hormonal interactions which induce the overexpression of MMTV-mediated erbB-2. However, whether or not accelerated tumor development is associated with modified cytogenetic patterns remains to be determined. In this study, chromosomal changes were characterized in mammary tumor cells derived from multiparous MMTV-erbB-2 transgenic mice, and compared with tumor cells derived from control virgin mice. Immunohistochemistry and Western blotting were used to detect erbB-2 overexpression in mammary tissues. Each of the five tumors from the multiparous MMTV-erbB-2 transgenic mice was found to exhibit a marked chromosomal imbalance, compared with only one tumor with aberrant chromosomes among the five tumors from the control virgin mice. In particular, trisomy 5 and loss of the X chromosome were recurrent cytogenetic lesions in tumors from the parous mice, which is a novel pattern compared with previous studies. The elevated number of genetic lesions in tumors from parous mice, which were characterized by enhanced erbB-2 overexpression and increased receptor tyrosine kinase activation in the mammary glands, suggest a causal role for erbB-2 in the genomic instability present in these tumors. These data advance our understanding of erbB-2-mediated pathogenesis and underscore the role of cytogenetic alteration in this process.
- Published
- 2011
- Full Text
- View/download PDF
31. Abstract 903: Upregulation of DARPP32/PPP1R1B is a critical mediator of acquired lapatinib resistance in ErbB2-overexpressing breast cancer cells
- Author
-
Amanda B. Parris, Erin W. Howard, Zhikun Ma, and Xiaohe Yang
- Subjects
Cancer Research ,Gene knockdown ,biology ,Cell growth ,Cell cycle ,CREB ,Lapatinib ,Small hairpin RNA ,Oncology ,Downregulation and upregulation ,Cell culture ,biology.protein ,Cancer research ,medicine ,skin and connective tissue diseases ,medicine.drug - Abstract
ErbB2/Her2 overexpression is detected in approximately 30% of human breast cancers and is associated with poor prognosis. Lapatinib, a small molecule dual inhibitor targeting ErbB2 and EGFR, is FDA-approved for the treatment of ErbB2-positive (ErbB2+) advanced or metastatic breast cancer. However, lapatinib resistance (LR) is emerging as a critical issue in clinical oncology. To understand the molecular mechanisms of LR, we first developed a lapatinib-resistant cell line from ErbB2-overexpressing BT474 breast cancer cells by prolonged exposure of parental BT474 cells to gradually increasing concentrations of lapatinib (up to 8 µM). We performed microarray analyses on parental and LR cells to identify novel factors/pathways that contribute to LR. Our microarray data indicated that the gene expression profile of the BT474/LR cells is significantly different from the parental BT474 cells. We found that DARPP32/PPP1R1B is one of the most significantly upregulated genes in the BT474/LR cells. To investigate the role of DARPP32 in LR development, we examined the effect of DARPP32 knockdown via lentiviral shRNA on BT474/LR cell proliferation and apoptosis. Results from MTT, clonogenic, cell cycle, and cleaved caspase-3 and PARP analyses indicated that DARPP32 knockdown significantly sensitized BT474/LR cells to lapatinib. We also found that DARPP32 knockdown renders MDA-MB-361 breast cancer cells, which express high endogenous levels of DARPP32, more sensitive to lapatinib. These data demonstrate that DARPP32 overexpression contributes to LR. To understand the mechanism of DARPP32 overexpression-mediated LR, we focused on the regulation of transcription factor cAMP response element-binding protein (CREB) in this process. As such, DARPP32 overexpression was generally associated with CREB upregulation. We also found that CREB-mediated transcription was increased in BT474/LR cells, and DARPP32 knockdown in BT474/LR and MDA-MB-361 cells significantly downregulated CREB protein levels and CREB-mediated transcription, as indicated by luciferase reporter assays. CREB overexpression rendered BT474 cells resistant to lapatinib and reversed DARPP32 knockdown-induced sensitization in BT474/LR cells. In contrast, CREB knockdown sensitized the cells to lapatinib. Further analysis with MG132 blockage suggests that DARPP32 may regulate CREB protein levels through the proteasomal degradation pathway. Taken together, we have identified DARPP32 overexpression as a critical mediator of acquired lapatinib resistance. The underlying mechanisms involve the deregulation of CREB protein and activity levels. These novel findings advance our understanding of the molecular mechanisms of LR and provide fundamental support for testing the DARPP32-CREB axis in clinical settings for the management of LR breast cancers, which is of significant translational value. Citation Format: Zhikun Ma, Erin W. Howard, Amanda B. Parris, Xiaohe Yang. Upregulation of DARPP32/PPP1R1B is a critical mediator of acquired lapatinib resistance in ErbB2-overexpressing breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 903.
- Published
- 2018
- Full Text
- View/download PDF
32. Gastric cardia adenocarcinoma microRNA profiling in Chinese patients
- Author
-
Shuo Liang, Feng Su, Xiaojuan Zhu, Jiangman Zhao, Zhikun Ma, Shegan Gao, Mengxi Zhang, Chen Zhao, Xiaoshan Feng, Gang Liu, Pengfei Zhang, Ruinuo Jia, Fuyou Zhou, Lu Wang, and Bo Peng
- Subjects
0301 basic medicine ,Male ,Microarray ,Biology ,Adenocarcinoma ,Bioinformatics ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,microRNA ,medicine ,Biomarkers, Tumor ,Humans ,RNA, Messenger ,skin and connective tissue diseases ,Survival analysis ,Neoplasm Staging ,Oligonucleotide Array Sequence Analysis ,Microarray analysis techniques ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Cancer ,General Medicine ,Middle Aged ,Gastric Cardia Adenocarcinoma ,medicine.disease ,Prognosis ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Lymphatic Metastasis ,Cancer research ,Female ,Follow-Up Studies - Abstract
Gastric cardia adenocarcinoma (GCA), which occurs at the gastroesophageal boundary, is one of the most malignant types of cancer. Over the past 30 years, the incidence of GCA has increased by approximately sevenfold, which has a more substantial increase than that of many other malignancies. However, as previous studies mainly focus on non-cardia gastric cancer, until now, the mechanisms behind GCA remain largely unknown. MicroRNAs (miRNAs) have been shown to play pivotal roles in carcinogenesis. To gain insight into the molecular mechanisms regulated by miRNAs in GCA development, we investigated miRNA expression profiles using 81 pairs of primary GCAs and corresponding non-tumorigenic tissues. First, 21 pairs of samples were used for microarray analysis, and then another 60 pairs of samples were used for further analysis. Our results showed that 464 miRNAs (237 upregulated, 227 downregulated, false discovery rate FDR
- Published
- 2015
33. The inhibition of histone deacetylase 8 suppresses proliferation and inhibits apoptosis in gastric adenocarcinoma
- Author
-
Shuo Liang, Zhikun Ma, Shiyuan Song, Guangping Zhang, Ruina Yang, Po Xu, and Ying Wang
- Subjects
Adult ,Male ,Cancer Research ,Cell cycle checkpoint ,Carcinogenesis ,Cell ,Apoptosis ,Biology ,Adenocarcinoma ,medicine.disease_cause ,Histone Deacetylases ,Stomach Neoplasms ,Cell Line, Tumor ,medicine ,Humans ,RNA, Messenger ,Aged ,Cell Proliferation ,Cell growth ,G1 Phase ,HDAC8 ,Cell cycle ,Middle Aged ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,medicine.anatomical_structure ,Oncology ,Cancer cell ,Cancer research ,Female - Abstract
Histone deacetylase 8 (HDAC8), a unique member of class I HDACs, shows remarkable correlation with advanced disease stage. The depletion of HDAC8 leads to inhibition of proliferation, apoptosis and cell cycle arrest in multiple malignant tumors. However, little is known about the contribution of HDAC8 to the tumorigenesis of gastric cancer (GC). The present study investigated expression of HDAC8 in GC cell lines and tissues, and the roles of HDAC8 inhibition in the proliferation, cell cycle and apoptosis of gastric cancer cells and explored the potential mechanisms. In the present study, quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry were used to examine the mRNA and protein expression of HDAC8 in GC cell lines and tissues. Then, the correlation between the clinicopathological parameters and the expression of HDAC8 was assessed. Finally, siRNA transfection and HDAC8 plasmid was performed to explore the functions of HDAC8 in GC progression in vitro. We found that the expression of HDAC8 was significantly upregulated both in GC cell lines and tumor tissues compared to human normal gastric epithelial cell, GES-1 and matched non-tumor tissues. Furthermore, depletion of HDAC8 remarkably inhibited GC cell proliferation, increased the apoptosis rate and G0/G1 phase percentage in vitro. Western blotting showed that the expression of protein promoting apoptosis such as, Bmf, activated caspase-3, caspase-6 were elevated following HDAC8 depletion. Our data exhibited an important role of HDAC8 in promoting gastric cancer tumorigenesis and identify this HDAC8 as a potential therapeutic target for the treatment of gastric cancer.
- Published
- 2015
34. In Utero Exposure to Low-Dose Alcohol Induces Reprogramming of Mammary Development and Tumor Risk in MMTV-erbB-2 Transgenic Mice
- Author
-
Zhikun Ma, Ming Zhao, Harry Lee, Xiaohe Yang, and Amanda J. Blackwelder
- Subjects
Receptor, ErbB-2 ,Estrogen receptor ,lcsh:Chemistry ,Mice ,breast cancer risk ,0302 clinical medicine ,Pregnancy ,lcsh:QH301-705.5 ,Spectroscopy ,0303 health sciences ,alcohol ,in utero exposure ,General Medicine ,Up-Regulation ,Computer Science Applications ,In utero ,Prenatal Exposure Delayed Effects ,030220 oncology & carcinogenesis ,Female ,Signal Transduction ,estrogen receptor ,medicine.medical_specialty ,Offspring ,Mice, Transgenic ,Biology ,Article ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Fetus ,Mammary Glands, Animal ,Mammary tumor virus ,ErbB ,Internal medicine ,medicine ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,Protein kinase B ,030304 developmental biology ,erbB-2 ,Ethanol ,Organic Chemistry ,Estrogen Receptor alpha ,Mammary Neoplasms, Experimental ,reprogramming ,Cell Transformation, Viral ,Endocrinology ,Mammary Tumor Virus, Mouse ,lcsh:Biology (General) ,lcsh:QD1-999 ,Estrogen receptor alpha - Abstract
There is increasing evidence that prenatal exposure to environmental factors may modify breast cancer risk later in life. This study aimed to investigate the effects of in utero exposure to low-dose alcohol on mammary development and tumor risk. Pregnant MMTV-erbB-2 mice were exposed to alcohol (6 g/kg/day) between day 13 and day 19 of gestation, and the female offspring were examined for tumor risk. Whole mount analysis indicated that in utero exposure to low-dose alcohol induced significant increases in ductal extension at 10 weeks of age. Molecular analysis showed that in utero alcohol exposure induced upregulation of ERα signaling and activation of Akt and Erk1/2 in pubertal mammary glands. However, enhanced signaling in the EGFR/erbB-2 pathway appeared to be more prominent in 10-week-old glands than did signaling in the other pathways. Interestingly, tumor development in mice with in utero exposure to low-dose alcohol was slightly delayed compared to control mice, but tumor multiplicity was increased. The results indicate that in utero exposure to low-dose alcohol induces the reprogramming of mammary development by mechanisms that include altered signaling in the estrogen receptor (ER) and erbB-2 pathways. The intriguing tumor development pattern might be related to alcohol dose and exposure conditions, and warrants further investigation.
- Published
- 2015
- Full Text
- View/download PDF
35. Abstract 1908: FGFR inhibitor, AZD4547, impedes the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice
- Author
-
Qingxia Zhao, Erin W. Howard, Ming Zhao, Ying Xing, Xiaohe Yang, Amanda B. Parris, and Zhikun Ma
- Subjects
Cancer Research ,Mammary tumor ,Cell ,Myoepithelial cell ,Wnt signaling pathway ,Cancer ,FGFR Inhibitor AZD4547 ,Biology ,medicine.disease ,Malignant transformation ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Stem cell - Abstract
The fibroblast growth factor receptor (FGFR) family (FGFR1-4) of receptor tyrosine kinases (RTKs) regulates signaling pathways involved in cell proliferation and differentiation. In particular, FGFR1 and FGFR2, which are found in the terminal end buds of developing mammary ducts, play a role in mammary development and glandular morphogenesis involving the regulation of mammary stem cells (MaSCs) in mice. As such, a number of FGFR inhibitors are being tested in preclinical studies and clinical trials for anti-tumor properties. Nevertheless, reports on FGFR inhibitor-mediated breast cancer prevention are sparse. In this study, we aimed to investigate the anti-cancer benefits of AZD4547, a small molecule inhibitor of FGFR1-3, on ErbB2-overexpressing breast cancer models. We particularly focus on the effects of AZD4547 on MaSCs and tumor-initiating cells (TICs) in the premalignant tissues of MMTV-ErbB2 transgenic mice. We first demonstrated the anti-proliferative effects of AZD4547 (1-5 µM) on human ErbB2-overexpressing breast cancer cell lines. We further showed that AZD4547 confers potent inhibition of the stemness of these breast cancer cells, as indicated by significant depletion of ALDH+ cells and impaired tumorsphere formation. To study the in vivo effects of AZD4547 on the stemness of mammary epithelial cells (MECs), MMTV-ErbB2 transgenic mice were administered AZD4547 (2-6 mg/kg/day) for 10 weeks (weeks 8-18 of age) during the ‘risk window’ for mammary tumor development. Histopathological analysis indicated that AZD4547 significantly inhibited ductal branching and MEC proliferation. To examine the effect of AZD4547 on MEC subpopulations and tissue hierarchy dynamics in the premalignant mammary tissues of this model, we performed flow cytometry analyses on the primary MECs using CD24/CD49f and CD61/CD49f cell surface markers. The results showed that AZD4547 treatment substantially reduced MaSC-derived luminal and myoepithelial cell populations. AZD4547 also selectively suppressed the CD61highCD49fhigh cell population, which is enriched with luminal progenitor cells that give rise to TICs during MMTV-ErbB2 mammary tumor development. Mammosphere and colony-forming cell (CFC) assays on primary MECs demonstrated that the stemness of these cells was also blocked by AZD4547 prior to malignant transformation. Consistently, AZD4547 inhibited the anchorage-independent growth of cells from spontaneous tumors. Moreover, we demonstrated that AZD4547 downregulated multiple pathways, including the inactivation of FGFR, EGFR, and Wnt/β-catenin signaling. Collectively, the morphogenic, MaSC/TIC, and signaling regulation associated with AZD4547 treatment provides critical evidence for AZD4547 as a breast cancer preventative and therapeutic agent, which ultimately reveals clues for more effective eradication of refractory mammary tumors. Citation Format: Qingxia Zhao, Amanda B. Parris, Erin W. Howard, Ming Zhao, Ying Xing, Zhikun Ma, Xiaohe Yang. FGFR inhibitor, AZD4547, impedes the stemness of mammary epithelial cells in the premalignant tissues of MMTV-ErbB2 transgenic mice [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1908. doi:10.1158/1538-7445.AM2017-1908
- Published
- 2017
- Full Text
- View/download PDF
36. MicroRNA-645, up-regulated in human adencarcinoma of gastric esophageal junction, inhibits apoptosis by targeting tumor suppressor IFIT2
- Author
-
Daiming Fan, Gang Liu, Shuoguo Li, Ying Wang, Shiyuan Song, Mengxi Zhang, Ruina Yang, Zhikun Ma, Shegan Gao, Xiaoshan Feng, and Shuo Liang
- Subjects
Adult ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,Esophageal Neoplasms ,Cell ,Adencarcinoma of gastric esophageal junction ,Apoptosis ,Adenocarcinoma ,medicine.disease_cause ,IFIT2 ,Stomach Neoplasms ,Cell Line, Tumor ,microRNA ,Genetics ,medicine ,Humans ,Aged ,Aged, 80 and over ,Binding Sites ,Oncogene ,business.industry ,Cancer ,Proteins ,RNA-Binding Proteins ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,Oncology ,Cancer research ,Ectopic expression ,Female ,Signal transduction ,Carcinogenesis ,business ,Apoptosis Regulatory Proteins ,microRNA-645 ,Signal Transduction ,Research Article - Abstract
Background An increasing body of evidence indicates that miRNAs have a critical role in carcinogenesis and cancer progression; however, the role of miRNAs in the tumorigenesis of adencarcinoma of gastric esophageal junction (AGEJ) remains largely unclear. Methods The SGC7901 and BGC-823 gastric cancer cell lines were used. The expressions of miR-645 and IFIT2 (Interferon-induced protein with tetratricopeptide repeats 2) were examined by qRT-PCR, The expressions of IFIT2 was examined by western blotting and immunohistochemistry assay. The cell apoptosis was determined by FACS. MiR-645 inhibitor, mimics and plasmid-IFIT2 transfections were performed to study the loss- and gain-function. Caspase-3/7 activity was examined by caspase-3/7 assay. Results In the present study, we have reported an increased expression of miR-645 in AGEJ clinical specimens compared with paired non-cancerous tissues. We also observed a significant miR-645 up-regulation in two gastric cancer (GC) cell lines, SGC7901 and BGC-823, which were used as cell models because there was no available AGEJ cell lines established to date. We found that inhibition of miR-645 could sensitize dramatically SGC7901 and BGC-823 cells to both serum starvation– and chemotherapeutic drug–induced apoptosis by up-regulating IFIT2, a mediator of apoptosis via a mitochondrial pathway, with a potential binding site for miR-645 in its mRNA’s 3′UTR. Further investigation exhibited that IFIT2 expression decreases in SGC7901 and BGC-823 cells and AGEJ tissues. IFIT2 ectopic expression leads to promotion of cell apoptosis, indicating that IFIT2 may function as a suppressor in the development of AGEJ. Furthermore, inhibition of miR-645 induces up-regulation of IFIT2 and increased caspase-3/7 activity compared with control groups. Conclusions Our data suggest that miR-645 functions as an oncogene in human AGEJ by, at least partially through, targeting IFIT2. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-633) contains supplementary material, which is available to authorized users.
- Published
- 2013
37. Sago-type palms were an important plant food prior to rice in southern subtropical China
- Author
-
Mingqi Li, Zhiwei Wan, Huw Barton, Jun Wei, Dan Zhang, Xiaoyan Yang, Zhikun Ma, and Quan Li
- Subjects
China ,lcsh:Medicine ,Plant Science ,Arecaceae ,Social and Behavioral Sciences ,Paleoanthropology ,Botany ,Humans ,Dominance (ecology) ,Paleobotany ,Domestication ,lcsh:Science ,Biology ,History, Ancient ,Nutrition ,Evolutionary Biology ,Multidisciplinary ,biology ,business.industry ,lcsh:R ,Paleontology ,food and beverages ,Starch ,biology.organism_classification ,Starch analysis ,Diet ,Archaeology ,Agronomy ,Agriculture ,Phytolith ,Anthropology ,Earth Sciences ,Medicine ,lcsh:Q ,Fern ,business ,Research Article - Abstract
Poor preservation of plant macroremains in the acid soils of southern subtropical China has hampered understanding of prehistoric diets in the region and of the spread of domesticated rice southwards from the Yangtze River region. According to records in ancient books and archaeological discoveries from historical sites, it is presumed that roots and tubers were the staple plant foods in this region before rice agriculture was widely practiced. But no direct evidences provided to test the hypothesis. Here we present evidence from starch and phytolith analyses of samples obtained during systematic excavations at the site of Xincun on the southern coast of China, demonstrating that during 3,350-2,470 aBC humans exploited sago palms, bananas, freshwater roots and tubers, fern roots, acorns, Job's-tears as well as wild rice. A dominance of starches and phytoliths from palms suggest that the sago-type palms were an important plant food prior to the rice in south subtropical China. We also believe that because of their reliance on a wide range of starch-rich plant foods, the transition towards labour intensive rice agriculture was a slow process.
- Published
- 2013
38. Abstract B31: In utero exposure to bisphenol A induces reprogramming of mammary development and tumor risk in MMTV-erbB-2 transgenic mice
- Author
-
Zhikun Ma, Amanda B. Parris, and Xiaohe Yang
- Subjects
Genetically modified mouse ,Cancer Research ,medicine.medical_specialty ,Mammary tumor ,Estrogen receptor ,Environmental exposure ,Biology ,medicine.disease ,Cyclin D1 ,Breast cancer ,Endocrinology ,Oncology ,In utero ,ErbB ,Internal medicine ,medicine ,Molecular Biology - Abstract
Increasing evidence indicates that environmental exposure plays a critical role in breast cancer etiology. Bisphenol A (BPA), the building block of polycarbonate plastics with estrogenic activity, is one of the most common chemicals exposed in daily life. Recent studies suggest a link between BPA exposure and increased breast cancer risk. However, specific conditions and the underlying mechanisms of BPA exposure associated breast cancer risk remain unclear. This study aimed to investigate the effects of in utero exposure to BPA on mammary tumor development in MMTV-erbB-2 transgenic mice, which exemplifies a scenario of gene-environmental interaction. Since erbB-2 is amplified/overexpressed in approximately 30% of breast cancer, which has been associated with poor prognosis and therapeutic resistance, use of this clinically relevant mammary tumor model is of high translational value. In this study, pregnant MMTV-erbB-2 mice were subcutaneously injected with 0, 50 ng, 500 ng and 250 μg/Kg body weight BPA daily between day 13 and day 19 of gestation. We found that in utero exposed mice in the low dose (50 and 500 ng) groups, but not the high dose group (250 μg), displayed earlier vaginal opening and prolonged estrous phase, suggesting a dose dependent pro-estrogenic effect of in utero exposure to BPA. Whole mount analysis showed that mammary glands of mice with in utero exposure to low dose BPA had a longer ductal extension at 6 weeks and increased lateral branching/alveolar structures at 10 weeks, which was more significant in the 500 ng group but less evident in the high dose group. Molecular analysis of mammary tissues at 10 weeks indicated that in utero exposure to BPA increased phosphorylation/activation of erbB-2, EGFR, erbB-3, Erk1/2 and Akt, and expression of erbB-3 and EGFR. Signaling of estrogen receptor (ER) pathway was upregulated concomitantly, as reflected by increased expression and phosphorylation of ERα and upregulation of cyclin D1 and c-myc. More importantly, mice in the 500 ng group but not in the high dose group, developed tumor earlier than the mice in the control group, suggesting that dose is a critical factor affecting the outcomes. These data demonstrated that in utero exposure to BPA may promote mammary tumor development in erbB-2 transgenic mice in a dose dependent manner. Concomitant activation of the erbB-2 and ER pathways in the premalignant tissues suggests that induction of ER-erbB-2 crosstalk may play a critical role in mammary tumorigenesis promoted by in utero exposure to BPA, which underscores the significance of gene-environment interaction in early exposure to BPA associated breast cancer risk. Citation Format: Zhikun Ma, Amanda Parris, Xiaohe Yang. In utero exposure to bisphenol A induces reprogramming of mammary development and tumor risk in MMTV-erbB-2 transgenic mice. [abstract]. In: Proceedings of the AACR Special Conference: Developmental Biology and Cancer; Nov 30-Dec 3, 2015; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(4_Suppl):Abstract nr B31.
- Published
- 2016
- Full Text
- View/download PDF
39. Chk1 knockdown confers radiosensitization in prostate cancer stem cells
- Author
-
Xiaoshan Feng, Zhongling Dou, Hui Liu, Zhikun Ma, Xiaobin Wang, Zheng Xiao, and Haijun Shi
- Subjects
Male ,Cancer Research ,DNA Repair ,Mitosis ,Apoptosis ,Biology ,Cyclin B ,medicine.disease_cause ,Radiation Tolerance ,Prostate cancer ,Cancer stem cell ,Antigens, CD ,Radioresistance ,Cell Line, Tumor ,CDC2 Protein Kinase ,medicine ,Humans ,cdc25 Phosphatases ,Radiosensitivity ,AC133 Antigen ,RNA, Small Interfering ,Glycoproteins ,CD44 ,Caspase 2 ,Cancer ,Prostatic Neoplasms ,General Medicine ,Cell Cycle Checkpoints ,medicine.disease ,Cyclin-Dependent Kinases ,Hyaluronan Receptors ,Oncology ,embryonic structures ,Checkpoint Kinase 1 ,biology.protein ,Cancer research ,Neoplastic Stem Cells ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Carcinogenesis ,Peptides ,Protein Kinases - Abstract
Radioresistance is responsible for treatment failure after radiotherapy in localized prostate cancer, while prostate cancer stem cells promote radioresistance by preferential activation of the DNA damage response. Chk1 inhibition has been shown to sensitize many tumor cells to radiation. However, whether Chk1 inhibition can potentiate the cytotoxic effects of radiation on prostate cancer stem cells remains to be elucidated. In this study, CD133+CD44+ cells were isolated using microbeads and were found to possess cancer stem cell properties. Using shRNA, Chk1 was knocked down in the sorted CD133+CD44+ cells. Our results demonstrated that Chk1 knockdown abrogated the radiation-induced G2/M arrest, inhibited DNA damage repair and promoted premature mitosis, leading to increased apoptosis in the radiated sorted CD133+CD44+ cells. Moreover, these effects were accompanied by caspase-2 activation and the inactivation of phosphorylated Cdc25C and Cdc2. Our results suggest that Chk1 knockdown increases the radiosensitivity of CD133+CD44+ prostate cancer stem cells. Chk1 knockdown in prostate cancer stem cells may be an effective therapeutic strategy against prostate cancer.
- Published
- 2012
40. The Chk1 inhibitor AZD7762 sensitises p53 mutant breast cancer cells to radiation in vitro and in vivo
- Author
-
Guoliang Yao, Shegan Gao, Xiaoshan Feng, Yonggang Fan, Zhikun Ma, and Bo Zhou
- Subjects
Cancer Research ,Radiation-Sensitizing Agents ,DNA damage ,medicine.medical_treatment ,Transplantation, Heterologous ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Thiophenes ,Biology ,medicine.disease_cause ,Biochemistry ,Mice ,Genetics ,medicine ,Animals ,Humans ,Urea ,CHEK1 ,Radiosensitivity ,Molecular Biology ,Mitotic catastrophe ,Protein Kinase Inhibitors ,Cancer ,Cell cycle ,medicine.disease ,Radiation therapy ,G2 Phase Cell Cycle Checkpoints ,Oncology ,Gamma Rays ,Checkpoint Kinase 1 ,Mutation ,Cancer research ,MCF-7 Cells ,Molecular Medicine ,M Phase Cell Cycle Checkpoints ,Female ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Protein Kinases ,DNA Damage - Abstract
AZD7762, a novel checkpoint kinase 1 (Chk 1)inhibitor, has been proven to sensitize various tumor cells to DNA damage. However, whether or not AZD7762 sensitizes breast cancer cells to radiation has not been defined. In the present study, we aimed to demonstrate for the first time, that AZD7762 not only promotes radiation-induced apoptosis and mitotic catastrophe of p53 mutant T47D breast cancer cells in vitro, but also delays their xenograft growth in response to radiation in vivo. Our mechanistic study showed that AZD7762 treatment resulted in the abrogation of radiation-induced G2/M arrest and the inhibition of radiation damage repair as demonstrated by increased radiation-induced γH2AX expression and decreased RAD51 protein expression. These results suggest that AZD7762 may effectively abrogate radiation-induced G2/M arrest and inhibit radiation damage repair in conferring radiosensitivity on p53 mutant T47D breast cancer cells, by promoting radiation-induced apoptosis and mitotic catastrophe. The clinical application of AZD7762, as an adjuvant in the radiotherapy of breast cancers, should be further explored.
- Published
- 2012
41. p53 inactivation upregulates p73 expression through E2F-1 mediated transcription
- Author
-
Zhikun Ma, Chaitali Tophkhane, Yunbo Jiang, Shrikant Anant, Wanguo Liu, Dharmalingam Subramaniam, Shihe Yang, Susan M. Edgerton, Xiaohe Yang, Ann D. Thor, Shingo Yogosawa, and Toshiyuki Sakai
- Subjects
Transcription, Genetic ,Gene Expression ,lcsh:Medicine ,Apoptosis ,Transcription (biology) ,Molecular Cell Biology ,Pathology ,Transcriptional regulation ,Tumor Protein p73 ,RNA, Small Interfering ,Promoter Regions, Genetic ,skin and connective tissue diseases ,lcsh:Science ,Regulation of gene expression ,Gene knockdown ,Multidisciplinary ,Cell Death ,Nuclear Proteins ,Up-Regulation ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Oncology ,MCF-7 Cells ,Medicine ,Research Article ,Plasmids ,Cyclin-Dependent Kinase Inhibitor p21 ,Transcriptional Activation ,Chromatin Immunoprecipitation ,Biology ,Cell Growth ,Molecular Genetics ,Genetic Mutation ,Diagnostic Medicine ,Cell Line, Tumor ,Genetics ,Cancer Genetics ,Cancer Detection and Diagnosis ,Humans ,Gene Regulation ,E2F ,neoplasms ,Tumor Suppressor Proteins ,lcsh:R ,Molecular biology ,Mutation ,Cancer cell ,lcsh:Q ,Tumor Suppressor Protein p53 ,Chromatin immunoprecipitation ,Biomarkers ,E2F1 Transcription Factor ,General Pathology - Abstract
While p73 overexpression has been associated with increased apoptosis in cancer tissues, p73 overexpressing tumors appear to be of high grade malignancy. Why this putative tumor suppressor is overexpressed in cancer cells and what the function of overexpressed p73 is in breast cancers are critical questions to be addressed. By investigating the effect of p53 inactivation on p73 expression, we found that both protein and mRNA levels of TAp73 were increased in MCF-7/p53siRNA cells, MCF-7/p53mt135 cells and HCT-116/p53-/- cells, as compared to wild type control, suggesting that p53 inactivation by various forms upregulates p73. We showed that p53 knockdown induced p73 was mainly regulated at the transcriptional level. However, although p53 has a putative binding site in the TAp73 promoter, deletion of this binding site did not affect p53 knockdown mediated activation of TAp73 promoter. Chromatin immuno-precipitation (ChIP) data demonstrated that loss of p53 results in enhanced occupancy of E2F-1 in the TAp73 promoter. The responsive sequence of p53 inactivation mediated p73 upregulation was mapped to the proximal promoter region of the TAp73 gene. To test the role of E2F-1 in p53 inactivation mediated regulation of p73 transcription, we found that p53 knockdown enhanced E2F-1 dependent p73 transcription, and mutations in E2F-1 binding sites in the TAp73 promoter abrogated p53 knockdown mediated activation of TAp73 promoter. Moreover, we demonstrated that p21 is a mediator of p53-E2F crosstalk in the regulation of p73 transcription. We concluded that p53 knockdown/inactivation may upregulate TAp73 expression through E2F-1 mediated transcriptional regulation. p53 inactivation mediated upregulation of p73 suggests an intrinsic rescuing mechanism in response to p53 mutation/inactivation. These findings support further analysis of the correlation between p53 status and p73 expression and its prognostic/predictive significance in human cancers.
- Published
- 2012
42. GASC1 expression to predict response to paclitaxel plus cisplatin in oesophageal squamous cell carcinoma
- Author
-
Na Li, Zhikun Ma, Shegan Gao, Shuo Liang, Xiaoshan Feng, Pengfei Zhang, Gang Liu, Xiang Yuan, Jinyu Kong, Ruinuo Jia, Yu-feng Wang, and Mengxi Zhang
- Subjects
Oncology ,Cisplatin ,Cancer Research ,medicine.medical_specialty ,Oncogene ,business.industry ,chemistry.chemical_compound ,Paclitaxel ,chemistry ,Internal medicine ,medicine ,Basal cell ,business ,medicine.drug - Abstract
e15065 Background: GASC1 is a novel driver oncogene involved in the development of multiple cancers. Our preliminary study reported GASC1 contributed to the occurrence and progression of esophageal...
- Published
- 2015
- Full Text
- View/download PDF
43. Abstract A09: In utero exposure to estrogen promotes mammary tumor risk in MMTV-erbB-2 transgenic mice through induction of ER-erbB-2 crosstalk
- Author
-
Zhikun Ma, Amanda Blackwelder, and Xiaohe Yang
- Subjects
Cancer Research ,Mammary tumor ,Oncogene ,medicine.drug_class ,Mammary gland ,Estrogen receptor ,Biology ,medicine.disease ,Breast cancer ,medicine.anatomical_structure ,Oncology ,Estrogen ,ErbB ,In utero ,Cancer research ,medicine ,Molecular Biology - Abstract
The mammary gland during in utero stage is in a largely undifferentiated state, which is particularly vulnerable to environmental factors. Increasing evidence indicates that intrauterine hormonal and nutritional alterations may significantly modify breast cancer risk later in life. A well-known case is the “DES Daughter” story. However, the underlying mechanisms are largely unknown and more prenatal risk factors remain to be identified. Therefore, development of sensitive and reliable model systems to identify those factors with mechanistic insight is of great significance in breast cancer prevention. Previously, most studies on in utero exposure-modulated breast cancer risk used carcinogenic models. While this system is valuable in providing proof of concept, due to challenges in elucidating molecular pathways induced by in utero exposures and by carcinogens, molecular mechanistic studies in these models are compromised. In this study, we aimed to establish an animal model for studying in utero exposure to environmental hormones-induced mammary tumor risk using MMTV-erbB-2 transgenic mice. erbB-2 is an oncogene that is amplified in ~25-30% of human breast cancer. The MMTV-erbB-2 mice have a defined genetic background and tumor development patterns, which is of advantage in mechanistic studies. When pregnant MMTV-erbB-2 mice were injected with 20 ng/mouse/day of Estradiol (E2) between day 13 and day 19 of gestation, mammary tumor development of the offspring was significantly accelerated, as compared to control. The average tumor latencies of the control and the E2 group were 36 and 32 weeks respectively. In utero exposure to E2 also induced earlier vaginal opening and prolonged length of estrous stages. Whole mount analysis indicated that in utero exposure to E2 increased mammary epithelial density at 6 and 10 weeks of age. Importantly, we also found that in utero exposure to E2 induced a concurrent upregulation of signaling in estrogen receptor α (ER α) and receptor tyrosine kinase pathways in the mammary tissues, as indicated by increased expression of ER target genes (c-myc, cyclin D1, EGFR, and Bcl-2) and phosphorylation of EGFR, erbB-2, Akt1 and Erk1/2. Use of gefitinib also, a small molecular inhibitor of EGFR, abrogated the upregulation of both pathways in mammary tissues with in utero exposure to E2, suggesting a role of EGFR in in utero exposure to E2 induced crosstalk between the two pathways. Our data indicates that in utero exposure to E2 induces a systematic pro-estrogenic effect and enhanced signaling and crosstalk between ER and EGFR/erbB-2 pathways. Deregulation of the two critical pathways in the premalignant tissues is associated with mammary tumor risk later in life. This will lead to further mechanistic studies, such as in utero exposure to hormones, induced epigenetic regulation, and reprogramming. This model system may represent a model for studying gene-environment interaction in breast carcinogenesis and can be used to study the effects of in utero exposure to other environmental and dietary factors modulated breast cancer risk. Citation Format: Zhikun Ma, Amanda Blackwelder, Xiaohe Yang. In utero exposure to estrogen promotes mammary tumor risk in MMTV-erbB-2 transgenic mice through induction of ER-erbB-2 crosstalk. [abstract]. In: Proceedings of the AACR Special Conference: The Translational Impact of Model Organisms in Cancer; Nov 5-8, 2013; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(11 Suppl):Abstract nr A09.
- Published
- 2014
- Full Text
- View/download PDF
44. Abstract 5345: DMBA promotes erbB-2 mediated carcinogenesis through activation of estrogen receptor and receptor tyrosine kinase pathways
- Author
-
Zhikun Ma, Morgan Carrington, Stanley D. Kosanke, and Xiaohe Yang
- Subjects
Cancer Research ,medicine.medical_specialty ,biology ,DMBA ,Estrogen receptor ,medicine.disease_cause ,Receptor tyrosine kinase ,Cyclin D1 ,Endocrinology ,Oncology ,ErbB ,Internal medicine ,medicine ,biology.protein ,Tumor promotion ,skin and connective tissue diseases ,Carcinogenesis ,neoplasms ,Carcinogen - Abstract
ErbB-2 is a receptor tyrosine kinase (RTK) that is amplified/overexpressed in 20-30% of all invasive breast malignancies; it has been associated with poor prognosis and therapeutic resistance in breast cancer patients. 7,12-dimethylbenz anthracene (DMBA) is a polycyclic aromatic hydrocarbon known for its carcinogenic effects. The objective of this study was to investigate the effect of DMBA exposure on erbB-2 mediated carcinogenesis. This represents a combination of genetic predisposition and environmental assault that synergistically promote carcinogenesis. In this study, female MMTV-erbB-2 transgenic mice at 6-weeks of age were treated weekly by oral gavage with either 1.0 mg DMBA in peanut oil or vehicle alone for 6 weeks. In the control group, palpable tumors appeared at week 25, with an average latency of 36.0 weeks. All mice in the DMBA group developed mammary tumors between week 16 and 26 with an average latency of 20 weeks. By the endpoint (first tumor ∼1.5cm3), the average number of palpable tumors per mouse were 1.15 and 2 for control and DMBA groups, respectively. Lung metastasis in the DMBA group was significantly higher than control mice (40% DMBA vs. 5% control). These data show that DMBA exposure significantly promoted erbB-2 mediated carcinogenesis. Whole mount analysis showed that at 14 weeks of age there was marked expansion of the ducts, whereas lateral branching of mammary glands from DMBA mice appeared to be inhibited, suggesting that DMBA stimulates glandular growth while inhibiting differentiation in this model. Western blot analysis of mammary tissues at 14 weeks of age showed that ERα, p-ERα, cyclin D1, Bcl-2, c-myc, EGFR, erbB-2 and erbB-3 protein levels were significantly increased in DMBA treated mice, which was accompanied by increased phosphorylation/activation of erbB-2, EGFR, ERK and Akt1. These data suggest that DMBA induced activation of both ER and receptor tyrosine kinase (RTK) pathways. Interestingly, tumor histology between tumors from control and DMBA treated mice was similar. However, analysis of tumor karotypes from control and DMBA mice showed that tumors from DMBA mice displayed more chromosomal alterations; an addition to chromosome 4 and trisomy 5 in the control mouse, trisomy 2, trisomy 3 and a deletion to chromosome 4 were detected from tumors of the DMBA mice, suggesting that DMBA induced chromosomal instability contributed to tumor promotion in this model. Taken together, our data suggest that DMBA promoted erbB-2 mediated carcinogenesis. In addition to its well known mutational effect, DMBA induced deregulation of ER and erbB-2 pathways plays a critical role in this process. Citation Format: Zhikun Ma, Stanley Kosanke, Morgan Carrington, Xiaohe Yang. DMBA promotes erbB-2 mediated carcinogenesis through activation of estrogen receptor and receptor tyrosine kinase pathways. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 5345. doi:10.1158/1538-7445.AM2014-5345
- Published
- 2014
- Full Text
- View/download PDF
45. Abstract 4790: Adult exposure to a soy-rich diet promotes mammary tumor development in MMTV-erbB-2 transgenic mice through induction of ER-erbB-2 crosstalk
- Author
-
Xiaohe Yang and Zhikun Ma
- Subjects
Cancer Research ,medicine.medical_specialty ,Mammary tumor ,biology ,business.industry ,medicine.drug_class ,Estrogen receptor ,Genistein ,medicine.disease ,Tyrosine-kinase inhibitor ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Endocrinology ,Breast cancer ,Cyclin D1 ,Oncology ,chemistry ,ErbB ,Internal medicine ,medicine ,biology.protein ,skin and connective tissue diseases ,business - Abstract
The impact of soy and soy isoflavones, such as genistein, on breast cancer risk remains controversial. Although meta-analysis of multiple studies suggests that soy intake may be associated with a small reduction in breast cancer risk, increasing reports show that soy/genistein may be deleterious to breast cancer under certain conditions. Therefore, it is urgent to identify the factors that may affect the safety and risk of soy/genistein on breast cancer. We tackled this clinically relevant problem by examining the effect of adult exposure to soy/genistein on a subgroup of breast cancers that are positive for both estrogen receptor (ER) and erbB-2. erbB-2 is a receptor tyrosine kinase that is frequently overexpressed in breast cancer. ER+/erbB-2+ breast cancer accounts for ∼15-20% of breast cancer. Available data suggest that low dose soy/genistein stimulates ER+ breast cancer cells. On the other hand, genistein, a well known tyrosine kinase inhibitor, may inhibit erbB-2 overexpressing cells. The effect soy/genistein on ER+/erbB-2+ breast cancer is a dilemmatic question to be addressed. In this study MMTV-erbB-2 transgenic mice fed an AIN-93G diet were switched to a soy-rich diet (Purina 5001) at 20 weeks of age. Mammary tumor development in these mice was compared to that in control mice on a lifelong AIN-93G diet. We found that the adult switch to a soy diet accelerated mammary tumor development in this model. Average tumor latency for the control and soy-exposed groups were 36 and 32 weeks, respectively. Whole mount analysis indicated that the soy-diet-switch induced proliferation in the mammary glands as indicated by increased BrdU incorporation and epithelial density. We also found that changes in signaling of ER and erbB-2 pathways were modest at 24 weeks but more evident at 32 weeks. The soy-diet-switch induced ERα expression and phosphorylation which led to increased expression of ER target genes, including c-myc, cyclin D1, and Bcl-2. These changes were accompanied by increased phosphorylation/activation of EGFR, erbB-2, erbB-3, AKT1, and ERK½. Expression of EGFR and erbB-3 was also increased. Although the mammary tumors developed in this model were ER negative, mammary epithelial cells in the premalignant tissues were ER+/erbB-2+. These results provide in vivo evidence that the soy-rich diet stimulates the proliferation of ER+/erbB-2+ mammary epithelial cells, suggesting that soy/genistein-associated estrogenic activity overrides its tyrosine kinase inhibitor activity, possibly through ER-erbB-2 crosstalk. Our data suggests that ER+/erbB-2+ breast cancer might be particularly vulnerable to the risk associated with adult exposure to soy/genistein, and therefore patients with ER+/erbB-2+ tumors should be alerted. Citation Format: XiaoHe Yang, Zhikun Ma. Adult exposure to a soy-rich diet promotes mammary tumor development in MMTV-erbB-2 transgenic mice through induction of ER-erbB-2 crosstalk. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4790. doi:10.1158/1538-7445.AM2013-4790
- Published
- 2013
- Full Text
- View/download PDF
46. Abstract 4874: Bisphenol A exposure negates tamoxifen-mediated chemoprevention in MMTV-erbB2 transgenic mice
- Author
-
Zhikun Ma, Xiaohe Yang, and Yunbo Jiang
- Subjects
endocrine system ,Cancer Research ,medicine.medical_specialty ,Mammary tumor ,urogenital system ,business.industry ,Cancer ,medicine.disease ,medicine.disease_cause ,Subcutaneous injection ,chemistry.chemical_compound ,Xenoestrogen ,Endocrinology ,Breast cancer ,Oncology ,chemistry ,Internal medicine ,medicine ,Tamoxifen Citrate ,Carcinogenesis ,business ,hormones, hormone substitutes, and hormone antagonists ,Tamoxifen ,medicine.drug - Abstract
Bisphenol A (BPA), the building block of polycarbonate plastics with estrogenic activity, is one of the most common chemicals people are exposed to in daily life. Increasing reports suggest a link between BPA exposure and increased breast cancer risk. Although the effect of early BPA exposure and breast cancer risk later in life has been extensively studied, whether this xenoestrogen interferes with hormonal therapeutic agents, such as tamoxifen, has hardly been documented. In this study, we tested the effect of BPA exposure on mammary tumor development and tamoxifen-mediated chemoprevention in MMTV-erbB2 transgenic mice. BPA (500 ng/kg in sesame oil) was administered daily by subcutaneous injection for 8 weeks, starting from 8 weeks of age. Citrate tamoxifen at a dose of 1 mg/kg/day was administered by oral gavage alone or in combination with BPA. Based on tumor latency, we found that BPA alone accelerated mammary tumor development whereas tamoxifen treatment profoundly delayed the tumorigenesis. A combination of tamoxifen-treatment and BPA exposure, however, significantly (p Citation Format: XiaoHe Yang, Zhikun Ma, Yunbo Jiang. Bisphenol A exposure negates tamoxifen-mediated chemoprevention in MMTV-erbB2 transgenic mice. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 4874. doi:10.1158/1538-7445.AM2013-4874
- Published
- 2013
- Full Text
- View/download PDF
47. Abstract 5452: In utero exposure to bisphenol A promotes mammary tumor development in MMTV-erbB-2 transgenic mice
- Author
-
Xia Cao, Xiaohe Yang, and Zhikun Ma
- Subjects
Genetically modified mouse ,Cancer Research ,Mammary tumor ,medicine.medical_specialty ,business.industry ,Estrogen receptor ,Environmental exposure ,medicine.disease ,Breast cancer ,Cyclin D1 ,Endocrinology ,Oncology ,In utero ,ErbB ,Internal medicine ,medicine ,business - Abstract
Increasing evidence indicates that environmental exposure plays a critical role in breast cancer etiology. Bisphenol A (BPA), the building block of polycarbonate plastics with estrogenic activity, is one of the most common chemicals exposed in daily life. Recent studies suggest a link between BPA exposure and increased breast cancer risk. However, specific conditions and the underlying mechanisms of BPA exposure associated breast cancer risk remain unclear. This study aimed to investigate the effects of in utero exposure to BPA on mammary tumor development in MMTV-erbB-2 transgenic mice, which exemplifies a scenario of gene-environmental interaction. Since erbB-2 is amplified/overexpressed in approximately 30% of breast cancer, which has been associated with poor prognosis and therapeutic resistance, use of this clinically relevant mammary tumor model is of high translational value. In this study, pregnant MMTV-erbB-2 mice were subcutaneously injected with 0, 25 ng, 250 ng and 250 µg/Kg body weight BPA daily between day 13 and day 19 of gestation. We found that in utero exposed mice in the low dose (25 and 250 ng) groups, but not the high dose group (250 µg), displayed earlier vaginal opening and prolonged estrous phase, suggesting a dose dependent pro-estrogenic effect of in utero exposure to BPA. Whole mount analysis showed that mammary glands of mice with in utero exposure to low dose BPA had a longer ductal extension at 6 weeks and increased lateral branching/alveolar structures at 10 weeks, which was more significant in the 250 ng group but less evident in the high dose group. Molecular analysis of mammary tissues at 10 weeks indicated that in utero exposure to BPA increased phosphorylation/activation of erbB-2, EGFR, erbB-3 Erk1/2 and Akt, and expression of erbB-3 and EGFR. Signaling of estrogen receptor (ER) pathway was upregulated concomitantly, as reflected by increased expression and phosphorylation of ERα and upregulation of cyclin D1 and c-myc. More importantly, mice in the 250 ng group but not the high dose group, developed tumor earlier than the mice in the control group, suggesting that dose is a critical factor affecting the outcomes. These data demonstrated that in utero exposure to BPA may promote mammary tumor development in erbB-2 transgenic mice in a dose dependent manner. Concomitant activation of the erbB-2 and ER pathways in the premalignant tissues suggests that induction of ER-erbB-2 crosstalk may play a critical role in mammary tumorigenesis promoted by in utero exposure to BPA, which underscores the significance of gene-environment interaction in early exposure to BPA associated breast cancer risk. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 5452. doi:1538-7445.AM2012-5452
- Published
- 2012
- Full Text
- View/download PDF
48. Abstract 548: Erythropoietin promotes mammary tumor development in MMTV-erbB-2 transgenic mice
- Author
-
Wei Qun Ding, Zhizhuang Joe Zhao, Xiaohe Yang, Xia Cao, Xiaoxi Zhang, Shihe Yang, Wanming Zhao, and Zhikun Ma
- Subjects
Tube formation ,Cancer Research ,medicine.medical_specialty ,Mammary tumor ,business.industry ,medicine.disease ,Metastasis ,Erythropoietin receptor ,Endocrinology ,Oncology ,Erythropoietin ,ErbB ,Tumor progression ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Erythropoiesis ,business ,medicine.drug - Abstract
Erythropoietin (EPO), a cytokine that stimulates erythropoiesis, is frequently used to treat anemia in cancer patients. Recent clinical studies indicate that EPO administration might be associated with tumor progression and reduced overall survival rate. However, the mechanisms of EPO associated risk in cancer patients remain poorly understood. In particular, a spontaneous tumor model that allows us to test the effects of EPO on tumor development/progression and provide mechanistic insights into multifactoral interactions in a physiological context has not been established. In this study, we used the MMTV-erbB-2 transgenic mouse model to test the specific effect of EPO on erbB-2 overexpressing breast cancers and aimed to establish a spontaneous tumor model for relevant studies. Female erbB-2 transgenic mice were injected with EPO? at 2000 U/Kg body weight, twice a week for three weeks (between week 20 and 22). We found that EPO treatment during the premalignant risk window not only increased hemoglobin levels but also significantly promoted mammary tumor development. In contrast to a 36-week mean latency of tumor development in control group, the mean latency for EPO exposed mice was 30 weeks. Moreover, EPO exposure resulted in increased pulmonary metastasis. Whole mount analysis of mammary gland at 23 weeks showed that EPO exposure induced ductal branching and alveolar outgrowth, which was consistent with increased BrdU incorporation in EPO exposed mammary tissues. Molecular analysis of mammary tissues at 23 weeks showed that the protein levels of p-Jak2, p-erbB-2, p-Erk and p-Akt1 were significantly increased, which was accompanied by increased expression of EGFR, erbB-3, IGFR, Bcl-2, cyclin D1 and VEGF. These data suggest that EPO exposure induced the activation of both EPOR/Jak2 and erbB-2/EGFR/erbB-3 pathways, and had a broad impact on signal transduction and gene expression in mammary tissues. Although the difference in signal transduction and gene expression of tumor tissues between control and EPO exposed groups was less significant, tumors from EPO treated mice showed increased microvessel density as compared to controls, suggesting EPO exposure induced a long lasting proangiogenic effect. Consistently, conditioned medium extracted from EPO treated mammary tissues at 23 weeks significantly increased tube formation of endothelial cells. Taken together, our results demonstrate that EPO treatment promotes the development and metastasis of erbB-2 overexpressing mammary tumors and established the MMTV-erbB-2 mouse model for the studies of EPO associated cancer risk. The underlying mechanisms involve enhanced activation of erbB-2 pathway through EPOR/Jak2 activation and increased proangiogenic activities in EPO exposed mammary tissues. Since erbB-2 is overexpressed in about 30% of human breast cancers, these data will have significant impact on safe use of EPO in breast cancer patients with anemia. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 548. doi:1538-7445.AM2012-548
- Published
- 2012
- Full Text
- View/download PDF
49. Abstract 2838: Brief exposure to gefitinib/Iressa during premalignant risk window provides lifelong protection from mammary tumor development in MMTV-erbB-2 transgenic mice
- Author
-
Zhikun Ma, Xiaohe Yang, Stanley D. Kosanke, Yunbo Jiang, Xiaoshan Feng, and Shihe Yang
- Subjects
Cancer Research ,medicine.medical_specialty ,Mammary tumor ,biology ,business.industry ,medicine.drug_class ,AKT1 ,Estrogen receptor ,Receptor tyrosine kinase ,Gefitinib ,Endocrinology ,Oncology ,ErbB ,Estrogen ,Internal medicine ,Cancer research ,biology.protein ,Medicine ,business ,Estrogen receptor alpha ,medicine.drug - Abstract
Although chemopreventive agents targeting estrogen/estrogen receptor (ER) pathway have been effective for ER+ breast cancer, hormonal resistance associated with increased receptor tyrosine kinase activities, such as erbB-2 and/or EGFR, remains a significant issue in beast cancer prevention. Previous studies demonstrated that administration of gefitinib, an orally active EGFR tyrosine kinase inhibitor, to MMTV-erbB-2 transgenic mice inhibited mammary tumor development in these mice, suggesting that gefitinib is a promising agent for the prevention of ER-negative breast cancer. However, the prevention was achieved by high dose (100 mg/kg) and prolonged drug exposure (from 12 to 50 weeks). The tolerance to high dose/long-term of drug administration for the prevention regimens raised concerns in clinical application. In this study, we tested the efficacy of brief exposure to gefitinib during the high risk window in the prevention of mammary tumor development in the MMTV-erbB-2 transgenic mice and studied the underlying mechanisms. We found that brief exposure of gefitinib to these mice at a dosage of 100 mg/kg/day from 16 to 23 weeks (total 8 wks) resulted in significant chemopreventive effects. In contrast to no tumor-free animals in the control group by 52 weeks, mice in gefitinib treated group remained 65% tumor-free at the same age, which was comparable to the results from a previous report that mice with lifelong gefitinib were 75% tumor-free by 45 weeks. We further demonstrated that delayed tumor development in the treated mice was preceded with decreased mammary epithelial density. Molecular analysis indicated that gefitinib inhibited phosphorylation and expression of EGFR, erbB-3 and endogenous erbB-2 in premalignant mammary tissues. Phosphorylation of Akt1 and Erk1/2 was also significantly downregulated. Importantly, although tumors developed from this model were ER-, mammary tissues in the premalignant stage were ERα+; and gefitinib treatment drastically inhibited the phosphorylation and expression of ERα and the transcription of ER target genes, including EGF, EGFR, ESR1, Bcl-2, cyclin D1 and c-myc. Moreover, BrdU incorporation analysis demonstrates that cell proliferation in the mammary glands in gefitinib treated mice was significantly inhibited. Taken together, these data demonstrate that brief treatment with EGFR/erbB-2 targeting agents at the appropriate risk window may provide lifelong protection from mammary tumors, and inhibition of ER-erbB-2 crosstalk may play a critical role in this long lasting preventive process. Given the concerns associated with high dose/long-term gefitinib treatment, our findings are of great significance in directing clinical application of EGFR/erbB-2 targeting agents as chemopreventive agents. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2838. doi:10.1158/1538-7445.AM2011-2838
- Published
- 2011
- Full Text
- View/download PDF
50. KDM4C, a H3K9me3 Histone Demethylase, is Involved in the Maintenance of Human ESCC-Initiating Cells by Epigenetically Enhancing SOX2 Expression
- Author
-
Shegan Gao, Yiwen Liu, Fuyou Zhou, Jinyu Kong, Ruinuo Jia, Qimin Zhan, Xiang Yuan, Zhikun Ma, Gang Liu, and Na Li
- Subjects
0301 basic medicine ,Cancer Research ,education.field_of_study ,biology ,Cell ,Population ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Phenotype ,lcsh:RC254-282 ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Histone ,SOX2 ,biology.protein ,Cancer research ,medicine ,Demethylase ,Ectopic expression ,education ,Intracellular - Abstract
Our studies investigating the existence of tumor-initiating cell (TIC) populations in human esophageal squamous cell carcinoma (ESCC) had identified a subpopulation of cells isolated from ESCC patient-derived tumor specimens marked by an ALDHbri+ phenotype bear stem cell-like features. Importantly, KDM4C, a histone demethylase was enhanced in ALDHbri+ subpopulation, suggesting that strategies interfering with KDM4C may be able to target these putative TICs. In the present study, by genetic and chemical means, we demonstrated that, KDM4C blockade selectively decreased the ESCC ALDHbri+ TICs population in vitro and specifically targeted the TICs in ALDHbri+-derived xenograft, retarding engraftment. Subsequent studies of the KDM4C functional network identified a subset of pluripotency-associated genes (PAGs) and aldehyde dehydrogenase family members to be preferentially down-regulated in KDM4C inhibited ALDHbri+ TICs. We further supported a model in which KDM4C maintains permissive histone modifications with a low level of H3K9 methylation at the promoters of several PAGs. Moreover, ectopic expression of SOX2 restored KDM4C inhibition-dependent ALDHbri+ TIC properties. We further confirmed these findings by showing that the cytoplasmic and nuclear KDM4C staining increased with adverse pathologic phenotypes and poor patient survival. Such staining pattern of intracellular KDM4C appeared to overlap with the expression of SOX2 and ALDH1. Collectively, our findings provided the insights into the development of novel therapeutic strategies based on the inhibition of KDM4C pathway for the eliminating of ESCC TIC compartment.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.