144 results on '"Binhua P. Zhou"'
Search Results
2. NAC1 promotes stemness and regulates myeloid-derived cell status in triple-negative breast cancer
- Author
-
Chrispus Ngule, Ruyi Shi, Xingcong Ren, Hongyan Jia, Felix Oyelami, Dong Li, Younhee Park, Jinhwan Kim, Hami Hemati, Yi Zhang, Xiaofang Xiong, Andrew Shinkle, Nathan L. Vanderford, Sara Bachert, Binhua P. Zhou, Jianlong Wang, Jianxun Song, Xia Liu, and Jin-Ming Yang
- Subjects
TNBC ,NAC1 ,Cancer stem cells ,MDSCs ,NK cells ,TME ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Triple negative breast cancer (TNBC) is a particularly lethal breast cancer (BC) subtype driven by cancer stem cells (CSCs) and an immunosuppressive microenvironment. Our study reveals that nucleus accumbens associated protein 1 (NAC1), a member of the BTB/POZ gene family, plays a crucial role in TNBC by maintaining tumor stemness and influencing myeloid-derived suppressor cells (MDSCs). High NAC1 expression correlates with worse TNBC prognosis. NAC1 knockdown reduced CSC markers and tumor cell proliferation, migration, and invasion. Additionally, NAC1 affects oncogenic pathways such as the CD44-JAK1-STAT3 axis and immunosuppressive signals (TGFβ, IL-6). Intriguingly, the impact of NAC1 on tumor growth varies with the host immune status, showing diminished tumorigenicity in natural killer (NK) cell-competent mice but increased tumorigenicity in NK cell-deficient ones. This highlights the important role of the host immune system in TNBC progression. In addition, high NAC1 level in MDSCs also supports TNBC stemness. Together, this study implies NAC1 as a promising therapeutic target able to simultaneously eradicate CSCs and mitigate immune evasion.
- Published
- 2024
- Full Text
- View/download PDF
3. Aldh2 deficiency plays a dual role in lung tumorigenesis and tumor progression
- Author
-
Hongjia Zhang, Xueqian Sun, Zhanming Li, Tingting Liu, Fang Zhang, Xinyu Meng, Kaimi Li, Jianhua Xu, Wei He, Bo Jing, Tong Wang, Na Ni, Beibei Sun, Feng Yao, Yadi Wu, Qi Wang, Jing Du, Eugene Y. Chin, Binhua P. Zhou, Ping Jiang, Lishun Wang, and Jiong Deng
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2024
- Full Text
- View/download PDF
4. NF-κB represses retinoic acid receptor–mediated GPRC5A transactivation in lung epithelial cells to promote neoplasia
- Author
-
Hongyong Song, Xiaofeng Ye, Yueling Liao, Siwei Zhang, Dongliang Xu, Shuangshuang Zhong, Bo Jing, Tong Wang, Beibei Sun, Jianhua Xu, Wenzheng Guo, Kaimi Li, Min Hu, Yanbin Kuang, Jing Ling, Tuo Zhang, Yadi Wu, Jing Du, Feng Yao, Y. Eugene Chin, Qi Wang, Binhua P. Zhou, and Jiong Deng
- Subjects
Oncology ,Medicine - Abstract
Chronic inflammation is associated with lung tumorigenesis, in which NF-κB–mediated epigenetic regulation plays a critical role. Lung tumor suppressor G protein–coupled receptor, family C, member 5A (GPRC5A), is repressed in most non–small cell lung cancer (NSCLC); however, the mechanisms remain unclear. Here, we show that NF-κB acts as a transcriptional repressor in suppression of GPRC5A. NF-κB induced GPRC5A repression both in vitro and in vivo. Intriguingly, transactivation of NF-κB downstream targets was not required, but the transactivation domain of RelA/p65 was required for GPRC5A repression. NF-κB did not bind to any potential cis-element in the GPRC5A promoter. Instead, p65 was complexed with retinoic acid receptor α/β (RARα/β) and recruited to the RA response element site at the GPRC5A promoter, resulting in disrupted RNA polymerase II complexing and suppressed transcription. Notably, phosphorylation on serine 276 of p65 was required for interaction with RARα/β and repression of GPRC5A. Moreover, NF-κB–mediated epigenetic repression was through suppression of acetylated histone H3K9 (H3K9ac), but not DNA methylation of the CpG islands, at the GPRC5A promoter. Consistently, a histone deacetylase inhibitor, but not DNA methylation inhibitor, restored GPRC5A expression in NSCLC cells. Thus, NF-κB induces transcriptional repression of GPRC5A via a complex with RARα/β and mediates epigenetic repression via suppression of H3K9ac.
- Published
- 2023
- Full Text
- View/download PDF
5. Metabolic reprogram associated with epithelial-mesenchymal transition in tumor progression and metastasis
- Author
-
Yifan Wang, Chenfang Dong, and Binhua P. Zhou
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Epithelial-mesenchymal Transition (EMT) is a de-differentiation program that imparts tumor cells with the phenotypic and cellular plasticity required for drug resistance, metastasis, and recurrence. This dynamic and reversible events is governed by a network of EMT-transcription factors (EMT-TFs) through epigenetic regulation. Many chromatin modifying-enzymes utilize metabolic intermediates as cofactors or substrates; this suggests that EMT is subjected to the metabolic regulation. Conversely, EMT rewires metabolic program to accommodate cellular changes during EMT. Here we summarize the latest findings regarding the epigenetic regulation of EMT, and discuss the mutual interactions among metabolism, epigenetic regulation, and EMT. Finally, we provide perspectives of how this interplay contributes to cellular plasticity, which may result in the clinical manifestation of tumor heterogeneity. Keywords: Epigenetics, Epithelial-mesenchymal transition, Heterogeneity, Metabolism, Plasticity
- Published
- 2020
- Full Text
- View/download PDF
6. Deletion of tetraspanin CD151 alters the Wnt oncogene-induced mammary tumorigenesis: A cell type-linked function and signaling
- Author
-
Hongxia Li, Jieming Li, Rongbo Han, Xinyu Deng, Junfong Shi, Huanhuan Huang, Nevean Hamad, Abigail McCaughley, Jinpeng Liu, Chi Wang, Kuey Chen, Dongping Wei, Jun Qiang, Sean Thatcher, Yadi Wu, Chunming Liu, Olivier Thibault, Xiaowei Wei, Song Chen, Hai Qian, Binhua P. Zhou, Pao Xu, and Xiuwei H. Yang
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Tetraspanin CD151 is increasingly implicated as a multifaceted mediator of cancer development and progression. Here we investigated the role of CD151 in breast cancer in the context of the Wnt oncogenic activation. Our data showed that removal of one or both of CD151 alleles in the MMTV-Wnt1 model significantly decreased the tumor-free survival of mice from 34 weeks on average to 22 weeks and 18 weeks, respectively. This effect coincided with an accelerated tumor growth and an increased number of Ki-67+ proliferative cells. Mechanistically, the CD151-deficient tumors were largely ER+, and exhibited hyperactivation of the Wnt pathway as reflected by a marked upregulation in β-catenin and Cyclin D1, and their target genes. In addition, E-cadherin displayed a cytosolic distribution and transcription factor Snail was markedly upregulated. Collectively, this data implies that CD151 suppresses the Wnt1-driven tumorigenesis, at least in part, via counteracting the epithelial-mesenchymal transition (EMT)-like program in luminal epithelial cells. Meanwhile, the proportion of tumor cells expressing CK5 or p63, the biomarkers of myoepithelial/basal cells, markedly decreased in the absence of CD151. This change was accompanied by a decreased invasiveness of tumors and their incompetence to form a long-term cell culture. Consistent with this basal cell-linked role, the CD151 downregulation impairs mammosphere formation in MCF-10A cells and the defect was rescued by re-expression of intact CD151 ORF, but not its integrin binding-defective mutant. Overall, our study suggests that CD151 is a key player in the Wnt oncogene-driven tumorigenesis and impacts breast cancer malignancy in a cell type-dependent manner.
- Published
- 2019
- Full Text
- View/download PDF
7. Targeting the BRD4/FOXO3a/CDK6 axis sensitizes AKT inhibition in luminal breast cancer
- Author
-
Jingyi Liu, Zhibing Duan, Weijie Guo, Lei Zeng, Yadi Wu, Yule Chen, Fang Tai, Yifan Wang, Yiwei Lin, Qiang Zhang, Yanling He, Jiong Deng, Rachel L. Stewart, Chi Wang, Pengnian Charles Lin, Saghi Ghaffari, B. Mark Evers, Suling Liu, Ming-Ming Zhou, Binhua P. Zhou, and Jian Shi
- Subjects
Science - Abstract
The molecular mechanism underlying the resistance of AKT inhibitors in breast cancer is still elusive. Here, the authors demonstrate that BRD4/FOXO3a axis upregulates CDK6 promoter activity to promote resistance to AKT inhibition in breast cancer cells and that blocking the action of CDK6 re-sensitizes resistant cancer cells to growth inhibition.
- Published
- 2018
- Full Text
- View/download PDF
8. Snail determines the therapeutic response to mTOR kinase inhibitors by transcriptional repression of 4E-BP1
- Author
-
Jun Wang, Qing Ye, Yanan Cao, Yubin Guo, Xiuping Huang, Wenting Mi, Side Liu, Chi Wang, Hsin-Sheng Yang, Binhua P. Zhou, B. Mark Evers, and Qing-Bai She
- Subjects
Science - Abstract
4E-BP1 is a translational repressor critical in mTOR signaling, whereas Snail is a critical promoter of epithelial to mesenchymal transition. Here the authors show that Snail induces resistance to mTOR inhibitors by repressing 4E-BP1 expression and promoting cell cycle progression via upregulating cycD.
- Published
- 2017
- Full Text
- View/download PDF
9. Dub3 inhibition suppresses breast cancer invasion and metastasis by promoting Snail1 degradation
- Author
-
Yadi Wu, Yu Wang, Yiwei Lin, Yajuan Liu, Yifan Wang, Jianhang Jia, Puja Singh, Young-In Chi, Chi Wang, Chenfang Dong, Wei Li, Min Tao, Dana Napier, Qiuying Shi, Jiong Deng, B Mark Evers, and Binhua P. Zhou
- Subjects
Science - Abstract
Snail1 is a key factor controlling epithelial-to-mesenchymal transition and cancer metastasis. While the E3 ligases responsible for Snail1 ubiquitination and degradation have been defined, the deubiquitinating enzyme is unknown. Here Zhou and colleagues show that Dub3 stabilizes Snail1 by removing ubiquitin, thus impacting breast cancer cell metastasis.
- Published
- 2017
- Full Text
- View/download PDF
10. Oncometabolite modification of Keap1 links GSTZ1 deficiency with cancer
- Author
-
Weijie Guo and Binhua P. Zhou
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Metabolic abnormalities are emerging as an active driver to the development, progression and metastasis of various tumors. In the recent issue of the EMBO Journal, Yang and colleagues identified that succinylacetone (SA) could act as an oncometabolite and that accumulation of SA activates the NRF2/IGF1R axis in hepatocellular carcinoma (HCC) development. These discoveries not only yield great insights in the understanding of tumor biology, but also hold significant clinical ramifications, as these findings may pave a new way for the early diagnosis and treatment of HCC.
- Published
- 2019
- Full Text
- View/download PDF
11. Metabolic Reprogramming of Cancer-Associated Fibroblasts by IDH3α Downregulation
- Author
-
Daoxiang Zhang, Yongbin Wang, Zhimin Shi, Jingyi Liu, Pan Sun, Xiaodan Hou, Jian Zhang, Shimin Zhao, Binhua P. Zhou, and Jun Mi
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Cancer-associated fibroblasts (CAFs) provide critical metabolites for tumor growth and undergo metabolic reprogramming to support glycolysis. However, the molecular mechanisms responsible for this change remain unclear. Here, we report that TGF-β1- or PDGF-induced CAFs switch from oxidative phosphorylation to aerobic glycolysis. We identify downregulation of isocitrate dehydrogenase 3α (IDH3α) as a marker for this switch. Furthermore, miR-424 downregulates IDH3α during CAF formation. Downregulation of IDH3α decreases the effective level of α-ketoglutarate (α-KG) by reducing the ratio of α-KG to fumarate and succinate, resulting in PHD2 inhibition and HIF-1α protein stabilization. The accumulation of HIF-1α, in turn, promotes glycolysis by increasing the uptake of glucose, upregulating expression of glycolytic enzymes under normoxic conditions, and inhibiting oxidative phosphorylation by upregulating NDUFA4L2. CAFs from tumor samples exhibit low levels of IDH3α, and overexpression of IDH3α prevents transformation of fibroblasts into CAFs. Our studies reveal IDH3α to be a critical metabolic switch in CAFs.
- Published
- 2015
- Full Text
- View/download PDF
12. The Deubiquitinase USP28 Stabilizes LSD1 and Confers Stem-Cell-like Traits to Breast Cancer Cells
- Author
-
Yadi Wu, Yifan Wang, Xiuwei H. Yang, Tiebang Kang, Yongxiang Zhao, Chi Wang, B. Mark Evers, and Binhua P. Zhou
- Subjects
Biology (General) ,QH301-705.5 - Abstract
LSD1 is a critical chromatin modulator that controls cellular pluripotency and differentiation through the demethylation of H3K4me1/2. Overexpression of LSD1 has been observed in many types of tumors and is correlated with its oncogenic effects in tumorigenesis. However, the mechanism leading to LSD1 upregulation in tumors remains unclear. Using an unbiased siRNA screening against all the human deubiquitinases, we identified USP28 as a bona fide deubiquitinase of LSD1. USP28 interacted with and stabilized LSD1 via deubiquitination. USP28 overexpression correlated with LSD1 upregulation in multiple cancer cell lines and breast tumor samples. Knockdown of USP28 resulted in LSD1 destabilization, leading to the suppression of cancer stem cell (CSC)-like characteristics in vitro and inhibition of tumorigenicity in vivo, which can be rescued by ectopic LSD1 expression. Our study reveals a critical mechanism underlying the epigenetic regulation by USP28 and provides another treatment approach against breast cancer.
- Published
- 2013
- Full Text
- View/download PDF
13. Inflammation A Driving Force Speeds Cancer Metastasis
- Author
-
Yadi WU and Binhua P. ZHOU
- Subjects
Lung neoplasms ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2009
14. Regulation of snail in epithelial-mesenchymal transition and breast cancer progression and metastasis
- Author
-
Binhua P. Zhou
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Published
- 2017
- Full Text
- View/download PDF
15. Identification of Active Bronchioalveolar Stem Cells as the Cell of Origin in Lung Adenocarcinoma
- Author
-
Huijing Yin, Bo Jing, Dongliang Xu, Wenzheng Guo, Beibei Sun, Jie Zhang, Yueling Liao, Hongyong Song, Tong Wang, Shuli Liu, Yanbin Kuang, Min Hu, Kaimi Li, Siwei Zhang, Hongjia Zhang, Jianhua Xu, Xue Li, Jing Du, Yadi Wu, Yingli Wu, Qi Wang, Feng Yao, Yueh Eugene Chin, Binhua P. Zhou, and Jiong Deng
- Subjects
Mice, Knockout ,Cancer Research ,Lung Neoplasms ,Carcinogenesis ,Stem Cells ,NF-kappa B ,Adenocarcinoma of Lung ,respiratory system ,Receptors, G-Protein-Coupled ,ErbB Receptors ,Mice ,Cell Transformation, Neoplastic ,Oncology ,Animals ,Humans ,Lung - Abstract
While initiation is established as a critical step in tumorigenesis, the identity of the cell of origin for lung adenocarcinoma and the mechanism controlling susceptibility to initiation remain elusive. Here we show that lung tumor suppressor Gprc5a-knockout (KO) mice are susceptible to initiation of lung tumorigenesis. Bronchioalveolar stem cells (BASC) and alveolar type 2 (AT2) cells were aberrantly expanded in Gprc5a-KO mouse lungs compared with those in wild-type (WT) mice, suggesting that Gprc5a-KO might confer susceptibility to initiation by increasing the cell of origin in mouse lungs. BASCs from Gprc5a-KO mice (KO-BASC) exhibited significantly increased stemness and self-renewal potential and reduced differentiation capacity compared with BASCs from WT mice (WT-BASC). AT2 cells did not possess self-renewal potential regardless of Gprc5a status. KO-BASCs expressed a stem-like gene profile with upregulated Abcg2, EGFR, and NF-κB signaling compared with WT-BASCs. Blockade of EGFR and NF-κB signaling inhibited both expansion of BASC and AT2 cells and lung tumorigenesis. Abcg2 was expressed in active KO-BASCs as well as in lung tumor cells but not in quiescent WT-BASCs or AT2 cells, supporting that lung adenocarcinoma cells are derived from Abcg2-positive KO-BASCs (active). Taken together, Gprc5a deletion leads to expansion of active BASCs via dysregulated EGFR and NF-κB signaling that confers susceptibility to initiation of lung tumorigenesis, marking Abcg2-positive BASCs as candidate cell of origin for lung adenocarcinoma. Significance: Identification of active bronchioalveolar stem cells as lung adenocarcinoma cells of origin provides insights into mechanisms of lung tumorigenesis and could facilitate development of effective strategies for cancer prevention and therapy. See related commentary by Osborne and Minna, p. 972
- Published
- 2022
- Full Text
- View/download PDF
16. Supplementary Data from Identification of Active Bronchioalveolar Stem Cells as the Cell of Origin in Lung Adenocarcinoma
- Author
-
Jiong Deng, Binhua P. Zhou, Yueh Eugene Chin, Feng Yao, Qi Wang, Yingli Wu, Yadi Wu, Jing Du, Xue Li, Jianhua Xu, Hongjia Zhang, Siwei Zhang, Kaimi Li, Min Hu, Yanbin Kuang, Shuli Liu, Tong Wang, Hongyong Song, Yueling Liao, Jie Zhang, Beibei Sun, Wenzheng Guo, Dongliang Xu, Bo Jing, and Huijing Yin
- Abstract
Supplementary Data from Identification of Active Bronchioalveolar Stem Cells as the Cell of Origin in Lung Adenocarcinoma
- Published
- 2023
- Full Text
- View/download PDF
17. Data from IL6/STAT3 Signaling Orchestrates Premetastatic Niche Formation and Immunosuppressive Traits in Lung
- Author
-
Jiong Deng, Feng Yao, Binhua P. Zhou, Tuo Zhang, Yanbin Kuang, Jing Ling, Siwei Zhang, Min Hu, Kaimi Li, Wenzheng Guo, Hongyong Song, Yueling Liao, Dongliang Xu, Jianhua Xu, Beibei Sun, Tong Wang, and Bo Jing
- Abstract
Cancer cells that succeed in forming metastasis need to be reprogrammed to evade immune surveillance and survive in a new microenvironment. This is facilitated by metastatic niches that are either postformed through reciprocal signaling between tumor cells and local stromal cells or preformed as premetastatic niches before tumor cell arrival. IL6/STAT3 signaling is aberrantly activated in lung tumorigenesis and metastasis, however, the roles and mechanisms of action of IL6 remain controversial. Here, we showed that blockade of intrinsic STAT3 signaling in lung tumor cells suppressed lung metastasis in immune-competent syngeneic mice, but not in immune-deficient nude mice. Consistently, repression of STAT3 signaling in tumor cells made them susceptible to T-cell–mediated cytotoxicity. Thus, STAT3-mediated immunosuppression is crucial for metastasis. Noticeably, lung metastasis was greatly increased in Gprc5a-knockout (ko; 5a−/−) mice compared with wild-type mice, which correlated with upregulated IL6 in the tumor microenvironment. Depletion of IL6 via combined deletion of Il6 and Gprc5a genes almost completely eliminated lung metastasis in Gprc5a-ko/Il6-ko (5a−/−;Il6−/−) mice. Mechanistically, dysregulated IL6 reprogrammed the STAT3 pathway in metastatic tumor cells, and induced recruitment of myeloid-derived suppressor cells and polarized macrophages to evade host immunity. Consistently, IHC staining showed that activated STAT3 correlated with repressed infiltration of CD8+ T cells in non–small cell lung cancer. Therefore, IL6/STAT3 signaling is crucial for orchestrating premetastatic niche formation and immunosuppression in lung.Significance: IL6 plays important roles not only in cell autonomous propensity for metastasis, but also in establishing the metastatic niche.
- Published
- 2023
- Full Text
- View/download PDF
18. Data from Identification of Active Bronchioalveolar Stem Cells as the Cell of Origin in Lung Adenocarcinoma
- Author
-
Jiong Deng, Binhua P. Zhou, Yueh Eugene Chin, Feng Yao, Qi Wang, Yingli Wu, Yadi Wu, Jing Du, Xue Li, Jianhua Xu, Hongjia Zhang, Siwei Zhang, Kaimi Li, Min Hu, Yanbin Kuang, Shuli Liu, Tong Wang, Hongyong Song, Yueling Liao, Jie Zhang, Beibei Sun, Wenzheng Guo, Dongliang Xu, Bo Jing, and Huijing Yin
- Abstract
While initiation is established as a critical step in tumorigenesis, the identity of the cell of origin for lung adenocarcinoma and the mechanism controlling susceptibility to initiation remain elusive. Here we show that lung tumor suppressor Gprc5a-knockout (KO) mice are susceptible to initiation of lung tumorigenesis. Bronchioalveolar stem cells (BASC) and alveolar type 2 (AT2) cells were aberrantly expanded in Gprc5a-KO mouse lungs compared with those in wild-type (WT) mice, suggesting that Gprc5a-KO might confer susceptibility to initiation by increasing the cell of origin in mouse lungs. BASCs from Gprc5a-KO mice (KO-BASC) exhibited significantly increased stemness and self-renewal potential and reduced differentiation capacity compared with BASCs from WT mice (WT-BASC). AT2 cells did not possess self-renewal potential regardless of Gprc5a status. KO-BASCs expressed a stem-like gene profile with upregulated Abcg2, EGFR, and NF-κB signaling compared with WT-BASCs. Blockade of EGFR and NF-κB signaling inhibited both expansion of BASC and AT2 cells and lung tumorigenesis. Abcg2 was expressed in active KO-BASCs as well as in lung tumor cells but not in quiescent WT-BASCs or AT2 cells, supporting that lung adenocarcinoma cells are derived from Abcg2-positive KO-BASCs (active). Taken together, Gprc5a deletion leads to expansion of active BASCs via dysregulated EGFR and NF-κB signaling that confers susceptibility to initiation of lung tumorigenesis, marking Abcg2-positive BASCs as candidate cell of origin for lung adenocarcinoma.Significance:Identification of active bronchioalveolar stem cells as lung adenocarcinoma cells of origin provides insights into mechanisms of lung tumorigenesis and could facilitate development of effective strategies for cancer prevention and therapy.See related commentary by Osborne and Minna, p. 972
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Data from IL6/STAT3 Signaling Orchestrates Premetastatic Niche Formation and Immunosuppressive Traits in Lung
- Author
-
Jiong Deng, Feng Yao, Binhua P. Zhou, Tuo Zhang, Yanbin Kuang, Jing Ling, Siwei Zhang, Min Hu, Kaimi Li, Wenzheng Guo, Hongyong Song, Yueling Liao, Dongliang Xu, Jianhua Xu, Beibei Sun, Tong Wang, and Bo Jing
- Abstract
Supplementary Fig. S1-S6, supplementary methods, supplementary table S1-S2. Supplementary Table S1. Primers used in the study; Supplementary Table S2. Antibodies information for flow cytometry analysis; Figure S1. IL6/ Gp130 pathway is involved in maintenance of stemness and in suppression of host immunity for lung metastasis; Figure S2. IL6 signaling is essential for lung metastasis in Gprc5a-ko mice lung tissues; Figure S3. IL6 signaling is essential for Lewis lung carcinoma cells in lung metastasis in Gprc5a-ko mice lung tissues; Figure S4. IL-6 signaling mediates the inhibition on innate immunity in Gprc5a-ko mouse lungs; Figure S5. IL-6 signaling orchestrates the inhibition on innate and adaptive immunity in Gprc5a-ko mouse lungs; Figure S6. Proposed model of IL-6/STAT3 signaling in lung metastasis
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure Legends 1-4 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 57K
- Published
- 2023
- Full Text
- View/download PDF
21. Supplemental Table S1 from Chaperone Hsp47 Drives Malignant Growth and Invasion by Modulating an ECM Gene Network
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Hanjiang Fu, Gaofeng Xiong, and Jieqing Zhu
- Abstract
Supplemental Table S1. Expression of Hsp47 and ECM protein genes is correlated in mammary cell lines.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Methods and References from Lung Tumor Suppressor GPRC5A Binds EGFR and Restrains Its Effector Signaling
- Author
-
Jiong Deng, Y. Eugene Chin, Binhua P. Zhou, Baohui Han, Wenzheng Guo, Xiaofeng Ye, Xiaofeng Lin, Beibei Sun, Chengyi Ding, Ruixu Yang, Zhi Pang, Lina Ma, Jingyi Liu, Yong Gao, Hongyong Song, Shuli Liu, Dongliang Xu, Yongxu Zhao, Huike Jiao, Jie Zhang, Qi Li, Feng Yao, Yueling Liao, Huijing Yin, and Shuangshuang Zhong
- Abstract
Supplementary Methods and References. Description of additional methods and procedures used in the study. Also includes Supplementary References.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Figure 1 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 1.3MB
- Published
- 2023
- Full Text
- View/download PDF
24. Supplemental Figure S4 from Chaperone Hsp47 Drives Malignant Growth and Invasion by Modulating an ECM Gene Network
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Hanjiang Fu, Gaofeng Xiong, and Jieqing Zhu
- Abstract
Supplemental Figure S4. Increased Hsp47 expression has been detected in ER negative cancer and is negatively associated with GATA3 levels.
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Figure 2 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 1.2MB
- Published
- 2023
- Full Text
- View/download PDF
26. Supplemental Table and Figure Legends from Chaperone Hsp47 Drives Malignant Growth and Invasion by Modulating an ECM Gene Network
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Hanjiang Fu, Gaofeng Xiong, and Jieqing Zhu
- Abstract
Supplemental Table and Figure Legends. Legends for Supplemental Table S1 and Supplemental Figures S1-S4.
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Table 2 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 53K
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Table 1 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 81K
- Published
- 2023
- Full Text
- View/download PDF
29. Supplementary Figures S1-S8 from Lung Tumor Suppressor GPRC5A Binds EGFR and Restrains Its Effector Signaling
- Author
-
Jiong Deng, Y. Eugene Chin, Binhua P. Zhou, Baohui Han, Wenzheng Guo, Xiaofeng Ye, Xiaofeng Lin, Beibei Sun, Chengyi Ding, Ruixu Yang, Zhi Pang, Lina Ma, Jingyi Liu, Yong Gao, Hongyong Song, Shuli Liu, Dongliang Xu, Yongxu Zhao, Huike Jiao, Jie Zhang, Qi Li, Feng Yao, Yueling Liao, Huijing Yin, and Shuangshuang Zhong
- Abstract
Supplementary Figures S1-S8. HEK293T cells were co-transfected with the plasmids of GPRC5A-myc, or myc-tagged GPRC5A mutants (∆C, ∆N, ∆7TM, and 7TM) as indicated ; Immunoblot was performed for p-EGFR (Y1068) and p-STAT3 (Y705) proteins from mouse lung tissues with the treatment as indicated ; IHC Staining for EGFR in the lung tissues of KO-NNK-12m (NNK-treated Gprc5a-/-mouse at age of 12 month) ; Representative pictures of IHC Staining for EGFR in two tumors (#282-1 and #282-2) identified in the same Gprc5a-/-mouse lung (#282) that was treated with NNK at age of 2 month and was sacrificed at age of 12 months (Gprc5a-ko-NNK-12m) ; Images of IHC Staining with normal rabbit IgG (upper) and anti-EGFR (bottom) in the tumor tissues (left) and the epithelium of small and terminal bronchioles (S/TB) of the lung from Gprc5a-ko mice treated with NNK at age of 12 months ; IHC Staining for p-STAT3 (Y705) in the lung tissues of KO-NNK-12m (NNK-treated Gprc5a-/- mouse at age of 12 month) ; Image of IHC Staining for Ki67 in the lung tissue of Gprc5a-ko-NNK-12m mouse which contains both the normal epithelium of small and terminal bronchioles (S/TB) (upper left) or tumor (bottom right) ; GPRC5A locus is located in the 12p12.3-p13 region which is frequently lost human NSCLC .
- Published
- 2023
- Full Text
- View/download PDF
30. Data from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
Inactivation of tumor suppressors and inhibitory microenvironmental factors is necessary for breast cancer invasion; therefore, identifying those suppressors and factors is crucial not only to advancing our knowledge of breast cancer, but also to discovering potential therapeutic targets. By analyzing gene expression profiles of polarized and disorganized human mammary epithelial cells in a physiologically relevant three-dimensional (3D) culture system, we identified retinoid orphan nuclear receptor alpha (RORα) as a transcription regulator of semaphorin 3F (SEMA3F), a suppressive microenvironmental factor. We showed that expression of RORα was downregulated in human breast cancer tissue and cell lines, and that reduced mRNA levels of RORα and SEMA3F correlated with poor prognosis. Restoring RORα expression reprogrammed breast cancer cells to form noninvasiveness structures in 3D culture and inhibited tumor growth in nude mice, accompanied by enhanced SEMA3F expression. Inactivation of RORα in nonmalignant human mammary epithelial cells inhibited SEMA3F transcription and impaired polarized acinar morphogenesis. Using chromatin immunoprecipitation and luciferase reporter assays, we showed that transcription of SEMA3F is directly regulated by RORα. Knockdown of SEMA3F in RORα-expressing cancer cells rescued the aggressive 3D phenotypes and tumor invasion. These findings indicate that RORα is a potential tumor suppressor and inhibits tumor invasion by inducing suppressive cell microenvironment. Cancer Res; 72(7); 1728–39. ©2012 AACR.
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Figure 3 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 2.3MB
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figure 4 from RORα Suppresses Breast Tumor Invasion by Inducing SEMA3F Expression
- Author
-
Ren Xu, Binhua P. Zhou, B. Mark Evers, Chi Wang, and Gaofeng Xiong
- Abstract
PDF file - 1730K
- Published
- 2023
- Full Text
- View/download PDF
33. Tumor Necrosis Factor Receptors: A Brief Digestion
- Author
-
Yiwei Lin, Zhibing Duan, and Binhua P. Zhou
- Published
- 2023
- Full Text
- View/download PDF
34. NFκB induces epigenetic repression of GPRC5A in lung epithelial cells to promote neoplasia
- Author
-
Hongyong, Song, Xiaofeng, Ye, Yueling, Liao, Siwei, Zhang, Dongliang, Xu, Shuangshuang, Zhong, Bo, Jing, Tong, Wang, Beibei, Sun, Jianhua, Xu, Wenzheng, Guo, Kaimi, Li, Min, Hu, Yanbin, Kuang, Jing, Ling, Tuo, Zhang, Yadi, Wu, Jing, Du, Feng, Yao, Yugene, Chin, Qi, Wang, Binhua P, Zhou, and Jiong, Deng
- Abstract
Chronic inflammation is associated with lung tumorigenesis, in which NF-κB-mediated epigenetic regulations play a critical role. Lung tumor suppressor GPRC5A is repressed in most non-small cell lung cancer (NSCLC), however the mechanisms remain unclear. Here, we show that NF-κB acts as a transcriptional repressor in suppression of GPRC5A. NF-κB induces GPRC5A repression both in vitro and in vivo. Intriguingly, trans-activation of NF-κB downstream targets is not required, but the trans-activation domain of RelA/p65 was required for GPRC5A repression. NF-κB did not bind to any potential cis-element in GPRC5A promoter. Instead, p65 was complexed with RARα/β, and recruited to the RA-response element (RARE) site at the GPRC5A promoter, resulting in disrupted RNA polymerase II complex, and suppressed transcription. Noticeably, phosphorylation on Serine276 of p65 is required for interaction with RARα/β and repression of GPRC5A. Moreover, NF-κB-mediated epigenetic repression is through suppression of histone H3K9ac, but not DNA methylation of the CpG islands, at the GPRC5A promoter. Consistently, a HDAC inhibitor, but not DNA methylation inhibitor, restored GPRC5A expression in NSCLC cells. Thus, NF-κB induces transcriptional repression of GPRC5A via complex with RARα/β and mediates epigenetic repression via suppression of H3K9ac.
- Published
- 2022
35. Dysregulated Glutamate Transporter SLC1A1 Propels Cystine Uptake via Xc− for Glutathione Synthesis in Lung Cancer
- Author
-
Kaimi Li, Qi Li, Hongyong Song, Binhua P. Zhou, Min Hu, Jing Ling, Feng Yao, Qi Wang, Shuli Liu, Beibei Sun, Wenzheng Guo, Jiong Deng, Yadi Wu, Tong Wang, Dongliang Xu, Lingfeng Tong, Shuhai Lin, Huijing Yin, Yanbin Kuang, Bo Jing, and Yueling Liao
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Chemistry ,SLC1A1 ,Glutamate receptor ,Cystine ,Glutathione ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Biosynthesis ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Extracellular ,Intracellular - Abstract
Cancer cells need to generate large amounts of glutathione (GSH) to buffer oxidative stress during tumor development. A rate-limiting step for GSH biosynthesis is cystine uptake via a cystine/glutamate antiporter Xc−. Xc− is a sodium-independent antiporter passively driven by concentration gradients from extracellular cystine and intracellular glutamate across the cell membrane. Increased uptake of cystine via Xc− in cancer cells increases the level of extracellular glutamate, which would subsequently restrain cystine uptake via Xc−. Cancer cells must therefore evolve a mechanism to overcome this negative feedback regulation. In this study, we report that glutamate transporters, in particular SLC1A1, are tightly intertwined with cystine uptake and GSH biosynthesis in lung cancer cells. Dysregulated SLC1A1, a sodium-dependent glutamate carrier, actively recycled extracellular glutamate into cells, which enhanced the efficiency of cystine uptake via Xc− and GSH biosynthesis as measured by stable isotope-assisted metabolomics. Conversely, depletion of glutamate transporter SLC1A1 increased extracellular glutamate, which inhibited cystine uptake, blocked GSH synthesis, and induced oxidative stress-mediated cell death or growth inhibition. Moreover, glutamate transporters were frequently upregulated in tissue samples of patients with non–small cell lung cancer. Taken together, active uptake of glutamate via SLC1A1 propels cystine uptake via Xc− for GSH biosynthesis in lung tumorigenesis. Significance: Cellular GSH in cancer cells is not only determined by upregulated Xc− but also by dysregulated glutamate transporters, which provide additional targets for therapeutic intervention.
- Published
- 2021
- Full Text
- View/download PDF
36. IL6/STAT3 Signaling Orchestrates Premetastatic Niche Formation and Immunosuppressive Traits in Lung
- Author
-
Jianhua Xu, Feng Yao, Siwei Zhang, Binhua P. Zhou, Min Hu, Tuo Zhang, Yanbin Kuang, Tong Wang, Bo Jing, Yueling Liao, Jing Ling, Jiong Deng, Beibei Sun, Wenzheng Guo, Dongliang Xu, Hongyong Song, and Kaimi Li
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Cancer Research ,Stromal cell ,Carcinogenesis ,Primary Cell Culture ,Mice, Nude ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,Receptors, G-Protein-Coupled ,Metastasis ,Carcinoma, Lewis Lung ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Tumor Microenvironment ,medicine ,Animals ,STAT3 ,Lung ,Mice, Knockout ,Tumor microenvironment ,biology ,Interleukin-6 ,Macrophages ,Myeloid-Derived Suppressor Cells ,Cellular Reprogramming ,medicine.disease ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Tumor Escape ,CD8 ,Signal Transduction - Abstract
Cancer cells that succeed in forming metastasis need to be reprogrammed to evade immune surveillance and survive in a new microenvironment. This is facilitated by metastatic niches that are either postformed through reciprocal signaling between tumor cells and local stromal cells or preformed as premetastatic niches before tumor cell arrival. IL6/STAT3 signaling is aberrantly activated in lung tumorigenesis and metastasis, however, the roles and mechanisms of action of IL6 remain controversial. Here, we showed that blockade of intrinsic STAT3 signaling in lung tumor cells suppressed lung metastasis in immune-competent syngeneic mice, but not in immune-deficient nude mice. Consistently, repression of STAT3 signaling in tumor cells made them susceptible to T-cell–mediated cytotoxicity. Thus, STAT3-mediated immunosuppression is crucial for metastasis. Noticeably, lung metastasis was greatly increased in Gprc5a-knockout (ko; 5a−/−) mice compared with wild-type mice, which correlated with upregulated IL6 in the tumor microenvironment. Depletion of IL6 via combined deletion of Il6 and Gprc5a genes almost completely eliminated lung metastasis in Gprc5a-ko/Il6-ko (5a−/−;Il6−/−) mice. Mechanistically, dysregulated IL6 reprogrammed the STAT3 pathway in metastatic tumor cells, and induced recruitment of myeloid-derived suppressor cells and polarized macrophages to evade host immunity. Consistently, IHC staining showed that activated STAT3 correlated with repressed infiltration of CD8+ T cells in non–small cell lung cancer. Therefore, IL6/STAT3 signaling is crucial for orchestrating premetastatic niche formation and immunosuppression in lung. Significance: IL6 plays important roles not only in cell autonomous propensity for metastasis, but also in establishing the metastatic niche.
- Published
- 2020
- Full Text
- View/download PDF
37. PTGES/PGE2 signaling links immunosuppression and lung metastasis in Gprc5a-knockout mouse model
- Author
-
Binhua P. Zhou, Yueling Liao, Tong Wang, Dongliang Xu, Yanbin Kuang, Bo Jing, Feng Yao, Kaimi Li, Siwei Zhang, Tuo Zhang, Beibei Sun, Jianhua Xu, Jing Ling, Hongyong Song, Wenzheng Guo, Shuli Liu, Jiong Deng, and Min Hu
- Subjects
0301 basic medicine ,Cancer Research ,Lung ,medicine.medical_treatment ,HEK 293 cells ,GPRC5A ,Immunosuppression ,Inflammation ,Biology ,medicine.disease_cause ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Knockout mouse ,Genetics ,medicine ,Cancer research ,medicine.symptom ,Carcinogenesis ,Molecular Biology - Abstract
Chronic inflammation has been linked to promotion of tumorigenesis and metastasis in lung. However, due to lack of a relevant animal model for characterization, the underlying mechanism remains elusive. Lung tumor suppressor gene Gprc5a-knockout (ko) mice are susceptible to lung inflammation, tumorigenesis and metastasis, which resembles the pathological features in human patients. Here, we showed that PTGES/PGE2 signaling was highly associated with lung tumorigenesis and metastasis in Gprc5a-ko mice. Interestingly, Ptges-knockout in mouse lung tumor cells, although reduced their stemness and EMT-like features, still formed tumors and lung metastasis in immune-deficient nude mice, but not in immune-competent mice. This suggests that the major role of PTGES/PGE2 signaling in tumorigenicity and lung metastasis is through immunosuppression. Mechanistically, PTGES/PGE2 signaling intrinsically endows tumor cells resistant to T-cell cytotoxicity, and induces cytokines extrinsically for MDSC recruitment, which is crucial for suppression of T-cell immunity. Importantly, targeting PGE2 signaling in Gprc5a-ko mice by PTGES inhibitor suppressed MDSC recruitment, restored T cells, and significantly repressed lung metastasis. Thus, PTGES/PGE2 signaling links immunosuppression and metastasis in an inflammatory lung microenvironment of Gprc5a-ko mouse model.
- Published
- 2020
- Full Text
- View/download PDF
38. Loss of GPRC5A Heterozygosity and Clinical Outcome in NSCLC
- Author
-
Jiong Deng, Beibei Sun, Wenzheng Guo, Yanbin Kuang, Song Hu, Jie Xing, Hongyong Song, Bingshun Wang, Dongliang Xu, Yueling Liao, Tong Wang, Bo Jing, Siwei Zhang, Shuli Liu, Min Hu, Kaimi Li, Xinyu Meng, Na Ni, Xueqian Sun, Tingting Liu, Mingming Tian, Eugene Y. Chin, Qi Wang, Yadi Wu, Feng Yao, Jing Du, Binhua P. Zhou, and Baohui Han
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
- Full Text
- View/download PDF
39. Deletion of tetraspanin CD151 alters the Wnt oncogene-induced mammary tumorigenesis: A cell type-linked function and signaling
- Author
-
Binhua P. Zhou, Olivier Thibault, Chi Wang, Sean E. Thatcher, Jinpeng Liu, Junfong Shi, Kuey Chen, Yadi Wu, Nevean Hamad, Dongping Wei, Jun Qiang, Hongxia Li, Hai Qian, Xinyu Deng, Rongbo Han, Xiuwei H. Yang, Jieming Li, Song Chen, Chunming Liu, Xiaowei Wei, Abigail McCaughley, Pao Xu, and Huanhuan Huang
- Subjects
0301 basic medicine ,Original article ,Cancer Research ,Cell type ,Mammary Neoplasms, Animal ,Wnt1 Protein ,Tetraspanin 24 ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cyclin D1 ,Tetraspanin ,Downregulation and upregulation ,Mammary tumor virus ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Oncogene ,Gene Expression Profiling ,Wnt signaling pathway ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunohistochemistry ,Wnt Proteins ,Cell Transformation, Neoplastic ,030104 developmental biology ,Mammary Tumor Virus, Mouse ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Carcinogenesis ,Biomarkers ,Gene Deletion ,Signal Transduction - Abstract
Tetraspanin CD151 is increasingly implicated as a multifaceted mediator of cancer development and progression. Here we investigated the role of CD151 in breast cancer in the context of the Wnt oncogenic activation. Our data showed that removal of one or both of CD151 alleles in the MMTV-Wnt1 model significantly decreased the tumor-free survival of mice from 34 weeks on average to 22 weeks and 18 weeks, respectively. This effect coincided with an accelerated tumor growth and an increased number of Ki-67+ proliferative cells. Mechanistically, the CD151-deficient tumors were largely ER+, and exhibited hyperactivation of the Wnt pathway as reflected by a marked upregulation in β-catenin and Cyclin D1, and their target genes. In addition, E-cadherin displayed a cytosolic distribution and transcription factor Snail was markedly upregulated. Collectively, this data implies that CD151 suppresses the Wnt1-driven tumorigenesis, at least in part, via counteracting the epithelial-mesenchymal transition (EMT)-like program in luminal epithelial cells. Meanwhile, the proportion of tumor cells expressing CK5 or p63, the biomarkers of myoepithelial/basal cells, markedly decreased in the absence of CD151. This change was accompanied by a decreased invasiveness of tumors and their incompetence to form a long-term cell culture. Consistent with this basal cell-linked role, the CD151 downregulation impairs mammosphere formation in MCF-10A cells and the defect was rescued by re-expression of intact CD151 ORF, but not its integrin binding-defective mutant. Overall, our study suggests that CD151 is a key player in the Wnt oncogene-driven tumorigenesis and impacts breast cancer malignancy in a cell type-dependent manner.
- Published
- 2019
- Full Text
- View/download PDF
40. Guidelines and definitions for research on epithelial–mesenchymal transition
- Author
-
Kyra Campbell, Ruby Yun-Ju Huang, Binhua P. Zhou, Yoshiko Takahashi, Yibin Kang, Cédric Blanpain, Antonio García de Herreros, Ben Z. Stanger, Amparo Cano, Gerhard Christofori, Jordi Casanova, Jean Paul Thiery, Jing Yang, Chaya Kalcheim, Geert Berx, Raghu Kalluri, Sendurai A. Mani, Marc P. Stemmler, Yeesim Khew-Goodall, Marianne E. Bronner, Thomas Brabletz, Heide L. Ford, Masatoshi Takeichi, Jonas Fuxe, Rik Derynck, Anna-Katerina Hadjantonakis, Shoukat Dedhar, Parker B. Antin, M. Angela Nieto, Donald F. Newgreen, Herbert Levine, Raymond B. Runyan, Guojun Sheng, Robert A. Weinberg, Gregory J. Goodall, Keith E. Mostov, Erik W. Thompson, Roberto Mayor, Eric Theveneau, Pierre Savagner, Alain Puisieux, Jinsong Liu, Joan Massagué, Elizabeth D. Williams, Gregory D. Longmore, Jianhua Xing, David R. McClay, Yang, Jing, Antin, Parker, Berx, Geert, Blanpain, Cédric, Goodall, Gregory J, Khew-Goodall, Yeesim, Sheng, Guojun, EMT International Association (TEMTIA), UAM. Departamento de Bioquímica, and Instituto de Investigaciones Biomédicas 'Alberto Sols' (IIBM)
- Subjects
Web of science ,cell migration ,Medicina ,Lung metastasis ,Diversity in experimental ,Regulació cel·lular -- Guies ,Definitions ,Guidelines ,Cellular organization ,Terminology ,EMBRYONIC HEART ,03 medical and health sciences ,0302 clinical medicine ,E-CADHERIN EXPRESSION ,Developmental biology ,Medicine and Health Sciences ,Epithelial–mesenchymal transition ,PLASTICITY ,epithelial–mesenchymal transition (EMT) ,Molecular Biology ,TRANSCRIPTION FACTOR SNAIL ,030304 developmental biology ,Research data ,FIBROBLAST-GROWTH-FACTOR ,Epithelial–mesenchymal transition (EMT) ,Cancer ,Cognitive science ,0303 health sciences ,Cèl·lules epitelials ,Nomenclature ,EMT ,Consensus Statement ,Biology and Life Sciences ,Biologie moléculaire ,Cell movement ,Cell Biology ,COLLAGEN GELS ,Epithelial-mesenchymal transition ,3. Good health ,Conceptual framework ,NEURAL CREST CELLS ,030220 oncology & carcinogenesis ,REPRESSES E-CADHERIN ,Consensus statement ,embryonic structures ,LUNG METASTASIS ,Biologie cellulaire ,Psychology - Abstract
On behalf of the EMT International Association (TEMTIA)., Epithelial–mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by ‘the EMT International Association’ (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.
- Published
- 2020
41. BRD4 modulates vulnerability of triple-negative breast cancer to targeting of integrin-dependent signaling pathways
- Author
-
Xucai Zheng, Xinlan Lu, Hai Qian, Olivier Thibault, Binhua P. Zhou, Chi Wang, Li Chen, Shuixiang He, Yang Zhang, Dongping Wei, Nevean Hamad, Chunming Liu, Rolf J. Craven, Jian-An Huang, Li Xu, Yadi Wu, Yueyin Pan, Jieming Li, Xiuwei H. Yang, Junfeng Shi, Helen Yang, Dana Napier, Zeyi Liu, Xiaowei Wei, and Bingwei Xu
- Subjects
0301 basic medicine ,Cancer Research ,Integrins ,Cell Survival ,medicine.medical_treatment ,Cell Cycle Proteins ,Triple Negative Breast Neoplasms ,Integrin ,Biology ,Targeted therapy ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,Triple-negative breast cancer ,Cell Line, Tumor ,medicine ,Humans ,RNA, Messenger ,Transcription factor ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Sulfonamides ,Original Paper ,Bcl-2-Like Protein 11 ,Cell Death ,FAK ,Kinase ,Genome, Human ,General Medicine ,Azepines ,Triazoles ,XIAP ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,c-Myc ,Oncology ,030220 oncology & carcinogenesis ,Focal Adhesion Protein-Tyrosine Kinases ,Pyrazines ,Benzamides ,Cancer research ,Molecular Medicine ,BRD4 ,Signal transduction ,Signal Transduction ,Transcription Factors - Abstract
Purpose Stemming from a myriad of genetic and epigenetic alterations, triple-negative breast cancer (TNBC) is tied to poor clinical outcomes and aspires for individualized therapies. Here we investigated the therapeutic potential of co-inhibiting integrin-dependent signaling pathway and BRD4, a transcriptional and epigenetic mediator, for TNBC. Methods Two independent patient cohorts were subjected to bioinformatic and IHC examination for clinical association of candidate cancer drivers. The efficacy and biological bases for co-targeting these drivers were interrogated using cancer cell lines, a protein kinase array, chemical inhibitors, RNAi/CRISPR/Cas9 approaches, and a 4 T1-Balb/c xenograft model. Results We found that amplification of the chromosome 8q24 region occurred in nearly 20% of TNBC tumors, and that it coincided with co-upregulation or amplification of c-Myc and FAK, a key effector of integrin-dependent signaling. This co-upregulation at the mRNA or protein level correlated with a poor patient survival (p p + myeloid-derived suppressor cells in vivo. Finally, we found that JQ1 and VS-6063 cooperatively induced apoptotic cell death by altering XIAP, Bcl2/Bcl-xl and Bim levels, impairing c-Src/p130Cas-, PI3K/Akt- and RelA-associated signaling, and were linked to EMT-inducing transcription factor Snail- and Slug-dependent regulation. Conclusion Based on our results, we conclude that the BRD4/c-Myc- and integrin/FAK-dependent pathways act in concert to promote breast cancer cell survival and poor clinical outcomes. As such, they represent promising targets for a synthetic lethal-type of therapy against TNBC.
- Published
- 2020
42. Dysregulated Glutamate Transporter SLC1A1 Propels Cystine Uptake via Xc
- Author
-
Wenzheng, Guo, Kaimi, Li, Beibei, Sun, Dongliang, Xu, Lingfeng, Tong, Huijing, Yin, Yueling, Liao, Hongyong, Song, Tong, Wang, Bo, Jing, Min, Hu, Shuli, Liu, Yanbin, Kuang, Jing, Ling, Qi, Li, Yadi, Wu, Qi, Wang, Feng, Yao, Binhua P, Zhou, Shu-Hai, Lin, and Jiong, Deng
- Subjects
Lung Neoplasms ,Cell Death ,Glutamine ,Glutamic Acid ,Mice, Nude ,Glutathione ,Antiporters ,Receptors, G-Protein-Coupled ,Up-Regulation ,Mice ,Oxidative Stress ,Excitatory Amino Acid Transporter 3 ,Stress, Physiological ,Cell Line, Tumor ,Animals ,Cystine - Abstract
Cancer cells need to generate large amounts of glutathione (GSH) to buffer oxidative stress during tumor development. A rate-limiting step for GSH biosynthesis is cystine uptake via a cystine/glutamate antiporter Xc
- Published
- 2020
43. Hsp47 promotes cancer metastasis by enhancing collagen-dependent cancer cell-platelet interaction
- Author
-
Jieqing Zhu, Shike Wang, Kunito Kawasaki, Zhenyu Li, Guoying Zhang, Yan Zhang, Ren Xu, Gaofeng Xiong, Binhua P. Zhou, Kazuhiro Nagata, and Jie Chen
- Subjects
Blood Platelets ,animal structures ,Epithelial-Mesenchymal Transition ,Medical Sciences ,extracellular matrix ,Breast Neoplasms ,Mice, SCID ,circulating tumor cell ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Circulating tumor cell ,breast cancer ,cancer metastasis ,Animals ,Humans ,Epithelial–mesenchymal transition ,Neoplasm Metastasis ,HSP47 Heat-Shock Proteins ,030304 developmental biology ,Heat shock protein 47 ,0303 health sciences ,Multidisciplinary ,biology ,Chemistry ,Mesenchymal stem cell ,Gene Amplification ,Biological Sciences ,Neoplastic Cells, Circulating ,Extravasation ,3. Good health ,030220 oncology & carcinogenesis ,Cancer cell ,embryonic structures ,Cancer research ,biology.protein ,Female ,Collagen ,Type I collagen - Abstract
Significance Cancer cell–platelet interaction is crucial for cancer metastasis; however, how this interaction is regulated remains largely unknown. We have identified Hsp47 as an EMT inducer and showed that Hsp47 and its dependent collagen secretion enhanced cancer cell–platelet interaction. Importantly, Hsp47-induced cancer cell–platelet interaction enhanced cancer cell clustering, which is crucial for cancer cell colonization at distant sites. We also found that Hsp47 gene amplification and expression were associated with breast cancer metastasis. These results reveal a link between the Hsp47/collagen axis and platelet recruitment, and provide additional insight into how mesenchymal phenotypes in cancer cells contribute to breast cancer metastasis., Increased expression of extracellular matrix (ECM) proteins in circulating tumor cells (CTCs) suggests potential function of cancer cell-produced ECM in initiation of cancer cell colonization. Here, we showed that collagen and heat shock protein 47 (Hsp47), a chaperone facilitating collagen secretion and deposition, were highly expressed during the epithelial-mesenchymal transition (EMT) and in CTCs. Hsp47 expression induced mesenchymal phenotypes in mammary epithelial cells (MECs), enhanced platelet recruitment, and promoted lung retention and colonization of cancer cells. Platelet depletion in vivo abolished Hsp47-induced cancer cell retention in the lung, suggesting that Hsp47 promotes cancer cell colonization by enhancing cancer cell–platelet interaction. Using rescue experiments and functional blocking antibodies, we identified type I collagen as the key mediator of Hsp47-induced cancer cell–platelet interaction. We also found that Hsp47-dependent collagen deposition and platelet recruitment facilitated cancer cell clustering and extravasation in vitro. By analyzing DNA/RNA sequencing data generated from human breast cancer tissues, we showed that gene amplification and increased expression of Hsp47 were associated with cancer metastasis. These results suggest that targeting the Hsp47/collagen axis is a promising strategy to block cancer cell–platelet interaction and cancer colonization in secondary organs.
- Published
- 2020
44. ITCH nuclear translocation and H1.2 polyubiquitination negatively regulate the DNA damage response
- Author
-
Binghui Shen, Jing Gao, Hangyi Pan, Li Zheng, Brian Armstrong, Yang Peng, Lufen Chang, Steven T. Rosen, Hu Zhou, Lei Shen, Binhua P Zhou, and Guang Peng
- Subjects
DNA Replication ,DNA damage ,Ubiquitin-Protein Ligases ,Active Transport, Cell Nucleus ,Breast Neoplasms ,Histones ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Downregulation and upregulation ,Cell Line, Tumor ,parasitic diseases ,Serine ,otorhinolaryngologic diseases ,Genetics ,Animals ,Humans ,skin and connective tissue diseases ,Molecular Biology ,Protein kinase B ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,biology ,Cell growth ,Ubiquitination ,DNA replication ,Ubiquitin ligase ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,HEK293 Cells ,Cancer cell ,biology.protein ,Female ,Tumor Suppressor p53-Binding Protein 1 ,Proto-Oncogene Proteins c-akt ,030217 neurology & neurosurgery ,DNA Damage - Abstract
The downregulation of the DNA damage response (DDR) enables aggressive tumors to achieve uncontrolled proliferation against replication stress, but the mechanisms underlying this process in tumors are relatively complex. Here, we demonstrate a mechanism through which a distinct E3 ubiquitin ligase, ITCH, modulates DDR machinery in triple-negative breast cancer (TNBC). We found that expression of a nuclear form of ITCH was significantly increased in human TNBC cell lines and tumor specimens. Phosphorylation of ITCH at Ser257 by AKT led to the nuclear localization of ITCH and ubiquitination of H1.2. The ITCH-mediated polyubiquitination of H1.2 suppressed RNF8/RNF168-dependent formation of 53BP1 foci, which plays important roles in DDR. Consistent with these findings, impaired ITCH nuclear translocation and H1.2 polyubiquitination sensitized cells to replication stress and limited cell growth and migration. AKT activation of ITCH-H1.2 axis may confer TNBC cells with a DDR repression to counteract the replication stress and increase cancer cell survivorship and growth potential.
- Published
- 2018
- Full Text
- View/download PDF
45. Targeting the BRD4/FOXO3a/CDK6 axis sensitizes AKT inhibition in luminal breast cancer
- Author
-
Yanling He, Lei Zeng, Zhibing Duan, Qiang Zhang, Fang Tai, Yifan Wang, Saghi Ghaffari, Pengnian Charles Lin, Yule Chen, Yiwei Lin, Jingyi Liu, B. Mark Evers, Yadi Wu, Suling Liu, Binhua P. Zhou, Weijie Guo, Chi Wang, Jiong Deng, Jian Shi, Rachel L. Stewart, and Ming-Ming Zhou
- Subjects
0301 basic medicine ,General Physics and Astronomy ,Cell Cycle Proteins ,Mice ,0302 clinical medicine ,Transcription (biology) ,Sirtuins ,Promoter Regions, Genetic ,lcsh:Science ,Regulation of gene expression ,Oxadiazoles ,Multidisciplinary ,Forkhead Box Protein O3 ,Nuclear Proteins ,Acetylation ,3. Good health ,Protein Transport ,030220 oncology & carcinogenesis ,Female ,Heterocyclic Compounds, 3-Ring ,Protein Binding ,SIRT6 ,BRD4 ,Science ,Mice, Nude ,Breast Neoplasms ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Breast cancer ,Protein Domains ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Pyrroles ,Protein Kinase Inhibitors ,Protein kinase B ,Cell Nucleus ,Promoter ,Cyclin-Dependent Kinase 6 ,General Chemistry ,medicine.disease ,Pyrimidines ,030104 developmental biology ,Gene Expression Regulation ,Cancer research ,biology.protein ,lcsh:Q ,Cyclin-dependent kinase 6 ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
BRD4 assembles transcriptional machinery at gene super-enhancer regions and governs the expression of genes that are critical for cancer progression. However, it remains unclear whether BRD4-mediated gene transcription is required for tumor cells to develop drug resistance. Our data show that prolonged treatment of luminal breast cancer cells with AKT inhibitors induces FOXO3a dephosphorylation, nuclear translocation, and disrupts its association with SirT6, eventually leading to FOXO3a acetylation as well as BRD4 recognition. Acetylated FOXO3a recognizes the BD2 domain of BRD4, recruits the BRD4/RNAPII complex to the CDK6 gene promoter, and induces its transcription. Pharmacological inhibition of either BRD4/FOXO3a association or CDK6 significantly overcomes the resistance of luminal breast cancer cells to AKT inhibitors in vitro and in vivo. Our study reports the involvement of BRD4/FOXO3a/CDK6 axis in AKTi resistance and provides potential therapeutic strategies for treating resistant breast cancer., The molecular mechanism underlying the resistance of AKT inhibitors in breast cancer is still elusive. Here, the authors demonstrate that BRD4/FOXO3a axis upregulates CDK6 promoter activity to promote resistance to AKT inhibition in breast cancer cells and that blocking the action of CDK6 re-sensitizes resistant cancer cells to growth inhibition.
- Published
- 2018
- Full Text
- View/download PDF
46. Twist promotes tumor metastasis in basal-like breast cancer by transcriptionally upregulating ROR1
- Author
-
Renxian Cao, Yuanzhong Wu, Xin Wang, Ruhua Zhang, Meifang Zhang, Jingying Cao, Binhua P. Zhou, Tao Dai, Jian Shi, Gang Wang, Rou Jiang, and Tiebang Kang
- Subjects
0301 basic medicine ,tumor metastasis ,Wnt signaling pathway ,EMT ,Medicine (miscellaneous) ,Cell migration ,basal-like breast cancer (BLBC) ,Biology ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,Breast cancer ,Cancer cell ,ROR1 ,medicine ,Cancer research ,Gene silencing ,Twist ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Transcription factor ,Research Paper - Abstract
Rationale: Twist is a key transcription factor for induction of epithelial-mesenchymal transition (EMT), which promotes cell migration, invasion, and cancer metastasis, confers cancer cells with stem cell-like characteristics, and provides therapeutic resistance. However, the functional roles and targeted genes of Twist in EMT and cancer progression remain elusive. Methods: The potential targeted genes of Twist were identified from the global transcriptomes of T47D/Twist cells by microarray analysis. EMT phenotype was detected by western blotting and immunofluorescence of marker proteins. The dual-luciferase reporter and chromatin immunoprecipitation assays were employed to observe the direct transcriptional induction of ROR1 by Twist. A lung metastasis model was used to study the pro-metastatic role of Twist and ROR1 by injecting MDA-MB-231 cells into tail vein of nude mice. Bio-informatics analysis was utilized to measure the metastasis-free survival of breast cancer patients. Results: Twist protein was proved to directly activate the transcription of ROR1 gene, a receptor of Wnt5a in non-canonical WNT signaling pathway. Silencing of ROR1 inhibited EMT process, cell migration, invasion, and cancer metastasis of basal-like breast cancer (BLBC) cells. Knockdown of ROR1 also ameliorated the pro-metastatic effect of Twist. Furthermore, analyses of clinical specimens indicated that high expression of both ROR1 and Twist tightly correlates with poor metastasis-free survival of breast cancer patients. Conclusion: ROR1 is a targeted gene of Twist. Twist/ROR1 signaling is critical for invasion and metastasis of BLBC cells.
- Published
- 2018
47. Author Correction: Guidelines and definitions for research on epithelial–mesenchymal transition
- Author
-
Jing Yang, Geert Berx, Elizabeth D. Williams, Sendurai A. Mani, Gregory J. Goodall, David R. McClay, Binhua P. Zhou, Parker B. Antin, Raghu Kalluri, Pierre Savagner, Erik W. Thompson, Roberto Mayor, Amparo Cano, Jinsong Liu, Robert A. Weinberg, Masatoshi Takeichi, Thomas Brabletz, Chaya Kalcheim, Donald F. Newgreen, Shoukat Dedhar, Kyra Campbell, Herbert Levine, Raymond B. Runyan, Jordi Casanova, Rik Derynck, Anna-Katerina Hadjantonakis, M. Angela Nieto, Marc P. Stemmler, Yeesim Khew-Goodall, Jean Paul Thiery, Heide L. Ford, Marianne E. Bronner, Yibin Kang, Antonio García de Herreros, Ben Z. Stanger, Cédric Blanpain, Keith E. Mostov, Jonas Fuxe, Eric Theveneau, Yoshiko Takahashi, Alain Puisieux, Ruby Yun-Ju Huang, Gerhard Christofori, Guojun Sheng, Joan Massagué, Gregory D. Longmore, and Jianhua Xing
- Subjects
Biomedical Research ,Consensus ,Epithelial-Mesenchymal Transition ,business.industry ,Published Erratum ,Cell Plasticity ,MEDLINE ,Cell Biology ,Cell Movement ,Neoplasms ,Terminology as Topic ,Cancer research ,Animals ,Humans ,Medicine ,Epithelial–mesenchymal transition ,Author Correction ,business ,Molecular Biology ,Developmental Biology - Abstract
Epithelial-mesenchymal transition (EMT) encompasses dynamic changes in cellular organization from epithelial to mesenchymal phenotypes, which leads to functional changes in cell migration and invasion. EMT occurs in a diverse range of physiological and pathological conditions and is driven by a conserved set of inducing signals, transcriptional regulators and downstream effectors. With over 5,700 publications indexed by Web of Science in 2019 alone, research on EMT is expanding rapidly. This growing interest warrants the need for a consensus among researchers when referring to and undertaking research on EMT. This Consensus Statement, mediated by 'the EMT International Association' (TEMTIA), is the outcome of a 2-year-long discussion among EMT researchers and aims to both clarify the nomenclature and provide definitions and guidelines for EMT research in future publications. We trust that these guidelines will help to reduce misunderstanding and misinterpretation of research data generated in various experimental models and to promote cross-disciplinary collaboration to identify and address key open questions in this research field. While recognizing the importance of maintaining diversity in experimental approaches and conceptual frameworks, we emphasize that lasting contributions of EMT research to increasing our understanding of developmental processes and combatting cancer and other diseases depend on the adoption of a unified terminology to describe EMT.
- Published
- 2021
- Full Text
- View/download PDF
48. Snail determines the therapeutic response to mTOR kinase inhibitors by transcriptional repression of 4E-BP1
- Author
-
Binhua P. Zhou, Chi Wang, Jun Wang, Side Liu, Wenting Mi, Yubin Guo, B. Mark Evers, Qing-Bai She, Xiuping Huang, Hsin-Sheng Yang, Qing Ye, and Yanan Cao
- Subjects
0301 basic medicine ,Male ,General Physics and Astronomy ,Cell Cycle Proteins ,Snail ,medicine.disease_cause ,environment and public health ,Madin Darby Canine Kidney Cells ,Mice ,Neoplasms ,lcsh:Science ,Multidisciplinary ,biology ,Kinase ,TOR Serine-Threonine Kinases ,3. Good health ,Gene Expression Regulation, Neoplastic ,MCF-7 Cells ,biological phenomena, cell phenomena, and immunity ,Science ,Mice, Nude ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,Dogs ,Downregulation and upregulation ,biology.animal ,Cell Line, Tumor ,parasitic diseases ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Cell growth ,fungi ,General Chemistry ,HCT116 Cells ,Phosphoproteins ,Xenograft Model Antitumor Assays ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,A549 Cells ,Cancer cell ,Cancer research ,NIH 3T3 Cells ,Ectopic expression ,lcsh:Q ,Snail Family Transcription Factors ,Carcinogenesis - Abstract
Loss of 4E-BP1 expression has been linked to cancer progression and resistance to mTOR inhibitors, but the mechanism underlying 4E-BP1 downregulation in tumors remains unclear. Here we identify Snail as a strong transcriptional repressor of 4E-BP1. We find that 4E-BP1 expression inversely correlates with Snail level in cancer cell lines and clinical specimens. Snail binds to three E-boxes present in the human 4E-BP1 promoter to repress transcription of 4E-BP1. Ectopic expression of Snail in cancer cell lines lacking Snail profoundly represses 4E-BP1 expression, promotes cap-dependent translation in polysomes, and reduces the anti-proliferative effect of mTOR kinase inhibitors. Conversely, genetic and pharmacological inhibition of Snail function restores 4E-BP1 expression and sensitizes cancer cells to mTOR kinase inhibitors by enhancing 4E-BP1-mediated translation-repressive effect on cell proliferation and tumor growth. Our study reveals a critical Snail-4E-BP1 signaling axis in tumorigenesis, and provides a rationale for targeting Snail to improve mTOR-targeted therapies., 4E-BP1 is a translational repressor critical in mTOR signaling, whereas Snail is a critical promoter of epithelial to mesenchymal transition. Here the authors show that Snail induces resistance to mTOR inhibitors by repressing 4E-BP1 expression and promoting cell cycle progression via upregulating cycD.
- Published
- 2017
- Full Text
- View/download PDF
49. Nanoparticle Delivery of miR-34a Eradicates Long-term-cultured Breast Cancer Stem Cells via Targeting C22ORF28 Directly
- Author
-
Xiaoti Lin, Xiaoming Xie, Fengqin Wei, Binhua P. Zhou, Weiyu Chen, and Mien Chie Hung
- Subjects
0301 basic medicine ,cancer stem cells ,C22ORF28 ,Medicine (miscellaneous) ,Mice, Nude ,therapeutic target ,Antineoplastic Agents ,Breast Neoplasms ,Docetaxel ,targeted drug delivery ,Metastasis ,03 medical and health sciences ,Mice ,Breast cancer ,breast cancer ,Cancer stem cell ,microRNA ,Medicine ,Animals ,Humans ,Telomerase reverse transcriptase ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Mice, Inbred BALB C ,business.industry ,Cell sorting ,medicine.disease ,MicroRNAs ,030104 developmental biology ,RNAi Therapeutics ,Cancer research ,MCF-7 Cells ,Neoplastic Stem Cells ,Nanoparticles ,Female ,Taxoids ,Stem cell ,business ,miR-34a ,medicine.drug ,Research Paper - Abstract
Rationale: Cancer stem cells (CSCs) have been implicated as the seeds of therapeutic resistance and metastasis, due to their unique abilities of self-renew, wide differentiation potentials and resistance to most conventional therapies. It is a proactive strategy for cancer therapy to eradicate CSCs. Methods: Tumor tissue-derived breast CSCs (BCSC), including XM322 and XM607, were isolated by fluorescence-activated cell sorting (FACS); while cell line-derived BCSC, including MDA-MB-231.SC and MCF-7.SC, were purified by magnetic-activated cell sorting (MACS). Analyses of microRNA and mRNA expression array profiles were performed in multiple breast cell lines. The mentioned nanoparticles were constructed following the standard molecular cloning protocol. Tissue microarray analysis has been used to study 217 cases of clinical breast cancer specimens. Results: Here, we have successfully established four long-term maintenance BCSC that retain their tumor-initiating biological properties. Our analyses of microarray and qRT-PCR explored that miR-34a is the most pronounced microRNA for investigation of BCSC. We establish hTERT promoter-driven VISA delivery of miR-34a (TV-miR-34a) plasmid that can induce high throughput of miR-34a expression in BCSC. TV-miR-34a significantly inhibited the tumor-initiating properties of long-term-cultured BCSC in vitro and reduced the proliferation of BCSC in vivo by an efficient and safe way. TV-miR-34a synergizes with docetaxel, a standard therapy for invasive breast cancer, to act as a BCSC inhibitor. Further mechanistic investigation indicates that TV-miR-34a directly prevents C22ORF28 accumulation, which abrogates clonogenicity and tumor growth and correlates with low miR-34 and high C22ORF28 levels in breast cancer patients. Conclusion: Taken together, we generated four long-term maintenance BCSC derived from either clinical specimens or cell lines, which would be greatly beneficial to the research progress in breast cancer patients. We further developed the non-viral TV-miR-34a plasmid, which has a great potential to be applied as a clinical application for breast cancer therapy.
- Published
- 2017
50. Stabilization of the transcription factors slug and twist by the deubiquitinase dub3 is a key requirement for tumor metastasis
- Author
-
Jing Feng, Qing Shi, Yadi Wu, Yu Wang, Yiwei Lin, Jiong Deng, Qian Yu, Binhua P. Zhou, B. Mark Evers, and Cuicui Liu
- Subjects
0301 basic medicine ,animal structures ,Slug ,Cellular differentiation ,Biology ,Deubiquitinating enzyme ,Metastasis ,03 medical and health sciences ,Cancer stem cell ,medicine ,metastasis ,Twist ,Transcription factor ,IL-6 ,fungi ,Cancer ,biology.organism_classification ,medicine.disease ,3. Good health ,030104 developmental biology ,Oncology ,embryonic structures ,Cancer cell ,biology.protein ,Cancer research ,Dub3 ,Research Paper - Abstract
// Yiwei Lin 1, 4, * , Yu Wang 2, 4, * , Qing Shi 1, 4 , Qian Yu 2, 4 , Cuicui Liu 1, 4, 5 , Jing Feng 5 , Jiong Deng 6 , B. Mark Evers 3, 4 , Binhua P. Zhou 1, 4 and Yadi Wu 2, 4 1 Department of Molecular and Cellular Biochemistry, Lexington, KY, USA 2 Department of Pharmacology & Nutritional Sciences, Lexington, KY, USA 3 Department of Surgery, Lexington, KY, USA 4 Markey Cancer Center, the University of Kentucky, College of Medicine, Lexington, KY, USA 5 Department of Laboratory Medicine & Central Laboratory, Shanghai Fengxian District Central Hospital, Shanghai, China 6 Key Laboratory of Cell Differentiation and Apoptosis of Chinese Minister of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China * Authors contributed equally to this work Correspondence to: Binhua P. Zhou, email: peter.zhou@uky.edu Yadi Wu, email: yadi.wu@uky.edu Keywords: Dub3, Slug, Twist, IL-6, metastasis Received: July 18, 2017 Accepted: August 09, 2017 Published: August 24, 2017 ABSTRACT The Epithelial-mesenchymal transition (EMT) represents a cellular de-differentiation process that provides cells with the increased plasticity required during embryonic development, tissue remodeling, wound healing and metastasis. Slug and Twist are two key EMT transcription factors (EMT-TFs) that are tightly regulated via ubiquitination and degradation. How Slug and Twist escape degradation and become stabilized in cancer cells remains unclear. One plausible mechanism of Slug and Twist stabilization involves removal of ubiquitin by deubiquitinases (DUBs). In this study, we identified Dub3 as a novel DUB for both Slug and Twist. We further found that Dub3 overexpression increased Slug and Twist protein levels in a dose-dependent manner, whereas Dub3-knockdown decreased their protein levels. Of importance, Dub3 interacted with Slug and Twist and prevented them from degradation, thereby promoting migration, invasion, and cancer stem cell (CSC)-like properties of breast cancer cells. Intriguingly, Dub3 was identified as an early response gene that was upregulated after exposure to inflammatory cytokines such as IL-6, which plays a critical role in the growth and metastasis of breast cancer cells, as well as the maintenance of breast CSCs. We found that Dub3 played an essential role in IL-6 induced EMT through stabilization of Slug and Twist. Our study has uncovered an IL-6-Dub3-Slug/Twist signaling axis during EMT and suggests potential approaches that could target Dub3 to prevent metastatic breast tumor.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.