21 results on '"Yunlu Jia"'
Search Results
2. Super-enhancer-driven TOX2 mediates oncogenesis in Natural Killer/T Cell Lymphoma
- Author
-
Jianbiao Zhou, Sabrina Hui-Min Toh, Tze King Tan, Kalpnaa Balan, Jing Quan Lim, Tuan Zea Tan, Sinan Xiong, Yunlu Jia, Siok-Bian Ng, Yanfen Peng, Anand D. Jeyasekharan, Shuangyi Fan, Soon Thye Lim, Chin-Ann Johnny Ong, Choon Kiat Ong, Takaomi Sanda, and Wee-Joo Chng
- Subjects
Cancer Research ,Oncology ,Molecular Medicine - Abstract
Background Extranodal natural killer/T-cell lymphoma (NKTL) is an aggressive type of non-Hodgkin lymphoma with dismal outcome. A better understanding of disease biology and key oncogenic process is necessary for the development of targeted therapy. Super-enhancers (SEs) have been shown to drive pivotal oncogenes in various malignancies. However, the landscape of SEs and SE-associated oncogenes remain elusive in NKTL. Methods We used Nano-ChIP-seq of the active enhancer marker histone H3 lysine 27 acetylation (H3K27ac) to profile unique SEs NKTL primary tumor samples. Integrative analysis of RNA-seq and survival data further pinned down high value, novel SE oncogenes. We utilized shRNA knockdown, CRISPR-dCas9, luciferase reporter assay, ChIP-PCR to investigate the regulation of transcription factor (TF) on SE oncogenes. Multi-color immunofluorescence (mIF) staining was performed on an independent cohort of clinical samples. Various function experiments were performed to evaluate the effects of TOX2 on the malignancy of NKTL in vitro and in vivo. Results SE landscape was substantially different in NKTL samples in comparison with normal tonsils. Several SEs at key transcriptional factor (TF) genes, including TOX2, TBX21(T-bet), EOMES, RUNX2, and ID2, were identified. We confirmed that TOX2 was aberrantly overexpressed in NKTL relative to normal NK cells and high expression of TOX2 was associated with worse survival. Modulation of TOX2 expression by shRNA, CRISPR-dCas9 interference of SE function impacted on cell proliferation, survival and colony formation ability of NKTL cells. Mechanistically, we found that RUNX3 regulates TOX2 transcription by binding to the active elements of its SE. Silencing TOX2 also impaired tumor formation of NKTL cells in vivo. Metastasis-associated phosphatase PRL-3 has been identified and validated as a key downstream effector of TOX2-mediated oncogenesis. Conclusions Our integrative SE profiling strategy revealed the landscape of SEs, novel targets and insights into molecular pathogenesis of NKTL. The RUNX3-TOX2-SE-TOX2-PRL-3 regulatory pathway may represent a hallmark of NKTL biology. Targeting TOX2 could be a valuable therapeutic intervene for NKTL patients and warrants further study in clinic.
- Published
- 2023
- Full Text
- View/download PDF
3. Super Enhancer-Mediated Upregulation of HJURP Promotes Growth and Survival of t(4;14)-Positive Multiple Myeloma
- Author
-
Yunlu Jia, Jianbiao Zhou, Tze King Tan, Tae-Hoon Chung, Yongxia Chen, Jing-Yuan Chooi, Takaomi Sanda, Melissa J. Fullwood, Sinan Xiong, Sabrina H.M. Toh, Kalpnaa Balan, Regina W.J. Wong, Julia S.L. Lim, Enfan Zhang, Zhen Cai, Peng Shen, Wee Joo Chng, School of Biological Sciences, Cancer Science Institute of Singapore, NUS, and Centre for Translational Medicine
- Subjects
Cancer Research ,Oncology ,Carcinogenesis ,Biological sciences [Science] ,Medicine [Science] ,Apoptosis - Abstract
Multiple myeloma is an incurable malignancy with marked clinical and genetic heterogeneity. The cytogenetic abnormality t(4;14) (p16.3;q32.3) confers aggressive behavior in multiple myeloma. Recently, essential oncogenic drivers in a wide range of cancers have been shown to be controlled by super-enhancers (SE). We used chromatin immunoprecipitation sequencing of the active enhancer marker histone H3 lysine 27 acetylation (H3K27ac) to profile unique SEs in t(4;14)-translocated multiple myeloma. The histone chaperone HJURP was aberrantly overexpressed in t(4;14)-positive multiple myeloma due to transcriptional activation by a distal SE induced by the histone lysine methyltransferase NSD2. Silencing of HJURP with short hairpin RNA or CRISPR interference of SE function impaired cell viability and led to apoptosis. Conversely, HJURP overexpression promoted cell proliferation and abrogated apoptosis. Mechanistically, the NSD2/BRD4 complex positively coregulated HJURP transcription by binding the promoter and active elements of its SE. In summary, this study introduces SE profiling as an efficient approach to identify new targets and understand molecular pathogenesis in specific subtypes of cancer. Moreover, HJURP could be a valuable therapeutic target in patients with t(4;14)-positive myeloma. SIGNIFICANCE: A super-enhancer screen in t(4;14) multiple myeloma serves to identify genes that promote growth and survival of myeloma cells, which may be evaluated in future studies as therapeutic targets. Published version The work was supported by the National Research Foundation Singapore and the Singapore Ministry of Education under its Research Centres of Excellence initiative (to W.J. Chng), the NMRC Clinician Scientist Investigator award (to W.J. Chng), the RNA Biology Center at CSI Singapore, NUS, from funding by the Singapore Ministry of Education’s Tier 3 grants, grant number MOE2014-T3–1-006 (to W.J. Chng), the National Natural Science Foundation of China (grant no. 82000212 to Y. Jia), Natural Science Foundation of Zhejiang Province (grant no. LQ21H160022 to Y. Jia), Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (grant no. 2021RC003 to Y. Jia). Y. Jia also thanks the China Scholarship Council (grant no. 201706320167) for financial support to visit National University of Singapore.
- Published
- 2021
- Full Text
- View/download PDF
4. Myeloma-specific superenhancers affect genes of biological and clinical relevance in myeloma
- Author
-
Sanjay de Mel, Regina Wan Ju Wong, Yunlu Jia, Cinnie Yentia Soekojo, Jianbiao Zhou, Yongxia Chen, Zhen Cai, Tze King Tan, Jian Ruan, Jing Yuan Chooi, Peng Shen, Melissa Ooi, Enfan Zhang, Wee Joo Chng, Tae-Hoon Chung, Julia Sze Lynn Lim, and Takaomi Sanda
- Subjects
Cell biology ,Biology ,lcsh:RC254-282 ,Article ,Transcription (biology) ,Cell Line, Tumor ,medicine ,Humans ,Gene Regulatory Networks ,Enhancer ,Gene ,Transcription factor ,Multiple myeloma ,Cancer ,Adaptor Proteins, Signal Transducing ,Cell growth ,Oncogenes ,Hematology ,Plasma cell neoplasm ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Gene Expression Regulation, Neoplastic ,Enhancer Elements, Genetic ,Oncology ,Proto-Oncogene Proteins c-maf ,Cancer cell ,Cancer research ,Multiple Myeloma ,Guanylate Kinases - Abstract
Multiple myeloma (MM) is an aggressive plasma cell neoplasm characterized by genomic heterogeneity. Superenhancers (SEs) are defined as large clusters of enhancers in close genomic proximity, which regulate genes for maintaining cellular identity and promote oncogenic transcription to which cancer cells highly addicted. Here, we analyzed cis-regulatory elements in MM samples with H3K27ac ChIP-seq, to identify novel SE-associated genes involved in the myeloma pathogenesis. SEs and their associated genes in cancerous tissue were compared with the control samples, and we found SE analysis alone uncovered cell-lineage-specific transcription factors and well-known oncogenes ST3GAL6 and ADM. Using a transcriptional CDK7 inhibitor, THZ1, coupled with H3K27ac ChlP-seq, we identified MAGI2 as a novel SE-associated gene of myeloma cells. Elevated MAGI2 was related to myelomagenesis with gradual increased expression from MGUS, SMM to newly diagnosed and relapsed MM. High prevalence of MAGI2 was also associated with poor survival of MM patients. Importantly, inhibition of the SE activity associated with MAGI2 decreased MAGI2 expression, inhibited cell growth and induced cell apoptosis. Mechanistically, we revealed that the oncogenic transcription factor, MAF, directly bound to the SE region and activated gene transcription. In summary, the discoveries of these acquired SEs-associated genes and the novel mechanism by which they are regulated provide new insights into MM biology and MAGI2-MAF-SE regulatory circuit offer potential novel targets for disease treatment.
- Published
- 2021
5. HJURP regulates cell proliferation and chemo-resistance via YAP1/NDRG1 transcriptional axis in triple-negative breast cancer
- Author
-
Misha Mao, Yunlu Jia, Yongxia Chen, Jingjing Yang, Ling Xu, Xun Zhang, Jichun Zhou, Zhaoqing Li, Cong Chen, Siwei Ju, and Linbo Wang
- Subjects
Cancer Research ,Transcription, Genetic ,Immunology ,Intracellular Signaling Peptides and Proteins ,Cell Cycle Proteins ,Triple Negative Breast Neoplasms ,YAP-Signaling Proteins ,Cell Biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Cellular and Molecular Neuroscience ,Gene Expression Regulation ,Cell Line, Tumor ,Humans ,Cell Proliferation - Abstract
Triple-negative breast cancer is still a difficult point in clinical treatment at present, and a deep study of its pathogenesis has great clinical value. Therefore, our research mainly focuses on exploring the progression of triple-negative breast cancer and determines the important role of the HJURP/YAP1/NDRG1 transcriptional regulation axis in triple-negative breast cancer. We observed significantly increased HJURP expression levels in triple-negative breast cancer compared to other subtypes. HJURP could affect the level of ubiquitination modification of YAP1 protein and then regulate its downstream transcriptional activity. Mechanistically, we found that YAP1 positively regulates NDRG1 transcription by binding the promoter region of the NDRG1 gene. And HJURP/YAP1/NDRG1 axis could affect cell proliferation and chemotherapy sensitivity in triple-negative breast cancer. Taken together, these findings provide insights into the transcriptional regulation axis of HJURP/YAP1/NDRG1 in triple-negative breast cancer progression and therapeutic response.
- Published
- 2022
- Full Text
- View/download PDF
6. Corrigendum: STAT5a Confers Doxorubicin Resistance to Breast Cancer by Regulating ABCB1
- Author
-
Zhaoqing Li, Cong Chen, Lini Chen, Dengdi Hu, Xiqian Yang, Wenying Zhuo, Yongxia Chen, Jingjing Yang, Yulu Zhou, Misha Mao, Xun Zhang, Ling Xu, Siwei Ju, Jun Shen, Qinchuan Wang, Minjun Dong, Shuduo Xie, Qun Wei, Yunlu Jia, Jichun Zhou, and Linbo Wang
- Subjects
Cancer Research ,Oncology - Published
- 2022
- Full Text
- View/download PDF
7. Immunotherapy of cholangiocarcinoma: Therapeutic strategies and predictive biomarkers
- Author
-
Ruyin, Chen, Dandan, Zheng, Qiong, Li, Shuaishuai, Xu, Chanqi, Ye, Qi, Jiang, Feifei, Yan, Yunlu, Jia, Xiaochen, Zhang, and Jian, Ruan
- Subjects
Cholangiocarcinoma ,Cancer Research ,Bile Ducts, Intrahepatic ,Bile Duct Neoplasms ,Oncology ,Tumor Microenvironment ,Humans ,Immunotherapy ,Biomarkers - Abstract
Cholangiocarcinoma (CCA) is a group of malignant heterogeneous cancer arising from the biliary tree. CCA has become a global health problem with rising incidence and mortality that threatens the health of human beings. The immune microenvironment of CCA is characterized by abundant cancer-associated fibroblast and suppressive immune components. The increasing body of knowledge and recent developments in transcriptomic studies have given insight into the immune landscape of CCA, paving the way for better application of immunotherapy. Immunotherapy mainly applies in a limited subset of CCA with deficient mismatch and high microsatellite instability. With limited response rates and treatment efficacy, researchers are looking into novel strategies on combination strategies and alternatives, such as immune vaccines and adoptive cell therapy. Biomarker identification is also critical for patient selection. We present an up-to-date summary of the current research on immunotherapy for CCA patients, covering pre-clinical and clinical exploration beyond immune checkpoint inhibitors, immune vaccines, and adoptive cell therapy. In addition, we review the promising biomarkers for CCA immunotherapy and discuss recent development.
- Published
- 2022
- Full Text
- View/download PDF
8. STAT5a Confers Doxorubicin Resistance to Breast Cancer by Regulating ABCB1
- Author
-
Zhaoqing Li, Cong Chen, Lini Chen, Dengdi Hu, Xiqian Yang, Wenying Zhuo, Yongxia Chen, Jingjing Yang, Yulu Zhou, Misha Mao, Xun Zhang, Ling Xu, Siwei Ju, Jun Shen, Qinchuan Wang, Minjun Dong, Shuduo Xie, Qun Wei, Yunlu Jia, Jichun Zhou, and Linbo Wang
- Subjects
0301 basic medicine ,Cancer Research ,animal structures ,stat ,STAT5A ,03 medical and health sciences ,breast cancer ,0302 clinical medicine ,Breast cancer ,Pimozide ,In vivo ,Medicine ,Doxorubicin ,skin and connective tissue diseases ,RC254-282 ,Original Research ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,food and beverages ,ABCB1 ,pimozide ,medicine.disease ,doxorubicin resistance ,030104 developmental biology ,Psychotropic drug ,Oncology ,030220 oncology & carcinogenesis ,STAT protein ,Cancer research ,business ,medicine.drug - Abstract
Chemoresistance is a daunting challenge to the prognosis of patients with breast cancer. Signal transducer and activator of transcription (STAT) 5a plays vital roles in the development of various cancers, but its function in breast cancer is controversial, and its role in chemoresistance in breast cancer remains unexplored. Here we identified STAT5a as a chemoresistance inducer that regulates the expression of ABCB1 in breast cancer and can be targeted by pimozide, an FDA-approved psychotropic drug. First, we found that STAT5a and ABCB1 were expressed at higher levels in doxorubicin-resistant cell lines and chemoresistant patients, and their expression was positively correlated. Then, we confirmed the essential roles of STAT5a and ABCB1 in doxorubicin resistance in breast cancer cells and the regulation of ABCB1 transcription by STAT5a. Subsequently, the efficacy of pimozide in inhibiting STAT5a and sensitizing doxorubicin-resistant breast cancer cells was tested. Finally, we verified the role of STAT5a in doxorubicin resistance in breast cancer and the efficacy of pimozide in reversing this resistance in vivo. Our study demonstrated the vital role of STAT5a in doxorubicin resistance in breast cancer. Targeting STAT5a might be a promising strategy for treating doxorubicin-resistant breast cancer. Moreover, repurposing pimozide for doxorubicin resensitization is attractive due to the safety profile of pimozide.
- Published
- 2021
- Full Text
- View/download PDF
9. P-098: Super-enhancer-driven PPP1R15B as an oncogenic and potential therapeutic target in Multiple Myeloma
- Author
-
Tze King Tan, Sinan Xiong, Sabrina Hui Min Toh, Takaomi Sanda, Yunlu Jia, Tae-Hoon Chung, Kalpnaa Balan, Jianbiao Zhou, and Wee Joo Chng
- Subjects
Cancer Research ,Oncogene ,Cell growth ,business.industry ,Hematology ,Cell biology ,Salubrinal ,Transcriptome ,chemistry.chemical_compound ,Oncology ,chemistry ,RNA interference ,Unfolded protein response ,Phosphatase complex ,Medicine ,Signal transduction ,business - Abstract
Background Growing evidence suggests that alterations in epigenetic landscape contribute to pathogenesis of multiple myeloma (MM). MM cells are highly dependent on unfolded protein response signaling pathways due to high level of endoplasmic reticulum stress. Phosphorylation of eIF2a can attenuate protein translation. The PPP1R15B(denoted as R15B hereafter) gene encodes a regulatory subunit of eIF2a-specific phosphatase complex. In this study,we identified super enhancer (SE)-driven oncogenes specific in MM with a particular focus on a candidate gene R15B, whose functional roles in MM remain largely elusive. Methods We performed H3K27Ac ChIP-seq on MM cell lines, primary MM patient samples, normal CD138+ plasma cells and memory B cells. ROSE analysis was used to annotate SEs and their associated genes. A combination of public data mining, RNAi, overexpression and CRISPR/Cas9 technologies were conducted to determine the oncogenic effects of R15B in MM. Transcriptome analysis of MM cell line H929 with R15B KD and scrambled control was performed. To further study the interactions between SE and its promoters, we are currently working on HiChIP. Results We have identified R15B as one of the SE-associated genes specific to MM patient samples and cell lines. SE activity was correlated with the expression level of R15B. Higher expression of R15B predicted poor overall survival of MM patients, suggesting its clinical relevance in MM pathogenesis. R15B KD or KO significantly reduced cell viability, clonogenicity and induced G2/M arrest. ChIP-qPCR assays showed that C/EBP-β is strongly enriched at R15B SE region. We also found that salubrinal, a selective inhibitor of eIF2a phosphorylation, inhibited MM cell proliferation in a dose-dependent manner. Conclusions Our integrative approaches identified R15B as a novel SE-driven oncogene. We propose that targeting R15B may serve as a new approach for effective anti-myeloma therapy, which warrants further clinical investigation.
- Published
- 2021
- Full Text
- View/download PDF
10. Abstract 2956: Superenhancer-drvien SMC4 promote cell growth and proliferation in multiple myeloma
- Author
-
Yunlu Jia, Jianbiao Zhou, Tze-King Tan, Tae-Hoon Chung, Yongxia Chen, Enfan Zhang, Zhen Cai, Takaomi Sanda, and Wee-Joo Chng
- Subjects
Cancer Research ,Oncology - Abstract
Background Multiple myeloma (MM) is an incurable malignancy characterized by complex heterogeneous cytogenetic abnormalities. Defined as large clusters of cis-acting enhancers, super-enhancers (SEs) could promote oncogenic transcription to which cancer cells highly addicted. Structural maintenance of chromosome 4 (SMC4) is a core subunit of condensin complexes, which contributes to chromosome condensation and segregation. Here, we identified SMC4 as a novel SE-associated oncogene of myeloma cells, whose functional roles in MM remain largely elusive. Methods We performed H3K27ac ChIP-seq and RNA-seq on primary MM patient samples, MM cell lines, normal CD138+ plasma cell and lymphoma cell lines as the controls, then the ROSE analysis was used to annotate SEs and their associated genes. THZ1 as a transcriptional CDK7 inhibitor was used to further filter THZ1-responsive genes. Combined analysis of THZ1-sensitive and SE-associated gene uncovered a number of promising MM oncogenes as SMC4. The public data mining, RNA interference, CRISPR/Cas9 gene editing system, overexpression and a variety of cellular functional assays were performed to determine the oncogenic effects of SMC4 on MM malignant. Transcriptome analysis with SMC4 silencing and overexpression, and the underlying molecular mechanism of SMC4 oncogenic role in MM are ongoing. Results We have demonstrated SMC4 as one of the SE-associated genes specific to MM patient samples and cell lines. SMC4 was related to myelomagenesis with increased expression from MGUS, SMM to MM cases, and overexpression of SMC4 predicted poor overall survival of MM patients, suggesting its clinical relevance. SMC4 silencing in MM cells suppressed cell proliferation and lead to cell apoptosis. Repression of the SMC4-SE region also led to impairment of cell viability and cell growth, consistent with an oncogenic function. Conclusion In summary, our integrative approached by combing H3H27ac ChIP-seq, RNA-seq and THZ1-sensitive transcripts identified the landscape of SEs and novel oncogenic transcripts as SMC4 in MM. Mapping these acquired SEs and their associated genes may provide novel insight into understanding MM biology, and SMC4 may serve as novel therapeutic targets in MM. Citation Format: Yunlu Jia, Jianbiao Zhou, Tze-King Tan, Tae-Hoon Chung, Yongxia Chen, Enfan Zhang, Zhen Cai, Takaomi Sanda, Wee-Joo Chng. Superenhancer-drvien SMC4 promote cell growth and proliferation in multiple myeloma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2956.
- Published
- 2022
- Full Text
- View/download PDF
11. PLAC8 promotes adriamycin resistance via blocking autophagy in breast cancer
- Author
-
Qun Wei, Jian Ruan, Zhaoqing Li, Yifeng Gu, Cong Chen, Linbo Wang, Dengdi Hu, Jingjing Yang, Misha Mao, Peng Shen, Yongxia Chen, Wenxian Hu, Jichun Zhou, Yunlu Jia, Lini Chen, Chenpu Xu, and Jun Shen
- Subjects
0301 basic medicine ,autophagy ,Cell ,PLAC8 ,Mice, Nude ,Aggressive phenotype ,law.invention ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Plasmid ,Breast cancer ,breast cancer ,law ,Medicine ,Animals ,Humans ,skin and connective tissue diseases ,adriamycin resistance ,Antibiotics, Antineoplastic ,Oncogene ,business.industry ,Autophagy ,p62 ,Mammary Neoplasms, Experimental ,Proteins ,Cell Biology ,Original Articles ,medicine.disease ,In vitro ,030104 developmental biology ,medicine.anatomical_structure ,Doxorubicin ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,MCF-7 Cells ,Molecular Medicine ,Suppressor ,Original Article ,Female ,business - Abstract
Background Adriamycin (ADM) is currently one of the most effective chemotherapeutic agents in breast cancer treatment. However, growing resistance to ADM can lead to treatment failure and poor outcome. The underlying molecular mechanisms in ADM resistance in breast cancer remains unclear. PLAC8 is reported as a novel highly-conserved protein and functions as an oncogene or tumor suppressor in various tumors. Methods Here, we analyzed the expression profile of PLAC8 in breast cancer tissues and breast cancer cell lines, and explored the correlation of PLAC8 expression levels with patients’ outcomes and ADM response. One ADM resistant MCF-7 breast cancer cell (MCF-7/ADM) and its parental cell was used as in vitro models to identify the underlying mechanism of PLAC8 and ADM resistance. Breast cancer cells were transfected with PLAC8 knockdown and overexpression vectors, and MTT and colony formation assays were performed to test the cell response to ADM. Then, we tested the effect of PLAC8 on autophagy pathway by flow cytometry and immunofluorescence analysis, and the change of main autophagy-correlated factors expressions: LC3 and p62. Next, combining treatment of autophagy inhibitor/inducer and PLAC8 downregulation/upregulation revealed the participation of PLAC8 in autophagy pathway to synergistically regulate ADM resistance in breast cancer. Results Here, higher PLAC8 expression was correlated with poorer outcome and aggressive phenotype in breast cancer, and breast cancer patients with higher PLAC8 expression showed potential ADM resistance. PLAC8 expression level was also significantly elevated in ADM resistant MCF-7 breast cancer cells (MCF-7/ADM), compared to parental MCF-7 cells. In vitro experiments further confirmed that PLAC8 inhibition by siRNA or enforced overexpression by infecting pcDNA3.1(C)-PLAC8 plasmid correspondingly decreased or increased the ADM resistance. Subsequently, we demonstrated that ectopic PLAC8 expression in MCF-7/ADM cell blocked the accumulation of the autophagy-associated protein LC3II, and resulted in cellular accumulation of p62. Rapamycin-triggered autophagy significantly increased cell response to ADM, while the autophagy inhibitor 3-MA enhanced ADM resistance. Actually, 3-MA and PLAC8 could synergistically enhance ADM resistance via blocking the autophagy process. Additionally, the downregulation of p62 by siRNA attenuated the activation of autophagy and PLAC8 expression in breast cancer cells. Conclusion Our findings suggest that PLAC8, through participation of p62, inhibits autophagy and consequently results in ADM resistance in breast cancer. PLAC8/p62/autophagy pathway may act as novel therapeutic targets in breast cancer treatment and has potential clinical application in overcoming ADM resistance.
- Published
- 2020
- Full Text
- View/download PDF
12. H19/let‑7/Lin28 ceRNA network mediates autophagy inhibiting epithelial‑mesenchymal transition in breast cancer
- Author
-
Zihan Chen, Linbo Wang, Shuduo Xie, Yunlu Jia, Jianguo Shen, Wenhe Zhao, Bojian Xie, Ushani Jayasinghe, Jichun Zhou, Jingjing Yang, Hanchu Xiong, and Ji Wang
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,Cell Survival ,Breast Neoplasms ,Biology ,LIN28 ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Movement ,Cell Line, Tumor ,microRNA ,Autophagy ,Humans ,Epithelial–mesenchymal transition ,Cell Proliferation ,Regulation of gene expression ,Oncogene ,Competing endogenous RNA ,RNA-Binding Proteins ,female genital diseases and pregnancy complications ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,embryonic structures ,MCF-7 Cells ,Cancer research ,Female ,RNA, Long Noncoding - Abstract
Long non‑coding RNA (lncRNA) H19 and Lin28 protein have been shown to participate in various pathophysiological processes, including cellular proliferation, autophagy and epithelial‑mesenchymal transition (EMT). A number of studies have investigated lncRNAs, microRNAs and mRNAs, and their roles in the initiation and progression of cancer, in doing so identifying competitive endogenous RNA (ceRNA) networks, including the H19/let‑7/Lin28 network. However, whether the H19/let‑7/Lin28 ceRNA network is involved in autophagy and EMT in breast cancer (BC) remains unclear. The present study demonstrated that the H19/let‑7/Lin28 loop was required for the downregulation of autophagy in BC cells via western blot analysis, reverse transcription‑quantitative PCR and autophagy flux monitoring. Using wound healing, migration and invasion assays, and morphological assays, the H19/let‑7/Lin28 loop was revealed to promote EMT in BC cells. Moreover, the H19/let‑7/Lin28 network was found to contribute to autophagy by inhibiting EMT in BC cells. To the best of our knowledge, the present study is the first to suggest the important roles of the H19/let‑7/Lin28 ceRNA network in BC autophagy and EMT, thus providing insight for the use of these molecules as prognostic biomarkers and therapeutic targets in BC metastasis.
- Published
- 2020
- Full Text
- View/download PDF
13. KLF4 overcomes tamoxifen resistance by suppressing MAPK signaling pathway and predicts good prognosis in breast cancer
- Author
-
Yongxia Chen, Wuguo Deng, Ji Wang, Miao Chen, Hanchu Xiong, Yunlu Jia, Wenhe Zhao, Wenxian Hu, Jichun Zhou, Linbo Wang, Xiaogang Ying, and Xiao Luo
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,medicine.medical_specialty ,Antineoplastic Agents, Hormonal ,p38 mitogen-activated protein kinases ,Kruppel-Like Transcription Factors ,Datasets as Topic ,Breast Neoplasms ,p38 Mitogen-Activated Protein Kinases ,Kruppel-Like Factor 4 ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,stomatognathic system ,Cell Line, Tumor ,Internal medicine ,Prohibitins ,medicine ,Humans ,Viability assay ,Extracellular Signal-Regulated MAP Kinases ,skin and connective tissue diseases ,Gene knockdown ,Cell growth ,business.industry ,Estrogen Receptor alpha ,Cell Biology ,Prognosis ,medicine.disease ,Survival Analysis ,Gene Expression Regulation, Neoplastic ,Tamoxifen ,030104 developmental biology ,Endocrinology ,Drug Resistance, Neoplasm ,KLF4 ,030220 oncology & carcinogenesis ,embryonic structures ,MCF-7 Cells ,Cancer research ,Female ,sense organs ,Signal transduction ,business ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Tamoxifen resistance represents a daunting challenge to the successful treatment for breast cancer. Krüppel-like factor 4 has critical roles in the development and progression of breast cancer, but its expression, function and regulation in the efficacy of TAM therapy in breast cancer have yet to be investigated. Here, we examined the clinical significance and biologic effects of KLF4 in breast cancer. Firstly, higher expression of KLF4 correlated with increased TAM sensitivity in breast cancer cells, and analysis of GEO datasets indicated that KLF4 expression was positively correlated with ERα and enhanced expression of KLF4 sensitized breast cancer patients to endocrine therapy. Knockdown of KLF4 in MCF-7 and BCAP37 cells led to increased TAM resistance, while ectopic KLF4 expression promoted the responsiveness to TAM in T47D and TAM-resistant MCF-7/TAM cells. Secondly, ectopic KLF4 overexpression suppressed MCF-7/TAM cell growth, invasion and migration. Moreover, KLF4 expression was down-regulated in breast cancer tumor tissues and high expression of KLF4 was associated with favorable outcomes. Mechanistically, KLF4 may enhance the responsiveness of breast cancer cells to TAM through suppressing mitogen-activated protein kinase (MAPK) signaling pathway. We found that ERK and p38 were more activated in MCF-7/TAM compared with MCF-7, and treatment with MAPK-specific inhibitors significantly suppressed cell viability. Knockdown of KLF4 activated ERK and p38 and drove MCF-7 cells to become resistant to TAM. Conversely, overexpression of KLF4 in MCF-7/TAM cells suppressed ERK and p38 signaling and resulted in increased sensitivity to TAM. Therefore, our findings suggested that KLF4 contributed to TAM sensitivity in breast cancer via phosphorylation modification of ERK and p38 signaling. Collectively, this study highlighted the significance of KLF4/MAPK signal interaction in regulating TAM resistance of breast cancer, and suggested that targeting KLF4/MAPK signaling may be a potential therapeutic strategy for breast cancer treatment, especially for the TAM-resistant patients.
- Published
- 2018
- Full Text
- View/download PDF
14. The long noncoding RNA H19 promotes tamoxifen resistance in breast cancer via autophagy
- Author
-
Cong Chen, Yulu Zhou, Chenyang Ye, Jingjing Yang, Yunlu Jia, Jichun Zhou, Linbo Wang, Xiaogang Ying, Hanchu Xiong, Yongxia Chen, Ji Wang, and Shuduo Xie
- Subjects
0301 basic medicine ,Cancer Research ,Apoptosis ,medicine.disease_cause ,DNA Methyltransferase 3A ,Mice ,Random Allocation ,Breast cancer ,0302 clinical medicine ,DNA (Cytosine-5-)-Methyltransferases ,Promoter Regions, Genetic ,skin and connective tissue diseases ,Gene knockdown ,Chemistry ,Tamoxifen resistance ,lcsh:Diseases of the blood and blood-forming organs ,Hematology ,Methylation ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,female genital diseases and pregnancy complications ,Oncology ,Doxycycline ,030220 oncology & carcinogenesis ,embryonic structures ,MCF-7 Cells ,Beclin-1 ,Female ,RNA, Long Noncoding ,LncRNA H19 ,Signal Transduction ,medicine.drug ,Mice, Nude ,Breast Neoplasms ,lcsh:RC254-282 ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,Autophagy ,medicine ,Animals ,Humans ,Beclin1 ,Molecular Biology ,Cell Proliferation ,lcsh:RC633-647.5 ,Research ,Adenosylhomocysteinase ,DNA Methylation ,medicine.disease ,Xenograft Model Antitumor Assays ,Tamoxifen ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer research ,Carcinogenesis ,Chromatin immunoprecipitation - Abstract
Background Tamoxifen resistance remains a clinical challenge for hormone receptor-positive breast cancer. Recently, dysregulations in autophagy have been suggested as a potential mechanism for tamoxifen resistance. Although the long noncoding RNA H19 is involved in various stages of tumorigenesis, its role in tamoxifen resistance remains unknown. Here, we assessed the role of H19 in the development of tamoxifen-resistant breast cancer. Methods Quantitative real-time PCR analyzed expression of H19 in tamoxifen-resistant breast cancer tissues. Knockdown of H19 was used to assess the sensitivity to tamoxifen in vitro and in vivo. Both knockdown and overexpression of H19 were used to analyze the status of autophagy. Real-time quantitative methylation-specific polymerase chain reaction, chromatin immunoprecipitation, immunofluorescence, and Western blot were used to explore the tamoxifen resistance mechanism of H19. Results In this study, we observed that the expression of H19 was substantially upregulated in tamoxifen-resistant breast cancer cell line and tumor tissues, and knockdown of H19 enhanced the sensitivity to tamoxifen both in vitro and in vivo. Furthermore, knockdown of H19 significantly inhibited autophagy in MCF7 tamoxifen-resistant (MCF7/TAMR) cells. Conversely, overexpression of H19 promoted autophagy. Interestingly, overexpression of H19 in MCF7 tamoxifen-sensitive cells could recapitulate tamoxifen resistance. Moreover, an increase in methylation in the promoter region of Beclin1 was observed in MCF7/TAMR-shH19 cells. In the double knockdown groups, both shH19+shSAHH and shH19+shDNMT3B rescued the Beclin1 promoter region methylation levels and reactivated autophagy functions. A chromatin immunoprecipitation assay further validated that DNMT3B binds to the Beclin1 promoter region and the knockdown of H19 increases this binding. Conclusions Our findings demonstrate that H19 induces autophagy activation via the H19/SAHH/DNMT3B axis, which could contribute to tamoxifen resistance in breast cancer. Electronic supplementary material The online version of this article (10.1186/s13045-019-0747-0) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
15. PLCA8 suppresses breast cancer apoptosis by activating the PI3k/AKT/NF-κB pathway
- Author
-
Cong Chen, Misha Mao, Zhaoqing Li, Lini Chen, Qun Wei, Yunlu Jia, Linbo Wang, Yongxia Chen, and Jingjing Yang
- Subjects
0301 basic medicine ,Cell Survival ,Mice, Nude ,PLAC8 ,Apoptosis ,Breast Neoplasms ,03 medical and health sciences ,chemistry.chemical_compound ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,breast cancer ,In vivo ,Cell Movement ,Cell Line, Tumor ,Gene silencing ,Animals ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Mice, Inbred BALB C ,Cell growth ,tumour growth ,NF-kappa B ,Proteins ,NF-κB ,Cell migration ,Cell Biology ,Original Articles ,Middle Aged ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,030220 oncology & carcinogenesis ,Caspases ,Cancer research ,Molecular Medicine ,Female ,Original Article ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
The cysteine‐rich lysosomal protein placenta‐specific 8 (PLAC8), also called onzin, has been shown to be involved in many types of cancers, and its role is highly dependent on cellular and physiological contexts. However, the precise function of PLAC8 in breast cancer (BC) progression remains unclear. In this study, we investigated both the clinical significance and biological functions of PLAC8 in BC progression. First, high PLAC8 expression was observed in primary BC tissues compared with adjacent normal tissues through immunohistochemistry analysis. The results of in vitro and in vivo assays further confirmed that PLAC8 overexpression promotes cell proliferation and suppress BC cell apoptosis, whereas PLAC8 silencing has the opposite effect. In addition, the forced expression of PLAC8 greatly induces cell migration, partially by affecting the EMT‐related genes, including down‐regulating E‐cadherin expression and facilitating vimentin expression. Further mechanistic analysis confirmed that PLAC8 contributes to cell proliferation and suppresses cell apoptosis in BC by activating the PI3K/AKT/NF‐κB pathway. The results of our study provide new insights into an oncogenic role of PLAC8 and reveal a novel PLAC8/ PI3K/AKT/NF‐κB pathway as a potential therapeutic target for BC.
- Published
- 2019
16. Super-Enhancer-Driven TOX2 Mediates Oncogenesis in Natural Killer/T Cell Lymphoma
- Author
-
Wee Joo Chng, Jing Quan Lim, Jianbiao Zhou, Choon Kiat Ong, Tuan Zea Tan, Kalpnaa Balan, Tze King Tan, Takaomi Sanda, Yunlu Jia, Sabrina Hui Min Toh, Siok Bian Ng, and Soon Thye Lim
- Subjects
Super-enhancer ,Immunology ,medicine ,Cancer research ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Natural killer T cell ,Carcinogenesis ,medicine.disease_cause ,Biochemistry ,Lymphoma - Abstract
Background: Extranodal natural killer/T-cell lymphoma (NKTL) is an EBV associated, aggressive malignancy. Activation of JAK/STAT pathway, overexpression of EZH2, and alternations in epigenetic program have been implicated in the pathogenesis of NKTL. Combined chemotherapy-radiotherapy is standard treatment for NKTL patients, but often associated with high relapse rate and serious side effects. New drugs, including anti-PD1 antibody pembrolizumab, have been explored. Overall, treatment for NKTL patients remains challenge in clinic. Super-enhancers (SEs) are defined as large clusters of cis-acting enhancers, marked by high level bindings of acetylation of histone H3 lysine 27 (H3K27ac), coactivators and transcription factors. SE associated oncogenes have been demonstrated to play key roles in various types of cancers. In this study, we aim to define the SE landscapes of NKTL for a better understanding of the molecular pathogenesis of NKTL and to identify novel therapeutic targets. Methods: We performed H3K27Ac ChIP-seq and RNA-seq on primary NKTL tumor samples and paired normal tonsil tissues as controls, then the Rank Ordering of Super Enhancers (ROSE) analysis was used to systematically annotate SEs and their associated genes between tumors and controls. A combination of RNA interference (RNAi), CRISPRi (dCas9-KRAB) technologies and overexpression experiments, followed by several functional assays were performed to determine the effects of candidate SE-genes on NKTL cells. Circularized chromatin conformation capture followed by sequencing (4C-seq) experiments which examine the direct contact of SE and its promoter are ongoing. Results: A total of 1266 SEs were identified in more than 2 out of 3 primary NKTL tumors but not in their normal tonsils. RNA-seq revealed overexpression of 1478 genes (false discovery rate Conclusions: Taken together, our integrative approaches defined the landscape of SE and identified high-value SE-associated oncogenes in NKTL. Among them, we demonstrated that TOX2 is a novel tumor driver, which promotes NKTL cell growth and enhances ability of colony formation, as well as protects cell viability under adverse condition. This study not only provides novel insight into the NKTL pathology, but also offer novel, potential therapeutic targets, such as TOX2 for the treatment of NKTL patients. Disclosures Lim: National Cancer Centre Singapore: Current Employment. Chng:Abbvie: Honoraria; Janssen: Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Novartis: Honoraria; Amgen: Honoraria, Research Funding.
- Published
- 2020
- Full Text
- View/download PDF
17. The novel KLF4/PLAC8 signaling pathway regulates lung cancer growth
- Author
-
Xiaogang Ying, Xiao Luo, Yongxia Chen, Linbo Wang, Shudu Xie, Qin chuan Wang, Jichun Zhou, Wenxian Hu, and Yunlu Jia
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Carcinogenesis ,Immunology ,Kruppel-Like Transcription Factors ,Mice, Nude ,Biology ,medicine.disease_cause ,Article ,Kruppel-Like Factor 4 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,lcsh:QH573-671 ,Promoter Regions, Genetic ,Lung cancer ,Aged ,Cell Proliferation ,Aged, 80 and over ,lcsh:Cytology ,Cell growth ,Proteins ,Cancer ,Cell Cycle Checkpoints ,Cell Biology ,Middle Aged ,medicine.disease ,Survival Analysis ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,KLF4 ,030220 oncology & carcinogenesis ,Multivariate Analysis ,Cancer research ,Female ,Signal transduction ,Chromatin immunoprecipitation ,Protein Binding ,Signal Transduction - Abstract
Accumulating evidence suggests that placenta-specific 8 (PLAC8) plays an important role in normal cellular process and human diseases, including multiple types of human tumors, and its role is highly relied upon in cellular and physiologic contexts. However, there are no reports on its expression profile and biological roles during lung cancer development. In the current study, both the clinical implications and biological effects of PLAC8 in lung cancer (LC) progression were investigated, and we identified and described the novel Krüppel-like factor 4 (KLF4)/PLAC8 regulatory pathway in cancer progression. Elevated PLAC8 levels were positively correlated with tumor size, histological grade, and tumor node metasis (TNM) stage, and LC patients with high PLAC8 expression suffered poor outcomes. In vitro and in vivo assays further revealed that endogenous PLAC8 promoted cell proliferation and tumor formation. We also found downregulated PLAC8 protein in several LC cell lines following the induction of KLF4, and immunohistochemistry analysis of LC tissues by microarray indicated a potential inverse correlation between PLAC8 and KLF4 expression. Luciferase reporter analysis and chromatin immunoprecipitation assays determined that KLF4 negatively regulated PLAC8 promoter activity via directly binding to the promoter region. Furthermore, the growth inhibition resulting from KLF4 overexpression was partially rescued by ectopic PLAC8 expression. Together, our data uncovered a previously unidentified role of PLAC8 as a central mediator in LC progression. PLAC8 was transcriptionally repressed by KLF4, and the novel KLF4/PLAC8 axis may act as a promising candidate target for LC diagnosis and therapy.
- Published
- 2018
- Full Text
- View/download PDF
18. Super-enhancer profiling of multiple myeloma in search of novel oncogenes and therapeutic targets
- Author
-
Takaomi Sanda, Yunlu Jia, Jianbiao Zhou, Tze King Tan, Tae-Hoon Chung, and Wee Joo Chng
- Subjects
Cancer Research ,Super-enhancer ,Oncology ,business.industry ,medicine ,Profiling (information science) ,Hematology ,Computational biology ,medicine.disease ,business ,Multiple myeloma - Published
- 2019
- Full Text
- View/download PDF
19. Ku80 cooperates with CBP to promote COX-2 expression and tumor growth
- Author
-
Wei Cheng, Wenlin Huang, Yao Xiao, Yiming Chen, Shilei Zhao, Quentin Liu, Canhui Yi, Yang Xuan, Xiangsheng Xiao, Yunlu Jia, Wendan Yu, Jingshu Wang, Wenxian Hu, Wuguo Deng, Meng Dai, Mei Li, Taihua Wu, Songshu Meng, Shusen Wang, Zhenglin Li, Wei Guo, Sha Du, Yu Qin, and Yuhui Yuan
- Subjects
MAPK/ERK pathway ,Ku80 ,Lung Neoplasms ,Mice ,Random Allocation ,Cell Movement ,2.1 Biological and endogenous factors ,RNA, Small Interfering ,Promoter Regions, Genetic ,Lung ,Cancer ,Gene knockdown ,Tumor ,Lung Cancer ,Antigens, Nuclear ,Transfection ,DNA-Binding Proteins ,Oncology ,Disease Progression ,Heterografts ,Biotechnology ,Research Paper ,Oncology and Carcinogenesis ,Adenocarcinoma of Lung ,Biology ,Adenocarcinoma ,Small Interfering ,CBP ,Cell Line ,Promoter Regions ,Rare Diseases ,Genetic ,Cell Line, Tumor ,Genetics ,medicine ,Gene silencing ,Animals ,Humans ,Nuclear ,Antigens ,Lung cancer ,Ku Autoantigen ,Cell Proliferation ,Cell growth ,Promoter ,COX-2 ,medicine.disease ,lung cancer ,Cyclooxygenase 2 ,Cancer research ,RNA - Abstract
Cyclooxygenase-2 (COX-2) plays an important role in lung cancer development and progression. Using streptavidin-agarose pulldown and proteomics assay, we identified and validated Ku80, a dimer of Ku participating in the repair of broken DNA double strands, as a new binding protein of the COX-2 gene promoter. Overexpression of Ku80 up-regulated COX-2 promoter activation and COX-2 expression in lung cancer cells. Silencing of Ku80 by siRNA down-regulated COX-2 expression and inhibited tumor cell growth in vitro and in a xenograft mouse model. Ku80 knockdown suppressed phosphorylation of ERK, resulting in an inactivation of the MAPK pathway. Moreover, CBP, a transcription co-activator, interacted with and acetylated Ku80 to co-regulate the activation of COX-2 promoter. Overexpression of CBP increased Ku80 acetylation, thereby promoting COX-2 expression and cell growth. Suppression of CBP by a CBP-specific inhibitor or siRNA inhibited COX-2 expression as well as tumor cell growth. Tissue microarray immunohistochemical analysis of lung adenocarcinomas revealed a strong positive correlation between levels of Ku80 and COX-2 and clinicopathologic variables. Overexpression of Ku80 was associated with poor prognosis in patients with lung cancers. We conclude that Ku80 promotes COX-2 expression and tumor growth and is a potential therapeutic target in lung cancer.
- Published
- 2015
20. KLF4 downregulates hTERT expression and telomerase activity to inhibit lung carcinoma growth
- Author
-
Yunlu Jia, Xiangsheng Xiao, Wenxian Hu, Wenlin Huang, Changlin Zhang, Tianze Liu, Zhenglong Yu, Wu Guo Deng, Kezhen Lv, Bing Sun, Linbo Wang, Yixin Li, Yongxia Chen, Xiao Luo, and Wenbin Li
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Male ,Telomerase ,Lung Neoplasms ,0302 clinical medicine ,Surgical oncology ,Promoter Regions, Genetic ,Middle Aged ,KLF4 ,Gene Expression Regulation, Neoplastic ,Oncology ,030220 oncology & carcinogenesis ,embryonic structures ,Female ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,hTERT ,Protein Binding ,Research Paper ,MAP Kinase Signaling System ,Transplantation, Heterologous ,Kruppel-Like Transcription Factors ,Down-Regulation ,Mice, Nude ,cancer prognosis ,Disease-Free Survival ,03 medical and health sciences ,Kruppel-Like Factor 4 ,stomatognathic system ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Telomerase reverse transcriptase ,Lung cancer ,Cell Proliferation ,Oncogene ,business.industry ,fungi ,Cancer ,medicine.disease ,MAPK ,lung cancer ,030104 developmental biology ,A549 Cells ,Immunology ,Cancer research ,sense organs ,business - Abstract
// Wenxian Hu 1, * , Yunlu Jia 1, * , Xiangsheng Xiao 2 , Kezhen Lv 3 , Yongxia Chen 1 , Linbo Wang 1 , Xiao Luo 1 , Tianze Liu 2 , Wenbin Li 2 , Yixin Li 2 , Changlin Zhang 2 , Zhenglong Yu 4 , Wenlin Huang 2, 5 , Bing Sun 4 , Wu-guo Deng 2, 5 1 Department of Surgical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China 2 Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China 3 Department of Breast Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China 4 Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China 5 State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China * These authors contributed equally to this work Correspondence to: Wenxian Hu, email: 13967169904@163.com Bing Sun, email: dysunbing@163.com Wu-guo Deng, email: dengwg@sysucc.org.cn Keywords: KLF4, hTERT, MAPK, lung cancer, cancer prognosis Received: January 07, 2016 Accepted: April 16, 2016 Published: May 02, 2016 ABSTRACT Kruppel-like factor 4 (KLF4) is a transcription factor that contributes to diverse cellular processes and serves as a tumor suppressor or oncogene in various cancers. Previously, we have reported on the tumor suppressive function of KLF4 in lung cancer; however, its precise regulatory mechanism remains elusive. In this study, we found that KLF4 negatively regulated hTERT expression and telomerase activity in lung cancer cell lines and a mouse model. In addition, the KLF4 and hTERT expression levels were significantly related to the clinicopathological features of lung cancer patients. Promoter reporter analyses revealed the decreased hTERT promoter activity in cells infected with Ad-KLF4, and chromatin immunoprecipitation analysis demonstrated that endogenous KLF4 directly bound to the promoter region of hTERT. Furthermore, the MAPK signaling pathway was revealed to be involved in the KLF4/hTERT modulation pathway. Forced expression of KLF4 profoundly attenuated lung cell proliferation and cancer formation in a murine model. Moreover, hTERT overexpression can partially rescue the KLF4-mediated suppressive effect in lung cancer cells. Taken together, these results demonstrate that KLF4 suppresses lung cancer growth by inhibiting hTERT and MAPK signaling. Additionally, the KLF4/hTERT/MAPK pathway is a potential new therapeutic target for human lung cancer.
- Published
- 2016
21. Abstract A46: KLF4 overcomes tamoxifen resistance by suppressing MAPK signaling pathway in breast cancer
- Author
-
Yunlu Jia and Wang Linbo
- Subjects
MAPK/ERK pathway ,Cancer Research ,Gene knockdown ,business.industry ,Cell growth ,Cancer ,medicine.disease ,Breast cancer ,stomatognathic system ,Oncology ,KLF4 ,embryonic structures ,Cancer research ,Medicine ,sense organs ,Signal transduction ,skin and connective tissue diseases ,business ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Tamoxifen ,medicine.drug - Abstract
Krüppel-like factor 4 (KLF4) has critical roles in breast cancer development and progression and several solid tumors. Tamoxifen (TAM) resistance represents a daunting challenge to the successful treatment of breast cancer. KLF4 expression, function, and regulation in the efficacy of TAM therapy in breast cancer have yet to be demonstrated. Here, we investigated the clinical significance and biologic effects of KLF4 in breast cancer. Firstly, higher expression of KLF4 was correlated with increased TAM sensitivity in breast cells, and analysis of GEO datasets indicated that KLF4 expression was positively correlated with ERa and enhanced expression of KLF4 sensitized breast cancer patients to endocrine therapy. Knockdown of KLF4 in MCF-7 and BCAP37 cells led to increased TAM resistance, while ectopic KLF4 expression promoted the responsiveness of MCF-7/TAM and T47D cells to TAM. Secondly, ectopic KLF4 overexpression suppressed MCF-7/TAM cell growth, invasion, and migration ability. Besides, KLF4 expression was downregulated in breast cancer tumor tissues and high expression of KLF4 linked to favorable outcome. Mechanistically, KLF4 may enhance the responsiveness of breast cancer cells to TAM through suppressing mitogen-activated protein kinase (MAPK) signaling pathway. We found that ERK and P38 were relatively activated in MCF-7/TAM compared with MCF-7, and treatment with MAPK-specific inhibitors can significantly suppress cell viability. Knockdown of KLF4 activated ERK and P38 and drove MCF-7 cells to become resistant to TAM. Conversely, overexpression of KLF4 in MCF-7/TAM cells suppressed ERK and P38 signaling and resulted in enhanced sensitivity to TAM. Therefore, our findings suggest that KLF4 contributes to TAM efficiency in breast cancer via phosphorylation modification of ERK and P38 signaling. Collectively, this study highlighted the significance of KLF4/MAPK signal interaction in regulating TAM resistance of breast cancer, and suggested that targeting the KLF4/MAPK signaling may be a potential therapeutic strategy for breast cancer treatment, especially for TAM-resistant patients. Note: This abstract was not presented at the conference. Citation Format: Yunlu Jia, Wang Linbo. KLF4 overcomes tamoxifen resistance by suppressing MAPK signaling pathway in breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A46.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.