1
|
Wen P, Jiang D, Qu F, Wang G, Zhang N, Shao Q, Huang Y, Li S, Wang L, Zeng X. PFDN5 plays a dual role in breast cancer and regulates tumor immune microenvironment: Insights from integrated bioinformatics analysis and experimental validation. Gene 2025; 933:149000. [PMID: 39396557 DOI: 10.1016/j.gene.2024.149000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Although the prognosis for patients with breast cancer has improved, breast cancer remains the leading cause of death for women worldwide. Prefoldin 5 (PFDN5), as a subunit of the prefoldin complex, plays a vital role in aiding the correct folding of newly synthesized proteins. However, the exact impact of PFDN5 on breast cancer development and its prognostic implications remain unclear. METHODS We conducted bioinformatics analysis to investigate the correlation between PFDN5 and patient survival, as well as various clinicopathological characteristics in breast cancer. Additionally, various assays were employed to validate the biological functions of PFDN5 in breast cancer. Finally, RNA sequencing (RNA-seq) was utilized to investigate the molecular mechanisms associated with PFDN5. RESULTS Compared to normal tissues, PFDN5 exhibited lower expression levels in breast cancer tissues, and lower expression of PFDN5 is associated with poorer prognosis. PFDN5 led to G2/M phase arrest in the cell cycle and reduced proliferative potential in breast cancer cells. However, PFDN5 also promoted migration and invasion of breast cancer cells. Also, RNA-seq analysis revealed an involvement of PFDN5 in the cell cycle and TGF-β signaling pathway. Furthermore, PFDN5 had a significant impact on tumor immune microenvironment by promoting macrophage polarization towards the M1 phenotype and exhibited a positive correlation with CD8+ T cell infiltration levels. CONCLUSIONS PFDN5 plays a dual role in breast cancer and serves as a key factor in tumor immune microenvironment. Therefore, PFDN5 holds promise as a valuable biomarker for predicting both metastatic and prognosis in breast cancer.
Collapse
Affiliation(s)
- Ping Wen
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Dongping Jiang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Fanli Qu
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Guanwen Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ningning Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Qing Shao
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Yuxin Huang
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Sisi Li
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Long Wang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaohua Zeng
- Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing 400030, China; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China; Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing 400030, China.
| |
Collapse
|
2
|
ZHU ZHAOYING, HU YANJIA, YE FENG, TENG HAIBO, YOU GUOLIANG, ZENG YUNHUI, TIAN MENG, XU JIANGUO, LI JIN, LIU ZHIYONG, LIU HAO, ZHENG NIANDONG. IKIP downregulates THBS1/FAK signaling to suppress migration and invasion by glioblastoma cells. Oncol Res 2024; 32:1173-1184. [PMID: 38948026 PMCID: PMC11211642 DOI: 10.32604/or.2024.042456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 02/04/2024] [Indexed: 07/02/2024] Open
Abstract
Background Inhibitor of NF-κB kinase-interacting protein (IKIP) is known to promote proliferation of glioblastoma (GBM) cells, but how it affects migration and invasion by those cells is unclear. Methods We compared levels of IKIP between glioma tissues and normal brain tissue in clinical samples and public databases. We examined the effects of IKIP overexpression and knockdown on the migration and invasion of GBM using transwell and wound healing assays, and we compared the transcriptomes under these different conditions to identify the molecular mechanisms involved. Results Based on data from our clinical samples and from public databases, IKIP was overexpressed in GBM tumors, and its expression level correlated inversely with survival. IKIP overexpression in GBM cells inhibited migration and invasion in transwell and wound healing assays, whereas IKIP knockdown exerted the opposite effects. IKIP overexpression in GBM cells that were injected into mouse brain promoted tumor growth but inhibited tumor invasion of surrounding tissue. The effects of IKIP were associated with downregulation of THBS1 mRNA and concomitant inhibition of THBS1/FAK signaling. Conclusions IKIP inhibits THBS1/FAK signaling to suppress migration and invasion of GBM cells.
Collapse
Affiliation(s)
- ZHAOYING ZHU
- Department of Neurosurgery, The Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - YANJIA HU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - FENG YE
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - HAIBO TENG
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - GUOLIANG YOU
- Department of Neurosurgery, The Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - YUNHUI ZENG
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - MENG TIAN
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - JIANGUO XU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - JIN LI
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - ZHIYONG LIU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - HAO LIU
- Department of Neurosurgery, Sichuan University West China Hospital, Chengdu, China
| | - NIANDONG ZHENG
- Department of Neurosurgery, The Affiliated Hospital of Southwestern Medical University, Luzhou, China
| |
Collapse
|
3
|
Jia Y, Feng G, Chen S, Li W, Jia Z, Wang J, Li H, Hong S, Dai F. Metabolic Heterogeneity of Tumor Cells and its Impact on Colon Cancer Metastasis: Insights from Single-Cell and Bulk Transcriptome Analyses. J Cancer 2024; 15:4175-4196. [PMID: 38947396 PMCID: PMC11212087 DOI: 10.7150/jca.94630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/17/2024] [Indexed: 07/02/2024] Open
Abstract
Background: Metabolic reprogramming plays a crucial role in the development of colorectal cancer (CRC), influencing tumor heterogeneity, the tumor microenvironment, and metastasis. While the interaction between metabolism and CRC is critical for developing personalized treatments, gaps remain in understanding how tumor cell metabolism affects prognosis. Our study introduces novel insights by integrating single-cell and bulk transcriptome analyses to explore the metabolic landscape within CRC cells and its mechanisms influencing disease progression. This approach allows us to uncover metabolic heterogeneity and identify specific metabolic genes impacting metastasis, which have not been thoroughly examined in previous studies. Methods: We sourced microarray and single-cell RNA sequencing datasets from the Gene Expression Omnibus (GEO) and bulk sequencing data for CRC from The Cancer Genome Atlas (TCGA). We employed Gene Set Variation Analysis (GSVA) to assess metabolic pathway activity, consensus clustering to identify CRC-specific transcriptome subtypes in bulkseq, and rigorous quality controls, including the exclusion of cells with high mitochondrial gene expression in scRNA seq. Advanced analyses such as AUCcell, infercnvCNV, Non-negative Matrix Factorization (NMF), and CytoTRACE were utilized to dissect the cellular landscape and evaluate pathway activities and tumor cell stemness. The hdWGCNA algorithm helped identify prognosis-related hub genes, integrating these findings using a random forest machine learning model. Results: Kaplan-Meier survival curves identified 21 significant metabolic pathways linked to prognosis, with consensus clustering defining three CRC subtypes (C3, C2, C1) based on metabolic activity, which correlated with distinct clinical outcomes. The metabolic activity of the 13 cell subpopulations, particularly the epithelial cell subpopulation with active metabolic levels, was evaluated using AUCcell in scRNA seq. To further analyze tumor cells using infercnv, NMF disaggregated these cells into 10 cellular subpopulations. Among these, the C2 subpopulation exhibited higher stemness and tended to have a poorer prognosis compared to C6 and C0. Conversely, the C8, C3, and C1 subpopulations demonstrated a higher level of the five metabolic pathways, and the C3 and C8 subpopulations tended to have a more favorable prognosis. hdWGCNA identified 20 modules, from which we selected modules primarily expressed in high metabolic tumor subgroups and highly correlated with clinical information, including blue and cyan. By applying variable downscaling of RF to a total of 50 hub genes, seven gene signatures were obtained. Furthermore, molecules that were validated to be protective in GEO were screened alongside related molecules, resulting in the identification of prognostically relevant molecules such as UQCRFS1 and GRSF1. Additionally, the expression of GRSF1 was examined in colon cancer cell lines using qPCR and phenotypically verified by in vitro experiments. Conclusion: Our findings emphasize that high activity in specific metabolic pathways, including pyruvate metabolism and the tricarboxylic acid cycle, correlates with improved colon cancer outcomes, presenting new avenues for metabolic-based therapies. The identification of hub genes like GRSF1 and UQCRFS1 and their link to favorable metabolic profiles offers novel insights into tumor neovascularization and metastasis, with significant clinical implications for targeting metabolic pathways in CRC therapy.
Collapse
Affiliation(s)
- Yiwen Jia
- Department of Gastroenterology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, China
| | - Guangming Feng
- Department of Gastroenterology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, China
| | - Siyuan Chen
- Department of Gastroenterology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, China
| | - Wenhao Li
- Department of Pulmonology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zeguo Jia
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jian Wang
- Department of Pathology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, 230032, China
| | - Hongxia Li
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, 230032, China
| | - Shaocheng Hong
- Department of Gastroenterology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, China
| | - Fu Dai
- Department of Gastroenterology, The Third Affiliated Hospital of Anhui Medical University (Hefei first people's Hospital), Hefei, China
| |
Collapse
|
4
|
Phan TTT, Truong NV, Wu WG, Su YC, Hsu TS, Lin LY. Tumor suppressor p53 mediates interleukin-6 expression to enable cancer cell evasion of genotoxic stress. Cell Death Discov 2023; 9:340. [PMID: 37696858 PMCID: PMC10495329 DOI: 10.1038/s41420-023-01638-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
The tumor suppressor p53 primarily functions as a mediator of DNA damage-induced cell death, thereby contributing to the efficacy of genotoxic anticancer therapeutics. Here, we show, on the contrary, that cancer cells can employ genotoxic stress-induced p53 to acquire treatment resistance through the production of the pleiotropic cytokine interleukin (IL)-6. Mechanistically, DNA damage, either repairable or irreparable, activates p53 and stimulates Caspase-2-mediated cleavage of its negative regulator mouse double minute 2 (MDM2) creating a positive feedback loop that leads to elevated p53 protein accumulation. p53 transcriptionally controls the major adenosine triphosphate (ATP) release channel pannexin 1 (Panx1), which directs IL-6 induction via a mechanism dependent on the extracellular ATP-activated purinergic P2 receptors as well as their downstream intracellular calcium (iCa2+)/PI3K/Akt/NF-ĸB signaling pathway. Thus, p53 silencing impairs Panx1 and IL-6 expression and renders cancer cells sensitive to genotoxic stress. Moreover, we confirm that IL-6 hampers the effectiveness of genotoxic anticancer agents by mitigating DNA damage, driving the expression of anti-apoptotic Bcl-2 family genes, and maintaining the migratory and invasive properties of cancer cells. Analysis of patient survival and relevant factors in lung cancer and pan-cancer cohorts supports the prognostic and clinical values of Panx1 and IL-6. Notably, IL-6 secreted by cancer cells during genotoxic treatments promotes the polarization of monocytic THP-1-derived macrophages into an alternative (M2-like) phenotype that exhibits impaired anti-survival activities but enhanced pro-metastatic effects on cancer cells as compared to nonpolarized macrophages. Our study reveals the precise mechanism for genotoxic-induced IL-6 and suggests that targeting p53-mediated IL-6 may improve the responsiveness of cancer cells to genotoxic anticancer therapy.
Collapse
Affiliation(s)
- Trinh T T Phan
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Nam V Truong
- Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Wen-Guey Wu
- Institute of Bioinformatics and Structural Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Yi-Chun Su
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC.
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan ROC.
| |
Collapse
|
5
|
Sun R, He J, Xiang Q, Feng Y, Gong Y, Ning Y, Deng C, Sun K, Zhang M, Cheng Z, Le X, Xiong Q, Dai F, Wu Y, Xiang T. NTF4 plays a dual role in breast cancer in mammary tumorigenesis and metastatic progression. Int J Biol Sci 2023; 19:641-657. [PMID: 36632451 PMCID: PMC9830504 DOI: 10.7150/ijbs.79435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
Breast cancer metastasis can happen even when the primary tumor is relatively small. But the mechanism for such early metastasis is poorly understood. Herein, we report that neurotrophin 4 (NTF4) plays a dual role in breast cancer proliferation and metastasis. Clinical data showed high levels of NTF4, especially in the early stage, to be associated with poor clinical outcomes, supporting the notion that metastasis, rather than primary cancer, was the major determinant of breast cancer mortality for patients. NTF4 promoted epithelial-mesenchymal transition (EMT), cell motility, and invasiveness of breast cancer cells in vitro and in vivo. Interestingly, NTF4 inhibited cell proliferation while promoting cellular apoptosis in vitro and inhibited xenograft tumorigenicity in vivo. Mechanistically, NTF4 elicited its pro-metastatic effects by activating PRKDC/AKT and ANXA1/NF-κB pathways to stabilize SNAIL protein, therefore decreasing the level of E-cadherin. Conversely, NTF4 increased ANXA1 phosphorylation and sumoylation and the interaction with importin β, leading to nuclear import and retention of ANXA1, which in turn activates the caspase-3 apoptosis cascade. Our findings identified an unexpected dual role for NTF4 in breast cancer which contributes to early metastasis of the disease. Therefore, NTF4 may serve as a prognostic marker and a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Ran Sun
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.,Department of Oncology, Jiulongpo People's Hospital, Chongqing 400050, China
| | - Jin He
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qin Xiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yixiao Feng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yijia Gong
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yijiao Ning
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chaoqun Deng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kexin Sun
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mingjun Zhang
- Department of Laboratory Medicine, Jiulongpo People's Hospital, Chongqing 400050, China
| | - Zhaobo Cheng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xin Le
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qi Xiong
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fengsheng Dai
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yongzhong Wu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.,✉ Corresponding authors: Tingxiu Xiang. Tel: (023) 65079282. E-mail: and Yongzhong Wu. E-mail:
| | - Tingxiu Xiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.,✉ Corresponding authors: Tingxiu Xiang. Tel: (023) 65079282. E-mail: and Yongzhong Wu. E-mail:
| |
Collapse
|
6
|
Rass M, Gizler L, Bayersdorfer F, Irlbeck C, Schramm M, Schneuwly S. The Drosophila functional Smad suppressing element fuss, a homologue of the human Skor genes, retains pro-oncogenic properties of the Ski/Sno family. PLoS One 2022; 17:e0262360. [PMID: 35030229 PMCID: PMC8759651 DOI: 10.1371/journal.pone.0262360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 12/21/2021] [Indexed: 11/19/2022] Open
Abstract
Over the years Ski and Sno have been found to be involved in cancer progression e.g. in oesophageal squamous cell carcinoma, melanoma, oestrogen receptor-positive breast carcinoma, colorectal carcinoma, and leukaemia. Often, their prooncogenic features have been linked to their ability of inhibiting the anti-proliferative action of TGF-ß signalling. Recently, not only pro-oncogenic but also anti-oncogenic functions of Ski/Sno proteins have been revealed. Besides Ski and Sno, which are ubiquitously expressed other members of Ski/Sno proteins exist which show highly specific neuronal expression, the SKI Family Transcriptional Corepressors (Skor). Among others Skor1 and Skor2 are involved in the development of Purkinje neurons and a mutation of Skor1 has been found to be associated with restless legs syndrome. But neither Skor1 nor Skor2 have been reported to be involved in cancer progression. Using overexpression studies in the Drosophila eye imaginal disc, we analysed if the Drosophila Skor homologue Fuss has retained the potential to inhibit differentiation and induce increased proliferation. Fuss expressed in cells posterior to the morphogenetic furrow, impairs photoreceptor axon pathfinding and inhibits differentiation of accessory cells. However, if its expression is induced prior to eye differentiation, Fuss might inhibit the differentiating function of Dpp signalling and might maintain proliferative action of Wg signalling, which is reminiscent of the Ski/Sno protein function in cancer.
Collapse
Affiliation(s)
- Mathias Rass
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
- * E-mail:
| | - Laura Gizler
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Florian Bayersdorfer
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Christoph Irlbeck
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Matthias Schramm
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Stephan Schneuwly
- Department of Developmental Biology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
7
|
Huang L, Zhu L, Pan S, Xu J, Xie M, Wang W, Xia G. Circ_0029803 serves as the sponge of miR-216b-5p to promote the progression of colorectal cancer by regulating SKIL expression. World J Surg Oncol 2021; 19:268. [PMID: 34479589 PMCID: PMC8417978 DOI: 10.1186/s12957-021-02368-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/10/2021] [Indexed: 01/17/2023] Open
Abstract
Background Circular RNA 0029803 (circ_0029803) was found to be upregulated in colorectal cancer (CRC) tissues, but its function and underlying molecular mechanism are not studied in CRC. Methods The expression levels of circ_0029803, microRNA-216b-5p (miR-216b-5p), and ski-oncogene-like (SKIL) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). RNase R treatment was used to affirm the existence of circ_0029803. Cell proliferation, apoptosis, migration, and invasion were assessed by colony formation, flow cytometry, and Transwell assays, respectively. A glucose and lactate assay kit was used to detect glucose consumption and lactate production. Western blot was applied to analyze the levels of all proteins. Dual-luciferase reporter assay was performed to assess the relationship between miR-216b-5p and circ_0029803 or SKIL. Tumor xenograft models were established to elucidate the effect of circ_0029803 in vivo. Results Circ_0029803 expression was enhanced in CRC tissues and cells, and the 5-year overall survival rate of patients with high circ_0029803 expression was substantially reduced. Circ_0029803 depletion retarded proliferation, migration, invasion, EMT and glycolysis of CRC cells in vitro as well as the tumor growth in vivo. Mechanically, circ_0029803 could serve as miR-216b-5p sponge to regulate its expression, and miR-216b-5p knockdown reversed the inhibition of si-circ_0029803 on the malignant behaviors of CRC cells. Additionally, as the target mRNA of miR-216b-5p, SKIL could counteract the inhibitory effect of miR-216b-5p on the development of CRC cells. Importantly, silencing circ_0029803 reduced SKIL expression via sponging miR-216b-5p. Conclusion Circ_0029803 knockdown hindered proliferation, migration, invasion, EMT, and glycolysis and promoted apoptosis in CRC cells by modulating the miR-216b-5p/SKIL axis.
Collapse
Affiliation(s)
- Linfei Huang
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China
| | - Lei Zhu
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China
| | - Sheng Pan
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China
| | - Jing Xu
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China
| | - Miao Xie
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China
| | - Ganlin Xia
- Department of Gastrointestinal Surgery, Wuhan Puren Hospital Affiliated to Wuhan University of Science and Technology, No.1 Benxi Street, Qingshan District, Wuhan City, 430080, Hubei Province, People's Republic of China.
| |
Collapse
|
8
|
SMAD-oncoprotein interplay: Potential determining factors in targeted therapies. Biochem Pharmacol 2020; 180:114155. [DOI: 10.1016/j.bcp.2020.114155] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022]
|
9
|
Pal A, Ashworth JC, Collier P, Probert C, Jones S, Leza EP, Meakin ML, A. Ritchie A, Onion D, Clarke PA, Allegrucci C, Grabowska AM. A 3D Heterotypic Breast Cancer Model Demonstrates a Role for Mesenchymal Stem Cells in Driving a Proliferative and Invasive Phenotype. Cancers (Basel) 2020; 12:E2290. [PMID: 32824003 PMCID: PMC7465555 DOI: 10.3390/cancers12082290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Accepted: 08/10/2020] [Indexed: 01/14/2023] Open
Abstract
Previous indirect 2D co-culture studies have demonstrated that mesenchymal stem cells (MSCs) promote breast cancer (BC) progression through secretion of paracrine factors including growth factors, cytokines and chemokines. In order to investigate this aspect of the tumour microenvironment in a more relevant 3D co-culture model, spheroids incorporating breast cancer cells (BCCs), both cell lines and primary BCCs expanded as patient-derived xenografts, and MSCs were established. MSCs in co-cultures were shown to enhance proliferation of estrogen receptor (ER)/progesterone receptor (PR)-positive BCCs. In addition, co-culture resulted in downregulation of E-cadherin in parallel with upregulation of the epithelial-mesenchymal transition (EMT)-relation transcription factor, SNAIL. Cytoplasmic relocalization of ski-related novel protein N (SnON), a negative regulator of transforming growth factor-beta (TGF-β) signalling, and of β-catenin, involved in a number of pathways including Wnt signalling, was also observed in BCCs in co-cultures in contrast to monocultures. In addition, the β-catenin inhibitor, 3-[[(4-methylphenyl)sulfonyl]amino]-benzoic acid methyl ester (MSAB), mediated reduced growth and invasion in the co-cultures. This study highlights the potential role for SnON as a biomarker for BC invasiveness, and the importance of interactions between TGF-β and Wnt signalling, involving SnON. Such pathways may contribute towards identifying possible targets for therapeutic intervention in BC patients.
Collapse
Affiliation(s)
- Amarnath Pal
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Jennifer C. Ashworth
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Pamela Collier
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Catherine Probert
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Sal Jones
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Eduardo Pernaut Leza
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Marian L. Meakin
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Alison A. Ritchie
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - David Onion
- Flow Cytometry Facility, School of Life Sciences, University of Nottingham, Nottingham NG7 2UH, UK;
| | - Philip A Clarke
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| | - Cinzia Allegrucci
- SVMS, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Anna M. Grabowska
- Ex Vivo Cancer Pharmacology Centre, Division of Cancer and Stem Cells, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.P.); (J.C.A.); (P.C.); (C.P.); (S.J.); (E.P.L.); (M.L.M.); (A.A.R.); (P.A.C.)
| |
Collapse
|
10
|
Shao H, Zhu Q, Lu H, Chang A, Gao C, Zhou Q, Luo K. HEXIM1 controls P-TEFb processing and regulates drug sensitivity in triple-negative breast cancer. Mol Biol Cell 2020; 31:1867-1878. [PMID: 32520633 PMCID: PMC7525814 DOI: 10.1091/mbc.e19-12-0704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 11/16/2022] Open
Abstract
The positive transcription elongation factor b (P-TEFb), composed of CDK9 and cyclin T, stimulates transcriptional elongation by RNA polymerase (Pol) II and regulates cell growth and differentiation. Recently, we demonstrated that P-TEFb also controls the expression of EMT regulators to promote breast cancer progression. In the nucleus, more than half of P-TEFb are sequestered in the inactive-state 7SK snRNP complex. Here, we show that the assembly of the 7SK snRNP is preceded by an intermediate complex between HEXIM1 and P-TEFb that allows transfer of the kinase active P-TEFb from Hsp90 to 7SK snRNP for its suppression. Down-regulation of HEXIM1 locks P-TEFb in the Hsp90 complex, keeping it in the active state to enhance breast cancer progression, but also rendering the cells highly sensitive to Hsp90 inhibition. Because HEXIM1 is often down-regulated in human triple-negative breast cancer (TNBC), these cells are particularly sensitive to Hsp90 inhibition. Our study provides a mechanistic explanation for the increased sensitivity of TNBC to Hsp90 inhibition.
Collapse
Affiliation(s)
- Hengyi Shao
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Huasong Lu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Amanda Chang
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Carol Gao
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Qiang Zhou
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| |
Collapse
|
11
|
Li DD, Deng L, Hu SY, Zhang FL, Li DQ. SH3BGRL2 exerts a dual function in breast cancer growth and metastasis and is regulated by TGF-β1. Am J Cancer Res 2020; 10:1238-1254. [PMID: 32368399 PMCID: PMC7191107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/09/2020] [Indexed: 06/11/2023] Open
Abstract
SH3 domain-binding glutamic acid-rich-like protein 2 (SH3BGRL2) is a poorly defined member of the SH3BGR gene family with potential roles in cell differentiation and tissue development. Here, we report for the first time that SH3BGRL2 exerts a dual function in breast tumor growth and metastasis. SH3BGRL2 was downregulated in a subset of primary breast tumors, and suppressed breast cancer cell proliferation and colony formation in vitro and xenograft tumor growth in vivo. Strikingly, SH3BGRL2 enhanced breast cancer cell migratory, invasive, and lung metastatic capacity. Mechanistic investigations revealed that SH3BGRL2 interacted with and transcriptionally repressed spectrin alpha, non-erythrocytic 1 (SPTAN1) and spectrin beta, non-erythrocytic 1 (SPTBN1), two important cytoskeletal proteins. Functional rescue assays further demonstrated that depletion of SH3BGRL2 reduced breast cancer cell invasive potential, which was partially rescued by knockdown of SPTAN1 and SPTBN1 using specific small interfering RNA. Moreover, transforming growth factor-β1 (TGF-β1) transcriptionally activated SH3BGRL2 expression in breast cancer cells through the canonical TGF-β receptor-Smad pathway. Collectively, these results establish a dual function of SH3BGRL2 in breast cancer growth and metastasis and uncover SH3BGRL2 as a downstream target of the TGF-β1 signaling pathway in breast cancer cells.
Collapse
Affiliation(s)
- Dou-Dou Li
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan UniversityShanghai 200032, China
- Cancer Institute, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Ling Deng
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan UniversityShanghai 200032, China
| | - Shu-Yuan Hu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan UniversityShanghai 200032, China
| | - Fang-Lin Zhang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan UniversityShanghai 200032, China
- Cancer Institute, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| | - Da-Qiang Li
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan UniversityShanghai 200032, China
- Cancer Institute, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Shanghai Key Laboratory of Breast Cancer, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| |
Collapse
|
12
|
Zhang KJ, Hu Y, Luo N, Li X, Chen FY, Yuan JQ, Guo L. miR‑574‑5p attenuates proliferation, migration and EMT in triple‑negative breast cancer cells by targeting BCL11A and SOX2 to inhibit the SKIL/TAZ/CTGF axis. Int J Oncol 2020; 56:1240-1251. [PMID: 32319565 DOI: 10.3892/ijo.2020.4995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/21/2019] [Indexed: 11/05/2022] Open
Abstract
Triple‑negative breast cancer (TNBC) is a subtype of breast cancer with a high degree of malignancy. TNBC is prone to distant metastasis and has a poor prognosis. A number of TNBC‑related microRNAs (miRNAs) have been studied and identified. However, the detailed roles of miR‑574‑5p in TNBC remain poorly understood. miR‑574‑5p, SRY (sex determining region Y)‑box 2 (SOX2), B‑cell lymphoma/leukaemia 11A (BCL11A), SKI like proto‑oncogene (SKIL) and epithelial‑mesenchymal transition (EMT)‑related miRNAs and proteins were measured by reverse transcription‑quantitative PCR and western blotting analysis, respectively. A luciferase reporter assay was employed to validate the direct targeting of SOX2 and BCL11A by miR‑574‑5p. MTT, colony formation and Transwell assays were performed to analyse the biological functions of miR‑574‑5p in TNBC cells. A nude mouse xenograft model was used to verify the effects of miR‑574‑5p on the tumorigenesis of TNBC in vivo. The results demonstrated that miR‑574‑5p levels were decreased in breast cancer tissues and cells. miR‑574‑5p repressed proliferation, migration and EMT in TNBC cells. Further experiments confirmed that miR‑574‑5p reduced tumour size and metastasis in vivo. miR‑574‑5p targeted BCL11A and SOX2 to inhibit the SKIL/transcriptional co‑activator with PDZ‑binding motif/connective tissue growth factor axis, and the inhibitory effect of miR‑574‑5p in TNBC cells was at least partly dependent on SOX2 and BCL11A. In addition, the regulation of downstream oncogenes by SOX2 was dependent on BCL11A. To the best of our knowledge, this is the first study to report the association between the miR‑574‑5p/BCL11A/SOX2 axis and the tumorigenesis of TNBC, which provides a new mechanism for understanding the progression of TNBC.
Collapse
Affiliation(s)
- Ke-Jing Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu Hu
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Na Luo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xin Li
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fei-Yu Chen
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jia-Qi Yuan
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lei Guo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
13
|
Are Ski and SnoN Involved in the Tumorigenesis of Oral Squamous Cell Carcinoma Through Smad4? Appl Immunohistochem Mol Morphol 2019; 27:626-630. [DOI: 10.1097/pai.0000000000000667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Zhu Q, Chang A, Xu A, Luo K. The regulatory protein SnoN antagonizes activin/Smad2 protein signaling and thereby promotes adipocyte differentiation and obesity in mice. J Biol Chem 2018; 293:14100-14111. [PMID: 30030373 DOI: 10.1074/jbc.ra118.003678] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/17/2018] [Indexed: 12/25/2022] Open
Abstract
Ski-related oncogene SnoN (SnoN or SKIL) regulates multiple signaling pathways in a tissue- and developmental stage-dependent manner and has broad functions in embryonic angiogenesis, mammary gland alveologenesis, cancer, and aging. Here, we report that SnoN also plays a critical role in white adipose tissue (WAT) development by regulating mesenchymal stem cell (MSC) self-renewal and differentiation. We found that SnoN promotes MSC differentiation in the adipocyte lineage by antagonizing activin A/Smad2, but not TGFβ/Smad3 signaling. Mice lacking SnoN or expressing a mutant SnoN defective in binding to the Smads were protected from high-fat diet-induced obesity and insulin resistance, and MSCs lacking a functional SnoN exhibited defective differentiation. We further demonstrated that activin, via Smad2, appears to be the major regulator of WAT development in vivo We also noted that activin A is abundantly expressed in WAT and adipocytes through an autocrine mechanism and promotes MSC self-renewal and inhibits adipogenic differentiation by inducing expression of the gene encoding the homeobox transcription factor Nanog. Of note, SnoN repressed activin/Smad2 signaling and activin A expression, enabling expression of adipocyte-specific transcription factors and promoting adipogenic differentiation. In conclusion, our study has revealed that SnoN plays an important in vivo role in adipocyte differentiation and WAT development in vivo by decreasing activity in the activin/Smad2 signaling pathway.
Collapse
Affiliation(s)
- Qingwei Zhu
- From the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Amanda Chang
- From the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Albert Xu
- the Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, California 94158-2140
| | - Kunxin Luo
- From the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, .,the Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 and
| |
Collapse
|
15
|
Sun R, Xie HY, Qian JX, Huang YN, Yang F, Zhang FL, Shao ZM, Li DQ. FBXO22 Possesses Both Protumorigenic and Antimetastatic Roles in Breast Cancer Progression. Cancer Res 2018; 78:5274-5286. [PMID: 29945959 DOI: 10.1158/0008-5472.can-17-3647] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 06/04/2018] [Accepted: 06/22/2018] [Indexed: 11/16/2022]
Abstract
The molecular underpinnings behind malignant progression of breast cancer from a localized lesion to an invasive and ultimately metastatic disease are incompletely understood. Here, we report that F-box only protein 22 (FBXO22) plays a dual role in mammary tumorigenesis and metastasis. FBXO22 was upregulated in primary breast tumors and promoted cell proliferation and colony formation in vitro and xenograft tumorigenicity in vivo Surprisingly, FBXO22 suppressed epithelial-mesenchymal transition (EMT), cell motility, and invasiveness in vitro and metastatic lung colonization in vivo Clinical data showed that expression levels of FBXO22 were associated with favorable clinical outcomes, supporting the notion that metastasis, rather than primary cancer, is the major determinant of the mortality of patients with breast cancer. Mechanistic investigations further revealed that FBXO22 elicits its antimetastatic effects by targeting SNAIL, a master regulator of EMT and breast cancer metastasis, for ubiquitin-mediated proteasomal degradation in a glycogen synthase kinase 3β phosphorylation-dependent manner. Importantly, expression of SNAIL rescued FBXO22-mediated suppression of EMT, cell migration, and invasion. A patient-derived tryptophan-to-arginine mutation at residue 52 (W52R) within the F-box domain impaired FBXO22 binding to the SKP1-Cullin1 complex and blocked FBXO22-mediated SNAIL degradation, thus abrogating the ability of FBXO22 to suppress cell migration, invasion, and metastasis. Collectively, these findings uncover an unexpected dual role for FBXO22 in mammary tumorigenesis and metastatic progression and delineate the mechanism of an oncogenic mutation of FBXO22 in breast cancer progression.Significance: These findings highlight the paradoxical roles of FBXO22 in breast cancer, as it promotes breast tumor cell proliferation but prevents EMT and metastasis. Cancer Res; 78(18); 5274-86. ©2018 AACR.
Collapse
Affiliation(s)
- Rui Sun
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Yan Xie
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Xian Qian
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan-Ni Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Lin Zhang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhi-Min Shao
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| | - Da-Qiang Li
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China. .,Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Breast Surgery, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Medical Epigenetics and Metabolism, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
17
|
Seoane J, Gomis RR. TGF-β Family Signaling in Tumor Suppression and Cancer Progression. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022277. [PMID: 28246180 DOI: 10.1101/cshperspect.a022277] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) induces a pleiotropic pathway that is modulated by the cellular context and its integration with other signaling pathways. In cancer, the pleiotropic reaction to TGF-β leads to a diverse and varied set of gene responses that range from cytostatic and apoptotic tumor-suppressive ones in early stage tumors, to proliferative, invasive, angiogenic, and oncogenic ones in advanced cancer. Here, we review the knowledge accumulated about the molecular mechanisms involved in the dual response to TGF-β in cancer, and how tumor cells evolve to evade the tumor-suppressive responses of this signaling pathway and then hijack the signal, converting it into an oncogenic factor. Only through the detailed study of this complexity can the suitability of the TGF-β pathway as a therapeutic target against cancer be evaluated.
Collapse
Affiliation(s)
- Joan Seoane
- Translational Research Program, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Roger R Gomis
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.,Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
18
|
Dichotomous roles of TGF-β in human cancer. Biochem Soc Trans 2017; 44:1441-1454. [PMID: 27911726 DOI: 10.1042/bst20160065] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-β (TGF-β) mediates numerous biological processes, including embryonic development and the maintenance of cellular homeostasis in a context-dependent manner. Consistent with its central role in maintaining cellular homeostasis, inhibition of TGF-β signaling results in disruption of normal homeostatic processes and subsequent carcinogenesis, defining the TGF-β signaling pathway as a tumor suppressor. However, once carcinogenesis is initiated, the TGF-β signaling pathway promotes cancer progression. This dichotomous function of the TGF-β signaling pathway is mediated through altering effects on both the cancer cells, by inducing apoptosis and inhibiting proliferation, and the tumor microenvironment, by promoting angiogenesis and inhibiting immunosurveillance. Current studies support inhibition of TGF-β signaling either alone, or in conjunction with anti-angiogenic therapy or immunotherapy as a promising strategy for the treatment of human cancers.
Collapse
|
19
|
Walldén K, Nyman T, Hällberg BM. SnoN Stabilizes the SMAD3/SMAD4 Protein Complex. Sci Rep 2017; 7:46370. [PMID: 28397834 PMCID: PMC5387736 DOI: 10.1038/srep46370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/15/2017] [Indexed: 12/30/2022] Open
Abstract
TGF-β signaling regulates cellular processes such as proliferation, differentiation and apoptosis through activation of SMAD transcription factors that are in turn modulated by members of the Ski-SnoN family. In this process, Ski has been shown to negatively modulate TGF-β signaling by disrupting active R-SMAD/Co-SMAD heteromers. Here, we show that the related regulator SnoN forms a stable complex with the R-SMAD (SMAD3) and the Co-SMAD (SMAD4). To rationalize this stabilization at the molecular level, we determined the crystal structure of a complex between the SAND domain of SnoN and the MH2-domain of SMAD4. This structure shows a binding mode that is compatible with simultaneous coordination of R-SMADs. Our results show that SnoN, and SMAD heteromers can form a joint structural core for the binding of other transcription modulators. The results are of fundamental importance for our understanding of the molecular mechanisms behind the modulation of TGF-β signaling.
Collapse
Affiliation(s)
- Karin Walldén
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Tomas Nyman
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - B Martin Hällberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.,Röntgen-Ångström-Cluster, Karolinska Institutet Outstation, Centre for Structural Systems Biology, DESY-Campus, 22603 Hamburg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, 22603 Hamburg, Germany
| |
Collapse
|
20
|
Zhu Q, Le Scolan E, Jahchan N, Ji X, Xu A, Luo K. SnoN Antagonizes the Hippo Kinase Complex to Promote TAZ Signaling during Breast Carcinogenesis. Dev Cell 2016; 37:399-412. [PMID: 27237790 DOI: 10.1016/j.devcel.2016.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/01/2016] [Accepted: 05/01/2016] [Indexed: 12/12/2022]
Abstract
SnoN regulates multiple signaling pathways, including TGF-β/Smad and p53, and displays both pro-oncogenic and anti-oncogenic activities in human cancer. We have observed previously that both its intracellular localization and expression levels are sensitive to cell density, suggesting that it may crosstalk with Hippo signaling. Here we report that, indeed, SnoN interacts with multiple components of the Hippo pathway to inhibit the binding of Lats2 to TAZ and the subsequent phosphorylation of TAZ, leading to TAZ stabilization. Consistently, SnoN enhances the transcriptional and oncogenic activities of TAZ, and reducing SnoN decreases TAZ expression as well as malignant progression of breast cancer cells. Interestingly, SnoN itself is downregulated by Lats2 that is activated by the Scribble basolateral polarity protein. Thus, SnoN is a critical component of the Hippo regulatory network that receives signals from the tissue architecture and polarity to coordinate the activity of intracellular signaling pathways.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, MC3204, Berkeley, CA 94720, USA
| | | | - Nadine Jahchan
- ORIC Pharmaceuticals, South San Francisco, CA 94080, USA
| | - Xiaodan Ji
- Departments of Cancer Biology, Cell & Developmental Biology, and Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 190104-6160, USA
| | - Albert Xu
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, MC3204, Berkeley, CA 94720, USA
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, MC3204, Berkeley, CA 94720, USA; Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
21
|
The Disintegrin and Metalloprotease ADAM12 Is Associated with TGF-β-Induced Epithelial to Mesenchymal Transition. PLoS One 2015; 10:e0139179. [PMID: 26407179 PMCID: PMC4583281 DOI: 10.1371/journal.pone.0139179] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/08/2015] [Indexed: 12/21/2022] Open
Abstract
The increased expression of the Disintegrin and Metalloprotease ADAM12 has been associated with human cancers, however its role remain unclear. We have previously reported that ADAM12 expression is induced by the transforming growth factor, TGF-β and promotes TGF-β-dependent signaling through interaction with the type II receptor of TGF-β. Here we explore the implication of ADAM12 in TGF-β-mediated epithelial to mesenchymal transition (EMT), a key process in cancer progression. We show that ADAM12 expression is correlated with EMT markers in human breast cancer cell lines and biopsies. Using a non-malignant breast epithelial cell line (MCF10A), we demonstrate that TGF-β-induced EMT increases expression of the membrane-anchored ADAM12L long form. Importantly, ADAM12L overexpression in MCF10A is sufficient to induce loss of cell-cell contact, reorganization of actin cytoskeleton, up-regulation of EMT markers and chemoresistance. These effects are independent of the proteolytic activity but require the cytoplasmic tail and are specific of ADAM12L since overexpression of ADAM12S failed to induce similar changes. We further demonstrate that ADAM12L-dependent EMT is associated with increased phosphorylation of Smad3, Akt and ERK proteins. Conversely, inhibition of TGF-β receptors or ERK activities reverses ADAM12L-induced mesenchymal phenotype. Together our data demonstrate that ADAM12L is associated with EMT and contributes to TGF-β-dependent EMT by favoring both Smad-dependent and Smad-independent pathways.
Collapse
|
22
|
Caligaris C, Vázquez-Victorio G, Sosa-Garrocho M, Ríos-López DG, Marín-Hernández A, Macías-Silva M. Actin-cytoskeleton polymerization differentially controls the stability of Ski and SnoN co-repressors in normal but not in transformed hepatocytes. Biochim Biophys Acta Gen Subj 2015; 1850:1832-41. [PMID: 26002202 DOI: 10.1016/j.bbagen.2015.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/21/2015] [Accepted: 05/12/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ski and SnoN proteins function as transcriptional co-repressors in the TGF-β pathway. They regulate cell proliferation and differentiation, and their aberrant expression results in altered TGF-β signalling, malignant transformation, and alterations in cell proliferation. METHODS We carried out a comparative characterization of the endogenous Ski and SnoN protein regulation by TGF-β, cell adhesion disruption and actin-cytoskeleton rearrangements between normal and transformed hepatocytes; we also analyzed Ski and SnoN protein stability, subcellular localization, and how their protein levels impact the TGF-β/Smad-driven gene transcription. RESULTS Ski and SnoN protein levels are lower in normal hepatocytes than in hepatoma cells. They exhibit a very short half-life and a nuclear/cytoplasmic distribution in normal hepatocytes opposed to a high stability and restricted nuclear localization in hepatoma cells. Interestingly, while normal cells exhibit a transient TGF-β-induced gene expression, the hepatoma cells are characterized by a strong and sustained TGF-β-induced gene expression. A novel finding is that Ski and SnoN stability is differentially regulated by cell adhesion and cytoskeleton rearrangements in the normal hepatocytes. The inhibition of protein turnover down-regulated both Ski and SnoN co-repressors impacting the kinetic of expression of TGF-β-target genes. CONCLUSION Normal regulatory mechanisms controlling Ski and SnoN stability, subcellular localization and expression are altered in hepatocarcinoma cells. GENERAL SIGNIFICANCE This work provides evidence that Ski and SnoN protein regulation is far more complex in normal than in transformed cells, since many of the normal regulatory mechanisms are lost in transformed cells.
Collapse
Affiliation(s)
- Cassandre Caligaris
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Genaro Vázquez-Victorio
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Marcela Sosa-Garrocho
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Diana G Ríos-López
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Alvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México D.F., 14080, México
| | - Marina Macías-Silva
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México.
| |
Collapse
|
23
|
Liu C, Zhang H, Zang X, Wang C, Kong Y, Zhang H. The influence of SnoN gene silencing by siRNA on the cell proliferation and apoptosis of human pancreatic cancer cells. Diagn Pathol 2015; 10:30. [PMID: 25907906 PMCID: PMC4407884 DOI: 10.1186/s13000-015-0267-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The prognosis for pancreatic cancer (PC) is very poor. The SnoN gene may have a role in cell proliferation and apoptosis in human cancer. However, the influence of SnoN on cell proliferation and apoptosis in human PC cells remains unknown. METHODS SnoN expression was assessed in SW1990 PC cell lines using real-time polymerase chain reaction (PCR). A luciferase reporter assay was used to confirm the target associations. The effect of SnoN on cell proliferation in vitro was confirmed using Cell Counting Kit-8. Apoptosis was confirmed using flow cytometry. Gene and protein expression were examined using real time PCR and Western blotting, respectively. RESULTS SnoN siRNA significantly inhibited the growth of SW1990 cells by decreasing cell proliferation (P < 0.05) and increasing cell apoptosis (P < 0.05), compared with the blank group and the negative control group. The highest inhibition of cell proliferation appeared at 3 days post-transfection. Cell apoptosis more obvious at 48 h after transfection. CONCLUSIONS In summary, our results reveal that the RNAi-mediated downregulation of SnoN effectively inhibited the proliferation of PC cells. SnoN-siRNA also enhanced SW1990 PC cell apoptosis. These findings indicate that SnoN gene plays an important role in pancreatic cancer development, and might serve as a potential therapeutic target for pancreatic cancer. However, further in vivo studies are needed to clarify the influence of SnoN gene silencing by siRNA on pancreatic cancer therapy. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/7609324661510147.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Hui Zhang
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Xiaoxia Zang
- Department of Stomatology, Air Force General Hospital of PLA, Beijing, China.
| | - Cheng Wang
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Yalin Kong
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Hongyi Zhang
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
24
|
Abstract
Activin/Nodal growth factors control a broad range of biological processes, including early cell fate decisions, organogenesis and adult tissue homeostasis. Here, we provide an overview of the mechanisms by which the Activin/Nodal signalling pathway governs stem cell function in these different stages of development. We describe recent findings that associate Activin/Nodal signalling to pathological conditions, focusing on cancer stem cells in tumorigenesis and its potential as a target for therapies. Moreover, we will discuss future directions and questions that currently remain unanswered on the role of Activin/Nodal signalling in stem cell self-renewal, differentiation and proliferation.
Collapse
Affiliation(s)
- Siim Pauklin
- Anne McLaren Laboratory For Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Ludovic Vallier
- Anne McLaren Laboratory For Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, West Forvie Building, Robinson Way, University of Cambridge, Cambridge CB2 0SZ, UK
| |
Collapse
|
25
|
Rashidian J, Le Scolan E, Ji X, Zhu Q, Mulvihill MM, Nomura D, Luo K. Ski regulates Hippo and TAZ signaling to suppress breast cancer progression. Sci Signal 2015; 8:ra14. [PMID: 25670202 DOI: 10.1126/scisignal.2005735] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Ski, the transforming protein of the avian Sloan-Kettering retrovirus, inhibits transforming growth factor-β (TGF-β)/Smad signaling and displays both pro-oncogenic and anti-oncogenic activities in human cancer. Inhibition of TGF-β signaling is likely responsible for the pro-oncogenic activity of Ski. We investigated the mechanism(s) underlying the tumor suppressor activity of Ski and found that Ski suppressed the activity of the Hippo signaling effectors TAZ and YAP to inhibit breast cancer progression. TAZ and YAP are transcriptional coactivators that can contribute to cancer by promoting proliferation, tumorigenesis, and cancer stem cell expansion. Hippo signaling activates the the Lats family of kinases, which phosphorylate TAZ and YAP, resulting in cytoplasmic retention and degradation and inhibition of their transcriptional activity. We showed that Ski interacted with multiple components of the Hippo pathway to facilitate activation of Lats2, resulting in increased phosphorylation and subsequent degradation of TAZ. Ski also promoted the degradation of a constitutively active TAZ mutant that is not phosphorylated by Lats, suggesting the existence of a Lats2-independent degradation pathway. Finally, we showed that Ski repressed the transcriptional activity of TAZ by binding to the TAZ partner TEAD and recruiting the transcriptional co-repressor NCoR1 to the TEAD-TAZ complex. Ski effectively reversed transformation and epithelial-to-mesenchyme transition in cultured breast cancer cells and metastasis in TAZ-expressing xenografted tumors. Thus, Ski inhibited the function of TAZ through multiple mechanisms in human cancer cells.
Collapse
Affiliation(s)
- Juliet Rashidian
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Erwan Le Scolan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Xiaodan Ji
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Melinda M Mulvihill
- Department of Nutritional Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel Nomura
- Department of Nutritional Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA. Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
26
|
Ji X, Lu H, Zhou Q, Luo K. LARP7 suppresses P-TEFb activity to inhibit breast cancer progression and metastasis. eLife 2014; 3:e02907. [PMID: 25053741 PMCID: PMC4126343 DOI: 10.7554/elife.02907] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Transcriptional elongation by RNA polymerase (Pol) II is essential for gene expression during cell growth and differentiation. The positive transcription elongation factor b (P-TEFb) stimulates transcriptional elongation by phosphorylating Pol II and antagonizing negative elongation factors. A reservoir of P-TEFb is sequestered in the inactive 7SK snRNP where 7SK snRNA and the La-related protein LARP7 are required for the integrity of this complex. Here, we show that P-TEFb activity is important for the epithelial–mesenchymal transition (EMT) and breast cancer progression. Decreased levels of LARP7 and 7SK snRNA redistribute P-TEFb to the transcriptionally active super elongation complex, resulting in P-TEFb activation and increased transcription of EMT transcription factors, including Slug, FOXC2, ZEB2, and Twist1, to promote breast cancer EMT, invasion, and metastasis. Our data provide the first demonstration that the transcription elongation machinery plays a key role in promoting breast cancer progression by directly controlling the expression of upstream EMT regulators. DOI:http://dx.doi.org/10.7554/eLife.02907.001 To express a gene to make a protein, the gene's DNA must first be transcribed to produce molecules of messenger RNA. The start of the transcription process features two milestones. First, an enzyme called RNA Polymerase II starts the process. Shortly afterwards, however, the process pauses and only starts again when other proteins are recruited. This second step, called transcriptional elongation, is essential for gene expression in cells that are growing and specializing into specific cell types. However, it is unclear how important this second step is for the progression of human cancers, such as breast cancer. In humans, two proteins join together to form a complex called ‘positive transcription elongation factor b’ (or P-TEFb for short). This elongation factor encourages the transcriptional elongation step by adding phosphate groups onto RNA Polymerase II and by outcompeting other proteins that act to stop the process. However, some of the P-TEFb proteins in the cell's nucleus are unable to do this because they are held within a complex, which also contains an RNA molecule and some other proteins including one called LARP7. This protein–RNA complex is thought to help to prevent a number of cancers, for example breast cancer or stomach cancer; however the effect of P-TEFb proteins on cancers in humans is not known. Less LARP7 protein is made in breast cancer cells compared to healthy cells. And when Ji et al. reduced the levels of the LARP7 protein (or the RNA molecule involved in the complex), the P-TEFb proteins were released from the complex and were free to encourage transcriptional elongation. This led to the increased expression of other proteins that switch other genes on or off, including genes that allow breast cancer cells to spread around the body. On the other hand, Ji et al. revealed that freeing the P-TEFb proteins from the complex in the nucleus did not appear to cause new tumors to develop or existing tumors to grow. Ji et al. suggest that the LARP7 protein normally helps to prevent the spread of breast cancers by keeping the P-TEFb proteins inactive as a part of the protein–RNA complex. One of the next challenges will be to see if drugs that can inhibit the P-TEFb proteins might be useful as new treatments for late stage breast cancer. DOI:http://dx.doi.org/10.7554/eLife.02907.002
Collapse
Affiliation(s)
- Xiaodan Ji
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Huasong Lu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Qiang Zhou
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| |
Collapse
|
27
|
The self-limiting dynamics of TGF-β signaling in silico and in vitro, with negative feedback through PPM1A upregulation. PLoS Comput Biol 2014; 10:e1003573. [PMID: 24901250 PMCID: PMC4105941 DOI: 10.1371/journal.pcbi.1003573] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/27/2014] [Indexed: 12/28/2022] Open
Abstract
The TGF-β/Smad signaling system decreases its activity through strong negative regulation. Several molecular mechanisms of negative regulation have been published, but the relative impact of each mechanism on the overall system is unknown. In this work, we used computational and experimental methods to assess multiple negative regulatory effects on Smad signaling in HaCaT cells. Previously reported negative regulatory effects were classified by time-scale: degradation of phosphorylated R-Smad and I-Smad-induced receptor degradation were slow-mode effects, and dephosphorylation of R-Smad was a fast-mode effect. We modeled combinations of these effects, but found no combination capable of explaining the observed dynamics of TGF-β/Smad signaling. We then proposed a negative feedback loop with upregulation of the phosphatase PPM1A. The resulting model was able to explain the dynamics of Smad signaling, under both short and long exposures to TGF-β. Consistent with this model, immuno-blots showed PPM1A levels to be significantly increased within 30 min after TGF-β stimulation. Lastly, our model was able to resolve an apparent contradiction in the published literature, concerning the dynamics of phosphorylated R-Smad degradation. We conclude that the dynamics of Smad negative regulation cannot be explained by the negative regulatory effects that had previously been modeled, and we provide evidence for a new negative feedback loop through PPM1A upregulation. This work shows that tight coupling of computational and experiments approaches can yield improved understanding of complex pathways. TGF-β signaling pathway regulates a variety of cellular responses, such as differentiation, migration and apoptosis. Phosphorylated R-Smad, the central signaling protein in this pathway, exhibits self-limiting behaviors: it not only decreases quickly after TGF-β is removed, but it also decreases slowly when TGF-β remains abundant. These two self-limiting behaviors are important to understand clearly because diseases such as cancer and fibrosis might benefit from treatments to decrease Smad signaling. Several negative regulatory effects have been reported previously, and we studied the dynamics of these effects with computational modeling. Analyzing the timing of negative regulation revealed that the three most widely accepted effects were not sufficient to explain the observed declines. After considering and excluding several alternative models, we arrived at a model in which TGF-β upregulated the phosphatase PPM1A. We tested for PPM1A upregulation in cell culture experiments. In addition, our model was able to explain why different durations of TGF-β exposure could cause seemingly opposite results about the importance of Smad degradation.
Collapse
|
28
|
Regulation of estrogen receptor signaling in breast carcinogenesis and breast cancer therapy. Cell Mol Life Sci 2014; 71:1549. [PMID: 25031550 PMCID: PMC3962223 DOI: 10.1007/s00018-013-1376-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 12/19/2022]
Abstract
Estrogen and estrogen receptors (ERs) are critical regulators of breast epithelial cell proliferation, differentiation, and apoptosis. Compromised signaling vis-à-vis the estrogen receptor is believed to be a major contributing factor in the malignancy of breast cells. Targeting the ER signaling pathway has been a focal point in the development of breast cancer therapy. Although approximately 75 % of breast cancer patients are classified as luminal type (ER(+)), which predicts for response to endocrine-based therapy; however, innate or acquired resistance to endocrine-based drugs remains a serious challenge. The complexity of regulation for estrogen signaling coupled with the crosstalk of other oncogenic signaling pathways is a reason for endocrine therapy resistance. Alternative strategies that target novel molecular mechanisms are necessary to overcome this current and urgent gap in therapy. A thorough analysis of estrogen-signaling regulation is critical. In this review article, we will summarize current insights into the regulation of estrogen signaling as related to breast carcinogenesis and breast cancer therapy.
Collapse
|
29
|
Expression and prognostic role of SKIP in human breast carcinoma. J Mol Histol 2013; 45:169-80. [PMID: 24150787 DOI: 10.1007/s10735-013-9546-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/09/2013] [Indexed: 10/26/2022]
Abstract
Ski-interacting protein (SKIP) is a nuclear hormone receptor-interacting cofactor, interactions with the proto-oncogene Ski, appears to modulate a number of signalling pathways involved in control of cell proliferation and differentiation, and may play a critical role in oncogenesis. In the present study, to investigate the potential roles of SKIP in breast cancer, expression patterns, interaction and the correlation with clinical/prognostic factors of SKIP and Ki-67 were examined among patients with breast cancer. Immunohistochemistry and Western blot analysis were performed for SKIP in 85 breast carcinoma samples. The data were correlated with clinicopathological features. The univariate and multivariate survival analyses were also performed to determine their prognostic significance. We found that SKIP was over expressed in breast carcinoma as compared with the adjacent normal tissues. High expression of SKIP was positively associated with histological grade (P = 0.01) and Ki-67 (P = 0.004). Univariate analysis showed that SKIP expression was associated with a poor prognosis (P = 0.006). While in vitro, following release of breast cancer cell lines from serum starvation, the expression of SKIP was up-regulated, whereas p27 was down-regulated. In addition, we employed small interfering RNA (siRNA) technique to knock down SKIP expression and observed it effects on MDA-MB-231 cells growth. SKIP depletion by siRNA inhibited cell proliferation, blocked S phase and decreased cyclin A and cyclin B levels. On the basis of these results, we suggested that SKIP overexpression was involved in the pathogenesis of breast cancer, which might serve as a future target for breast cancer.
Collapse
|
30
|
Zhu Q, Kim YH, Wang D, Oh SP, Luo K. SnoN facilitates ALK1-Smad1/5 signaling during embryonic angiogenesis. ACTA ACUST UNITED AC 2013; 202:937-50. [PMID: 24019535 PMCID: PMC3776356 DOI: 10.1083/jcb.201208113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In endothelial cells, two type I receptors of the transforming growth factor β (TGF-β) family, ALK1 and ALK5, coordinate to regulate embryonic angiogenesis in response to BMP9/10 and TGF-β. Whereas TGF-β binds to and activates ALK5, leading to Smad2/3 phosphorylation and inhibition of endothelial cell proliferation and migration, BMP9/10 and TGF-β also bind to ALK1, resulting in the activation of Smad1/5. SnoN is a negative regulator of ALK5 signaling through the binding and repression of Smad2/3. Here we uncover a positive role of SnoN in enhancing Smad1/5 activation in endothelial cells to promote angiogenesis. Upon ligand binding, SnoN directly bound to ALK1 on the plasma membrane and facilitated the interaction between ALK1 and Smad1/5, enhancing Smad1/5 phosphorylation. Disruption of this SnoN-Smad interaction impaired Smad1/5 activation and up-regulated Smad2/3 activity. This resulted in defective angiogenesis and arteriovenous malformations, leading to embryonic lethality at E12.5. Thus, SnoN is essential for TGF-β/BMP9-dependent biological processes by its ability to both positively and negatively modulate the activities of Smad-dependent pathways.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | | | | | | | | |
Collapse
|
31
|
Wang X, Liu J, Wang Z, Huang Y, Liu W, Zhu X, Cai Y, Fang X, Lin S, Yuan L, Ouyang G. Periostin contributes to the acquisition of multipotent stem cell-like properties in human mammary epithelial cells and breast cancer cells. PLoS One 2013; 8:e72962. [PMID: 24009721 PMCID: PMC3756944 DOI: 10.1371/journal.pone.0072962] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 07/21/2013] [Indexed: 12/22/2022] Open
Abstract
Periostin (POSTN), a recently characterised matricellular protein, is frequently dysregulated in various malignant cancers and promotes tumor metastatic growth. POSTN plays a critical role in the crosstalk between murine breast cancer stem cells (CSCs) and their niche to permit metastatic colonization. However, whether pro-metastatic capability of POSTN is associated with multipotent potentials of mesenchymal stem cells (MSCs) has not been documented. Here we demonstrate that POSTN promotes a stem cell-like trait and a mesenchymal phenotype in human mammary epithelial cells and breast cancer cells. Interestingly, ectopic overexpression of POSTN or recombinant POSTN treatment can induce human mammary epithelial cells and breast cancer cells differentiation into multiple cell lineages that recapitulate part of the multilineage differentiation potentials of MSCs. Moreover, POSTN is highly expressed in bone marrow-derived MSCs and their derived adipocytes, chondrocytes, and osteoblasts in vitro. Furthermore, POSTN promotes the growth of xenograft tumors in vivo. POSTN-overexpressing human mammary epithelial cells enhance breast tumor growth and metastasis. These data thus provide evidence of a new role for POSTN in mammary epithelial neoplasia and metastasis, suggesting that epithelial cancer cells might acquire CSC-like traits and a mesenchymal phenotype, as well as the multipotent potentials of MSCs to promote tumorigenesis and metastasis. Therefore, targeting POSTN and other extracellular matrix components of tumor microenvironment may help to develop new therapeutical strategies to inhibit tumor metastasis.
Collapse
Affiliation(s)
- Xiaowei Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jia Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhe Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yangmei Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Weiping Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiao Zhu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yao Cai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiaoguang Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Shuyong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Li Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Gaoliang Ouyang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
- Laboratory of Stem Cells and Tumor Metastasis, School of Life Sciences, Xiamen University, Xiamen, China
- * E-mail:
| |
Collapse
|
32
|
Stepanenko AA, Vassetzky YS, Kavsan VM. Antagonistic functional duality of cancer genes. Gene 2013; 529:199-207. [PMID: 23933273 DOI: 10.1016/j.gene.2013.07.047] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 05/08/2013] [Accepted: 07/09/2013] [Indexed: 12/21/2022]
Abstract
Cancer evolution is a stochastic process both at the genome and gene levels. Most of tumors contain multiple genetic subclones, evolving in either succession or in parallel, either in a linear or branching manner, with heterogeneous genome and gene alterations, extensively rewired signaling networks, and addicted to multiple oncogenes easily switching with each other during cancer progression and medical intervention. Hundreds of discovered cancer genes are classified according to whether they function in a dominant (oncogenes) or recessive (tumor suppressor genes) manner in a cancer cell. However, there are many cancer "gene-chameleons", which behave distinctly in opposite way in the different experimental settings showing antagonistic duality. In contrast to the widely accepted view that mutant NADP(+)-dependent isocitrate dehydrogenases 1/2 (IDH1/2) and associated metabolite 2-hydroxyglutarate (R)-enantiomer are intrinsically "the drivers" of tumourigenesis, mutant IDH1/2 inhibited, promoted or had no effect on cell proliferation, growth and tumorigenicity in diverse experiments. Similar behavior was evidenced for dozens of cancer genes. Gene function is dependent on genetic network, which is defined by the genome context. The overall changes in karyotype can result in alterations of the role and function of the same genes and pathways. The diverse cell lines and tumor samples have been used in experiments for proving gene tumor promoting/suppressive activity. They all display heterogeneous individual karyotypes and disturbed signaling networks. Consequently, the effect and function of gene under investigation can be opposite and versatile in cells with different genomes that may explain antagonistic duality of cancer genes and the cell type- or the cellular genetic/context-dependent response to the same protein. Antagonistic duality of cancer genes might contribute to failure of chemotherapy. Instructive examples of unexpected activity of cancer genes and "paradoxical" effects of different anticancer drugs depending on the cellular genetic context/signaling network are discussed.
Collapse
Affiliation(s)
- A A Stepanenko
- State Key Laboratory of Molecular and Cellular Biology, Institute of Molecular Biology and Genetics, 150 Zabolotnogo Street, Kyiv 03680, Ukraine.
| | | | | |
Collapse
|
33
|
Hernández-Damián J, Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Vázquez-Macías A, Caligaris C, Sosa-Garrocho M, Flores-Pérez B, Romero-Avila M, Macías-Silva M. Downregulation of SnoN oncoprotein induced by antibiotics anisomycin and puromycin positively regulates transforming growth factor-β signals. Biochim Biophys Acta Gen Subj 2013; 1830:5049-58. [PMID: 23872350 DOI: 10.1016/j.bbagen.2013.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/26/2013] [Accepted: 07/09/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND SnoN and Ski proteins function as Smad transcriptional corepressors and are implicated in the regulation of diverse cellular processes such as proliferation, differentiation and transformation. Transforming growth factor-β (TGF-β) signaling causes SnoN and Ski protein degradation via proteasome with the participation of phosphorylated R-Smad proteins. Intriguingly, the antibiotics anisomycin (ANS) and puromycin (PURO) are also able to downregulate Ski and SnoN proteins via proteasome. METHODS We explored the effects of ANS and PURO on SnoN protein downregulation when the activity of TGF-β signaling was inhibited by using different pharmacological and non-pharmacological approaches, either by using specific TβRI inhibitors, overexpressing the inhibitory Smad7 protein, or knocking-down TβRI receptor or Smad2 by specific shRNAs. The outcome of SnoN and Ski downregulation induced by ANS or PURO on TGF-β signaling was also studied. RESULTS SnoN protein downregulation induced by ANS and PURO did not involve the induction of R-Smad phosphorylation but it was abrogated after TGF-β signaling inhibition; this effect occurred in a cell type-specific manner and independently of protein synthesis inhibition or any other ribotoxic effect. Intriguingly, antibiotics seem to require components of the TGF-β/Smad pathway to downregulate SnoN. In addition, SnoN protein downregulation induced by antibiotics favored gene transcription induced by TGF-β signaling. CONCLUSIONS ANS and PURO require TGF-β/Smad pathway to induce SnoN and Ski protein downregulation independently of inducing R-Smad2 phosphorylation, which facilitates TGF-β signaling. GENERAL SIGNIFICANCE Antibiotic analogs lacking ribotoxic effects are useful as pharmacological tools to study TGF-β signaling by controlling Ski and SnoN protein levels.
Collapse
Affiliation(s)
- Jacqueline Hernández-Damián
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., 04510 Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sengupta S, Jana S, Biswas S, Mandal PK, Bhattacharyya A. Cooperative involvement of NFAT and SnoN mediates transforming growth factor-β (TGF-β) induced EMT in metastatic breast cancer (MDA-MB 231) cells. Clin Exp Metastasis 2013; 30:1019-31. [PMID: 23832742 DOI: 10.1007/s10585-013-9600-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 06/27/2013] [Indexed: 12/17/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a secondary phenomenon concomitantly associated with the tumor progression. The regulatory signals and mechanistic details of EMT are not fully elucidated. Here, we shared a TGF-β mediated mechanism of EMT in breast cancer (MDA-MB 231) cells. Initial exposure of TGF-β for 48 h, enhanced the rate of cell proliferation and associated with EMT of MDA-MB 231 cells. The EMT was characterized by observing the increased N-cadherin, fibronectin, Snail expression and associated with the morphological change with a reduced E-cadherin expression. NFAT, a transcription factor, alters tumor suppressive function of TGF-β towards tumor progression. Up regulation of NFAT, coupled with a foremost translocation of one oncogenic protein SnoN from cytoplasm to nucleus was noticed during this TGF-β mediated EMT. Silencing of NFAT also showed the inhibition of TGF-β mediated EMT characterized by down regulation of N-cadherin and associated with reduced expression of SnoN. In addition, it was also observed that NFAT sequestering the Smad3 prevents the proteasome mediated degradation of SnoN and this SnoN has a role on the regulation of MMP-2, MMP-9 activity. Increased Smad3-SnoN interaction and proteasome mediated degradation of SnoN were detected after silencing of NFAT with a reduced MMP-2, MMP-9 activity. All of these observations provide a fresh mechanism in which by a twofold involvement of NFAT and SnoN plays a crucial role in TGF-β mediated EMT by recruiting the effector molecules N-cadherin and MMP-2, MMP-9.
Collapse
Affiliation(s)
- Suman Sengupta
- Immunology Lab, Department of Zoology, University of Calcutta, Kolkata, 700019, West Bengal, India
| | | | | | | | | |
Collapse
|
35
|
Briones-Orta MA, Levy L, Madsen CD, Das D, Erker Y, Sahai E, Hill CS. Arkadia regulates tumor metastasis by modulation of the TGF-β pathway. Cancer Res 2013; 73:1800-10. [PMID: 23467611 PMCID: PMC3672972 DOI: 10.1158/0008-5472.can-12-1916] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
TGF-β can act as a tumor suppressor at early stages of cancer progression and as a tumor promoter at later stages. The E3 ubiquitin ligase Arkadia (RNF111) is a critical component of the TGF-β signaling pathway, being required for a subset of responses, those mediated by Smad3-Smad4 complexes. It acts by mediating ligand-induced degradation of Ski and SnoN (SKIL), which are 2 potent transcriptional repressors. Here, we investigate the role of Arkadia in cancer using model systems to address both potential tumor-suppressive and tumor-promoting roles. Stable reexpression of Arkadia in lung carcinoma NCI-H460 cells, which we show contain a hemizygous nonsense mutation in the Arkadia/RNF111 gene, efficiently restored TGF-β-induced Smad3-dependent transcription, and substantially decreased the ability of these cells to grow in soft agar in vitro. However, it had no effect on tumor growth in vivo in mouse models. Moreover, loss of Arkadia in cancer cell lines and human tumors is rare, arguing against a prominent tumor-suppressive role. In contrast, we have uncovered a potent tumor-promoting function for Arkadia. Using 3 different cancer cell lines whose tumorigenic properties are driven by TGF-β signaling, we show that loss of Arkadia function, either by overexpression of dominant negative Arkadia or by siRNA-induced knockdown, substantially inhibited lung colonization in tail vein injection experiments in immunodeficient mice. Our findings indicate that Arkadia is not critical for regulating tumor growth per se, but is required for the early stages of cancer cell colonization at the sites of metastasis.
Collapse
Affiliation(s)
- Marco A. Briones-Orta
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Laurence Levy
- INSERM UMR S 938, Hôpital St-Antoine, 184 rue du Faubourg St-Antoine, 75012 Paris, France
| | - Chris D. Madsen
- Tumour Cell Biology Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Debipriya Das
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Yigit Erker
- INSERM UMR S 938, Hôpital St-Antoine, 184 rue du Faubourg St-Antoine, 75012 Paris, France
| | - Erik Sahai
- Tumour Cell Biology Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| | - Caroline S. Hill
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, 44 Lincoln’s Inn Fields, London WC2A 3LY, United Kingdom
| |
Collapse
|
36
|
Wang W, Liu C, Wang Y, Cao L. Effects of the downregulation of SnoN expression on HepG2 cell proliferation and apoptosis. Mol Med Rep 2013; 7:1324-8. [PMID: 23446947 DOI: 10.3892/mmr.2013.1340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 01/31/2013] [Indexed: 11/06/2022] Open
Abstract
Ski‑novel protein (SnoN) is a proto‑oncogene that belongs to the Ski protein family and is involved in regulating processes such as cell proliferation and apoptosis. To investigate the role of SnoN in the proliferation and apoptosis of HepG2 cells, we downregulated its expression by the use of small interfering RNA (siRNA). Three fragments predicted to have RNAi capacity were designed and synthesized as the target siRNAs (siRNA‑A, ‑B and ‑C). Following transfection, inhibition efficiency was detected by reverse transcription PCR (RT‑PCR) and western blot analysis. The siRNA with the optimal inhibition efficiency was used for the cell proliferation and apoptosis analysis. Cell proliferation was analyzed by the Cell Counting Kit‑8 (CCK‑8) and cell apoptosis was investigated by flow cytometry. In our study, all three siRNAs efficiently inhibited SnoN expression, and siRNA‑C demonstrated the optimal inhibition efficiency. We found that following downregulation of SnoN expression, HepG2 cell proliferation was significantly inhibited (P<0.05), while HepG2 cell apoptosis was significantly increased (P<0.05). SnoN‑specific siRNA is capable of effectively inhibiting the expression of SnoN in human HepG2 cells, and the downregulation of SnoN expression induces growth inhibition and apoptosis.
Collapse
Affiliation(s)
- Wenqi Wang
- Department of Gastroenterology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China.
| | | | | | | |
Collapse
|
37
|
Jahchan NS, Ouyang G, Luo K. Expression profiles of SnoN in normal and cancerous human tissues support its tumor suppressor role in human cancer. PLoS One 2013; 8:e55794. [PMID: 23418461 PMCID: PMC3572181 DOI: 10.1371/journal.pone.0055794] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 12/30/2012] [Indexed: 11/18/2022] Open
Abstract
SnoN is a negative regulator of TGF-β signaling and also an activator of the tumor suppressor p53 in response to cellular stress. Its role in human cancer is complex and controversial with both pro-oncogenic and anti-oncogenic activities reported. To clarify its role in human cancer and provide clinical relevance to its signaling activities, we examined SnoN expression in normal and cancerous human esophageal, ovarian, pancreatic and breast tissues. In normal tissues, SnoN is expressed in both the epithelium and the surrounding stroma at a moderate level and is predominantly cytoplasmic. SnoN levels in all tumor epithelia examined are lower than or similar to that in the matched normal samples, consistent with its anti-tumorigenic activity in epithelial cells. In contrast, SnoN expression in the stroma is highly upregulated in the infiltrating inflammatory cells in high-grade esophageal and ovarian tumor samples, suggesting that SnoN may potentially promote malignant progression through modulating the tumor microenvironment in these tumor types. The overall levels of SnoN expression in these cancer tissues do not correlate with the p53 status. However, in human cancer cell lines with amplification of the snoN gene, a strong correlation between increased SnoN copy number and inactivation of p53 was detected, suggesting that the tumor suppressor SnoN-p53 pathway must be inactivated, either through downregulation of SnoN or inactivation of p53, in order to allow cancer cell to proliferate and survive. These data strongly suggest that SnoN can function as a tumor suppressor at early stages of tumorigenesis in human cancer tissues.
Collapse
Affiliation(s)
- Nadine S. Jahchan
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Gaoliang Ouyang
- College of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Tsuneyoshi N, Tan EK, Sadasivam A, Poobalan Y, Sumi T, Nakatsuji N, Suemori H, Dunn NR. The SMAD2/3 corepressor SNON maintains pluripotency through selective repression of mesendodermal genes in human ES cells. Genes Dev 2013; 26:2471-6. [PMID: 23154981 DOI: 10.1101/gad.201772.112] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Activin/Nodal signaling via SMAD2/3 maintains human embryonic stem cell (hESC) pluripotency by direct transcriptional regulation of NANOG or, alternatively, induces mesoderm and definitive endoderm (DE) formation. In search of an explanation for these contrasting effects, we focused on SNON (SKIL), a potent SMAD2/3 corepressor that is expressed in hESCs but rapidly down-regulated upon differentiation. We show that SNON predominantly associates with SMAD2 at the promoters of primitive streak (PS) and early DE marker genes. Knockdown of SNON results in premature activation of PS and DE genes and loss of hESC morphology. In contrast, enforced SNON expression inhibits DE formation and diverts hESCs toward an extraembryonic fate. Thus, our findings provide novel mechanistic insight into how a single signaling pathway both regulates pluripotency and directs lineage commitment.
Collapse
Affiliation(s)
- Norihiro Tsuneyoshi
- Institute of Medical Biology, A*STAR (Agency for Science, Technology, and Research), Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Lebrun JJ. The Dual Role of TGFβ in Human Cancer: From Tumor Suppression to Cancer Metastasis. ISRN MOLECULAR BIOLOGY 2012; 2012:381428. [PMID: 27340590 PMCID: PMC4899619 DOI: 10.5402/2012/381428] [Citation(s) in RCA: 239] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 10/12/2012] [Indexed: 12/31/2022]
Abstract
The transforming growth factor-beta (TGFβ) superfamily encompasses widespread and evolutionarily conserved polypeptide growth factors that regulate and orchestrate growth and differentiation in all cell types and tissues. While they regulate asymmetric cell division and cell fate determination during early development and embryogenesis, TGFβ family members play a major regulatory role in hormonal and immune responses, cell growth, cell death and cell immortalization, bone formation, tissue remodeling and repair, and erythropoiesis throughout adult life. The biological and physiological functions of TGFβ, the founding member of this family, and its receptors are of central importance to human diseases, particularly cancer. By regulating cell growth, death, and immortalization, TGFβ signaling pathways exert tumor suppressor effects in normal cells and early carcinomas. Thus, it is not surprising that a high number of human tumors arise due to mutations or deletions in the genes coding for the various TGFβ signaling components. As tumors develop and progress, these protective and cytostatic effects of TGFβ are often lost. TGFβ signaling then switches to promote cancer progression, invasion, and tumor metastasis. The molecular mechanisms underlying this dual role of TGFβ in human cancer will be discussed in depth in this paper, and it will highlight the challenge and importance of developing novel therapeutic strategies specifically aimed at blocking the prometastatic arm of the TGFβ signaling pathway without affecting its tumor suppressive effects.
Collapse
Affiliation(s)
- Jean-Jacques Lebrun
- Division of Medical Oncology, Department of Medicine, Royal Victoria Hospital, McGill University Health Center, Montreal, QC, Canada H3A 1A1
| |
Collapse
|
40
|
Pan D, Zhu Q, Conboy MJ, Conboy IM, Luo K. SnoN activates p53 directly to regulate aging and tumorigenesis. Aging Cell 2012; 11:902-911. [PMID: 22805162 DOI: 10.1111/j.1474-9726.2012.00857.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We have identified SnoN as a direct activator of p53 to accelerate aging and inhibit tumorigenesis. SnoN has been shown previously to promote proliferation and transformation by antagonizing TGFβ signaling. We show that elimination of this TGFβ antagonistic activity of SnoN in vivo results in accelerated aging and resistance to tumorigenesis. The SnoN knockin mice display a shortened lifespan, decreased reproductivity, osteoporosis, reduced regenerative capacity, and other aging phenotypes, similar to that found in mice expressing an active p53. These activities of SnoN rely on the ability of SnoN to activate p53. SnoN can bind directly to p53 and compete with Mdm2 for binding to p53, preventing p53 ubiquitination and degradation and additionally facilitating p53 acetylation and phosphorylation. SnoN also binds to p53 on the promoter of p53 responsive genes to promote transcription activation. This activation of p53 by SnoN is necessary for its antitumorigenic and progeria activities in vivo because elimination of one copy of p53 reverses the aging phenotypes and accelerates tumorigenesis. Thus, we have revealed a novel function of SnoN in regulating aging and tumorigenesis by directly activating p53.
Collapse
Affiliation(s)
- Deng Pan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Michael J Conboy
- Department of Bioengineering and QB3 University of California, Berkeley, CA 94720
| | - Irina M Conboy
- Department of Bioengineering and QB3 University of California, Berkeley, CA 94720
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| |
Collapse
|
41
|
Tecalco-Cruz AC, Sosa-Garrocho M, Vázquez-Victorio G, Ortiz-García L, Domínguez-Hüttinger E, Macías-Silva M. Transforming growth factor-β/SMAD Target gene SKIL is negatively regulated by the transcriptional cofactor complex SNON-SMAD4. J Biol Chem 2012; 287:26764-76. [PMID: 22674574 PMCID: PMC3411014 DOI: 10.1074/jbc.m112.386599] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Indexed: 12/23/2022] Open
Abstract
The human SKI-like (SKIL) gene encodes the SMAD transcriptional corepressor SNON that antagonizes TGF-β signaling. SNON protein levels are tightly regulated by the TGF-β pathway: whereas a short stimulation with TGF-β decreases SNON levels by its degradation via the proteasome, longer TGF-β treatment increases SNON levels by inducing SKIL gene expression. Here, we investigated the molecular mechanisms involved in the self-regulation of SKIL gene expression by SNON. Bioinformatics analysis showed that the human SKIL gene proximal promoter contains a TGF-β response element (TRE) bearing four groups of SMAD-binding elements that are also conserved in mouse. Two regions of 408 and 648 bp of the human SKIL gene (∼2.4 kb upstream of the ATG initiation codon) containing the core promoter, transcription start site, and the TRE were cloned for functional analysis. Binding of SMAD and SNON proteins to the TRE region of the SKIL gene promoter after TGF-β treatment was demonstrated by ChIP and sequential ChIP assays. Interestingly, the SNON-SMAD4 complex negatively regulated basal SKIL gene expression through binding the promoter and recruiting histone deacetylases. In response to TGF-β signal, SNON is removed from the SKIL gene promoter, and then the activated SMAD complexes bind the promoter to induce SKIL gene expression. Subsequently, the up-regulated SNON protein in complex with SMAD4 represses its own expression as part of the negative feedback loop regulating the TGF-β pathway. Accordingly, when the SNON-SMAD4 complex is absent as in some cancer cells lacking SMAD4 the regulation of some TGF-β target genes is modified.
Collapse
Affiliation(s)
- Angeles C. Tecalco-Cruz
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Marcela Sosa-Garrocho
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Genaro Vázquez-Victorio
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Layla Ortiz-García
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Elisa Domínguez-Hüttinger
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Marina Macías-Silva
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| |
Collapse
|
42
|
Vo BT, Cody B, Cao Y, Khan SA. Differential role of Sloan-Kettering Institute (Ski) protein in Nodal and transforming growth factor-beta (TGF-β)-induced Smad signaling in prostate cancer cells. Carcinogenesis 2012; 33:2054-64. [PMID: 22843506 DOI: 10.1093/carcin/bgs252] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) signaling pathways contain both tumor suppressor and tumor promoting activities. We have demonstrated that Nodal, another member of the TGF-β superfamily, and its receptors are expressed in prostate cancer cells. Nodal and TGF-β exerted similar biological effects on prostate cells; both inhibited proliferation in WPE, RWPE1 and DU145 cells, whereas neither had any effect on the proliferation of LNCaP or PC3 cells. Interestingly, Nodal and TGF-β induced migration in PC3 cells, but not in DU145 cells. TGF-β induced predominantly phosphorylation of Smad3, whereas Nodal induced phosphorylation of only Smad2. We also determined the expression and differential role of Ski, a corepressor of Smad2/3, in Nodal and TGF-β signaling in prostate cancer cells. Similar levels of Ski mRNA were found in several established prostate cell lines; however, high levels of Ski protein were only detected in prostate cancer cells and prostate cancer tissue samples. Exogenous Nodal and TGF-β had no effects on Ski mRNA levels. On the other hand, TGF-β induced a rapid degradation of Ski protein mediated by the proteasomal pathway, whereas Nodal had no effect on Ski protein. Reduced Ski levels correlated with increased basal and TGF-β-induced Smad2/3 phosphorylation. Knockdown of endogenous Ski reduced proliferation in DU145 cells and enhanced migration of PC3 cells. We conclude that high levels of Ski expression in prostate cancer cells may be responsible for repression of TGF-β and Smad3 signaling, but Ski protein levels do not influence Nodal and Smad2 signaling.
Collapse
Affiliation(s)
- BaoHan T Vo
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | | | | | | |
Collapse
|
43
|
Jahchan NS, Wang D, Bissell MJ, Luo K. SnoN regulates mammary gland alveologenesis and onset of lactation by promoting prolactin/Stat5 signaling. Development 2012; 139:3147-56. [PMID: 22833129 DOI: 10.1242/dev.079616] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mammary epithelial cells undergo structural and functional differentiation at late pregnancy and parturition to produce and secrete milk. Both TGF-β and prolactin pathways are crucial regulators of this process. However, how the activities of these two antagonistic pathways are orchestrated to initiate lactation has not been well defined. Here, we show that SnoN, a negative regulator of TGF-β signaling, coordinates TGF-β and prolactin signaling to control alveologenesis and lactogenesis. SnoN expression is induced at late pregnancy by the coordinated actions of TGF-β and prolactin. The elevated SnoN promotes Stat5 signaling by enhancing its stability, thereby sharply increasing the activity of prolactin signaling at the onset of lactation. SnoN-/- mice display severe defects in alveologenesis and lactogenesis, and mammary epithelial cells from these mice fail to undergo proper morphogenesis. These defects can be rescued by an active Stat5. Thus, our study has identified a new player in the regulation of milk production and revealed a novel function of SnoN in mammary alveologenesis and lactogenesis in vivo through promotion of Stat5 signaling.
Collapse
Affiliation(s)
- Nadine S Jahchan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
44
|
Liu R, Wang Y, Xiao Y, Shi M, Zhang G, Guo B. SnoN as a key regulator of the high glucose-induced epithelial-mesenchymal transition in cells of the proximal tubule. Kidney Blood Press Res 2012; 35:517-28. [PMID: 22813962 DOI: 10.1159/000339172] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 04/27/2012] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIMS Ski-related protein N (SnoN) suppression is essential to transforming growth factor-β1 induction and the epithelial-mesenchymal transition (EMT) in several cancer cells. The role of SnoN in diabetic nephropathy is unknown. We aimed to determine the role of SnoN in the EMT of proximal tubule cells (PTCs) maintained under high glucose conditions. METHODS Immunohistochemistry, immunocytochemistry, Western blotting, small interfering RNA gene silencing, viral transduction and RT-PCR were used to assess changes in SnoN, E-cadherin, cytokeratin-18, α-smooth muscle actin and fibronectin expression using an in vivo streptozotocin-induced rat diabetic nephropathy model, and PTCs exposed to high glucose (25 mmol/l). RESULTS High glucose induced EMT in vitro and in vivo. Exposure of PTCs to a high concentration of glucose suppressed SnoN expression in a time-dependent manner compared with normal glucose and high osmolarity-treated groups. SnoN gene silencing under high glucose conditions appears to enhance the transition of PTC phenotype. Conversely, ectopic expression of exogenous SnoN after transfection conferred tubular epithelial cell resistance to high glucose-induced EMT. CONCLUSION SnoN plays a negative role in high glucose-induced EMT in PTCs. The effect of SnoN downregulation in vivo and in vitro suggests that SnoN may be a potential therapeutic target.
Collapse
Affiliation(s)
- Ruixia Liu
- Department of Pathophysiology, Guiyang Medical University, Guiyang, China
| | | | | | | | | | | |
Collapse
|
45
|
Zhu Q, Luo K. SnoN in regulation of embryonic development and tissue morphogenesis. FEBS Lett 2012; 586:1971-6. [PMID: 22710172 DOI: 10.1016/j.febslet.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/03/2012] [Accepted: 03/05/2012] [Indexed: 01/15/2023]
Abstract
SnoN (Ski-novel protein) plays an important role in embryonic development, tumorigenesis and aging. Past studies largely focused on its roles in tumorigenesis. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed that SnoN interacts with multiple signaling molecules at different cellular levels to modulate the activities of several signaling pathways in a tissue context and developmental stage dependent manner. These studies suggest that SnoN may have broad functions in the embryonic development and tissue morphogenesis.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
46
|
Bonni S, Bonni A. SnoN signaling in proliferating cells and postmitotic neurons. FEBS Lett 2012; 586:1977-83. [PMID: 22710173 DOI: 10.1016/j.febslet.2012.02.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 01/28/2023]
Abstract
The transcriptional regulator SnoN plays a fundamental role as a modulator of transforming growth factor beta (TGFβ)-induced signal transduction and biological responses. In recent years, novel functions of SnoN have been discovered in both TGFβ-dependent and TGFβ-independent settings in proliferating cells and postmitotic neurons. Accumulating evidence suggests that SnoN plays a dual role as a corepressor or coactivator of TGFβ-induced transcription. Accordingly, SnoN exerts oncogenic or tumor-suppressive effects in epithelial tissues. At the cellular level, SnoN antagonizes or mediates the ability of TGFβ to induce cell cycle arrest in a cell-type specific manner. SnoN also exerts key effects on epithelial-mesenchymal transition (EMT), with implications in cancer biology. Recent studies have expanded SnoN functions to postmitotic neurons, where SnoN orchestrates key aspects of neuronal development in the mammalian brain, from axon growth and branching to neuronal migration and positioning. In this review, we will highlight our understanding of SnoN biology at the crossroads of cancer biology and neurobiology.
Collapse
Affiliation(s)
- Shirin Bonni
- Department of Biochemistry and Molecular Biology, Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada T2N 4N1.
| | | |
Collapse
|
47
|
Band AM, Laiho M. SnoN oncoprotein enhances estrogen receptor-α transcriptional activity. Cell Signal 2011; 24:922-30. [PMID: 22227247 DOI: 10.1016/j.cellsig.2011.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 12/19/2011] [Indexed: 12/13/2022]
Abstract
Estrogen receptor-α (ERα) and transforming growth factor-beta (TGF-β) signaling pathways are essential regulators during mammary gland development and tumorigenesis. Ski-related novel gene (SnoN) is an oncoprotein and a negative feedback inhibitor of TGF-β signaling. We have previously reported that low expression of SnoN in ERα positive breast carcinomas is associated with favorable prognosis (Zhang et al. Cancer Res. (2003) 63, 5005-5010). Here we have studied the mechanism of a possible cross-talk between ERα and SnoN. We find that SnoN interacts with the estrogen-activated form of ERα in the nucleus. SnoN contains two highly conserved nuclear receptor binding LxxLL-like motifs and we show that mutations in these motifs reduce the interaction of SnoN with ERα. Over-expression of SnoN enhanced the transcriptional activity of ERα in estrogen response element (ERE)-reporter assays, augmented the expression of several ERα target genes and increased the proliferation of MCF7 breast carcinoma cells in an estrogen-dependent manner. Chromatin immunoprecipitation demonstrated that SnoN interacts with ERα at the TTF1 (pS2) gene promoter. Conversely, silencing of SnoN reduced both ERE-reporter activity and the expression of ERα target genes in MCF7 and T-47D breast cancer cells. Histone deacetylase inhibition increased the level of SnoN and SnoN-dependent enhancement of ERα-dependent transcription and SnoN supported the recruitment of p300 histone acetylase to ERα. This study reveals a novel mechanism that interconnects ERα and TGF-β signaling pathways by SnoN. Accordingly, the results indicate that high SnoN level promotes ERα signaling and possibly breast cancer progression.
Collapse
Affiliation(s)
- Arja M Band
- Molecular Cancer Biology Program, Biomedicum Helsinki and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
48
|
Marcelain K, Armisen R, Aguirre A, Ueki N, Toro J, Colmenares C, Hayman MJ. Chromosomal instability in mouse embryonic fibroblasts null for the transcriptional co-repressor Ski. J Cell Physiol 2011; 227:278-87. [PMID: 21412778 DOI: 10.1002/jcp.22733] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Ski is a transcriptional regulator that has been considered an oncoprotein given its ability to induce oncogenic transformation in avian model systems. However, studies in mouse and in some human tumor cells have also indicated a tumor suppressor activity for this protein. We found that Ski-/- mouse embryo fibroblasts exhibit high levels of genome instability, namely aneuploidy, consistent with a tumor suppressor function for Ski. Time-lapse microscopy revealed lagging chromosomes and chromatin/chromosome bridges as the major cause of micronuclei (MN) formation and the subsequent aneuploidy. Although these cells arrested in mitosis after treatment with spindle disrupting drugs and exhibited a delayed metaphase/anaphase transition, spindle assembly checkpoint (SAC) was not sufficient to prevent chromosome missegregation, consistent with a weakened SAC. Our in vivo analysis also showed dynamic metaphase plate rearrangements with switches in polarity in cells arrested in metaphase. Importantly, after ectopic expression of Ski the cells that displayed this metaphase arrest died directly during metaphase or after aberrant cell division, relating SAC activation and mitotic cell death. This increased susceptibility to undergo mitosis-associated cell death reduced the number of MN-containing cells. The presented data support a new role for Ski in the mitotic process and in maintenance of genetic stability, providing insights into the mechanism of tumor suppression mediated by this protein.
Collapse
Affiliation(s)
- Katherine Marcelain
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | | | | | | | | | | | | |
Collapse
|
49
|
Tran DD, Corsa CAS, Biswas H, Aft RL, Longmore GD. Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast cancer recurrence. Mol Cancer Res 2011; 9:1644-57. [PMID: 22006115 PMCID: PMC4922748 DOI: 10.1158/1541-7786.mcr-11-0371] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a normal developmental program that is considered to also play an important role in cancer metastasis. Ultimate inducers of EMT are transcriptional repressors that individually can induce experimental EMT, yet in many cells, particularly cancer cells, multiple inducers are expressed simultaneously. Why, and if, and how they interact to regulate EMT is unanswered. Using RNA interference technology to affect protein knockdown and avoid potential overexpression artifact coupled with transient TGFβ treatment to better mimic in vivo conditions we show, in both nontumorigenic and tumorigenic epithelial cancer cells, that Snail1 is uniquely required for EMT initiation, whereas Twist1 is required to maintain late EMT. Twist1, present in resting epithelial cells, is dispensable for EMT initiation. Mechanistically, in response to transient TGFβ treatment, transient Snail1 expression represses Twist1 transcription directly, which is subsequently upregulated, as Snail1 levels decrease, to sustain E-cadherin downregulation and growth arrest of EMT. Persistent Twist1 expression is associated with a p38 and extracellular signal-regulated kinase signal feedback loop that sustains growth-inhibitory signals characteristic of quiescent micrometastatic tumors. This Snail1-Twist1 temporal and spatial cooperation was also observed in vivo during human breast cancer progression to metastasis. Twist1 level, but not Snail1 level, and Twist1:Snail1 ratio in disseminated micrometastatic bone marrow tumor cells was found to correlate with survival and treatment resistance and is highly predictive of metastatic or recurrent disease.
Collapse
Affiliation(s)
- David D. Tran
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Callie Ann S. Corsa
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- The BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Hirak Biswas
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- The BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| | - Rebecca L. Aft
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- John Cochran Veterans Administration Hospital, 915 North Grand Boulevard, Saint Louis, MO 63106, USA
| | - Gregory D. Longmore
- Departments of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- The BRIGHT Institute, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
- Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, Saint Louis, MO 63110, USA
| |
Collapse
|
50
|
Band AM, Laiho M. Crosstalk of TGF-β and estrogen receptor signaling in breast cancer. J Mammary Gland Biol Neoplasia 2011; 16:109-15. [PMID: 21390570 DOI: 10.1007/s10911-011-9203-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 02/28/2011] [Indexed: 01/17/2023] Open
Abstract
Estrogen receptor-α (ERα) and transforming growth factor (TGF)-β signaling pathways are major regulators during mammary gland development, function and tumorigenesis. Predominantly, they have opposing roles in proliferation and apoptosis. While ERα signaling supports growth and differentiation and is antiapoptotic, mammary gland epithelia cells are very sensitive to TGF-β-induced cell cycle arrest and apoptosis. Their regulatory pathways intersect, and ERα blocks TGF-β pathway by multiple means, including direct interactions of its signaling components, Smads. However, relatively little is known of the dysfunction of their interactions in cancer. A better understanding would help to develop new strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Arja M Band
- Molecular Cancer Biology Program, Biomedicum Helsinki and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|