1
|
Nicaise Y, Delmas C, Cohen‐Jonathan‐Moyal E, Seva C. PEA3 Transcription Factors, Role in Invasion, Proliferation and Radioresistance of Glioblastoma Stem Cells. J Cell Mol Med 2025; 29:e70533. [PMID: 40275610 PMCID: PMC12022000 DOI: 10.1111/jcmm.70533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
The presence of glioblastoma stem cells (GSCs), known for their high invasiveness and resistance to radiation, is one of the reasons for systematic recurrence. It is therefore important to understand the resistance mechanisms of these cells to optimise therapies. We focused on the PEA3 family of transcription factors, ETV1, ETV4 and ETV5, in patient-derived GSCs. We demonstrate that ETV1 is over-expressed in high invasive GSCs. In 3D invasion assays, inhibiting ETV1 expression using specific siRNAs significantly reduces cell invasion. Furthermore, we show a significant correlation between ETV1 and ZEB1, a major driver of invasion. Blocking ETV1 decreases ZEB1 expression in GSCs. The study also demonstrates the essential role of ETV1, ETV4 and ETV5 in the radioresistance of GSCs and their ability to form neurospheres. Using specific siRNAs to inhibit the expression of these transcription factors led to an increased sensitivity of GSCs to radiation and a decrease in both the number and size of neurospheres. These findings suggest that PEA3 transcription factors play a major role in GSCs aggressiveness by regulating invasion, radioresistance and the ability to form neurospheres. Trial Registration: Registry and the Registration N° of the study/trial: 12TETE01, ID-RCB No. 2012-A00585-38, approval Date: May 7, 2012.
Collapse
Affiliation(s)
- Yvan Nicaise
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
| | - Caroline Delmas
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
- IUCT‐Oncopole, Institut Claudius RegaudToulouseFrance
| | - Elizabeth Cohen‐Jonathan‐Moyal
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
- IUCT‐Oncopole, Institut Claudius RegaudToulouseFrance
| | - Catherine Seva
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
| |
Collapse
|
2
|
Arakil N, Akhund SA, Elaasser B, Mohammad KS. Intersecting Paths: Unraveling the Complex Journey of Cancer to Bone Metastasis. Biomedicines 2024; 12:1075. [PMID: 38791037 PMCID: PMC11117796 DOI: 10.3390/biomedicines12051075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/27/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
The phenomenon of bone metastases presents a significant challenge within the context of advanced cancer treatments, particularly pertaining to breast, prostate, and lung cancers. These metastatic occurrences stem from the dissemination of cancerous cells into the bone, thereby interrupting the equilibrium between osteoblasts and osteoclasts. Such disruption results in skeletal complications, adversely affecting patient morbidity and quality of life. This review discusses the intricate interplay between cancer cells and the bone microenvironment, positing the bone not merely as a passive recipient of metastatic cells but as an active contributor to cancer progression through its distinctive biochemical and cellular makeup. A thorough examination of bone structure and the dynamics of bone remodeling is undertaken, elucidating how metastatic cancer cells exploit these processes. This review explores the genetic and molecular pathways that underpin the onset and development of bone metastases. Particular emphasis is placed on the roles of cytokines and growth factors in facilitating osteoclastogenesis and influencing osteoblast activity. Additionally, this paper offers a meticulous critique of current diagnostic methodologies, ranging from conventional radiography to advanced molecular imaging techniques, and discusses the implications of a nuanced understanding of bone metastasis biology for therapeutic intervention. This includes the development of targeted therapies and strategies for managing bone pain and other skeletal-related events. Moreover, this review underscores the imperative of ongoing research efforts aimed at identifying novel therapeutic targets and refining management approaches for bone metastases. It advocates for a multidisciplinary strategy that integrates advancements in medical oncology and radiology with insights derived from molecular biology and genetics, to enhance prognostic outcomes and the quality of life for patients afflicted by this debilitating condition. In summary, bone metastases constitute a complex issue that demands a comprehensive and informed approach to treatment. This article contributes to the ongoing discourse by consolidating existing knowledge and identifying avenues for future investigation, with the overarching objective of ameliorating patient care in the domain of oncology.
Collapse
Affiliation(s)
| | | | | | - Khalid S. Mohammad
- Department of Anatomy, College of Medicine, Alfaisal University, Riyadh 1153, Saudi Arabia; (N.A.); (S.A.A.); (B.E.)
| |
Collapse
|
3
|
Sánchez P, Serrano Falcón C, Martínez Rodríguez S, Torres JM, Serrano S, Ortega E. mRNA Levels of Aromatase, 5α-Reductase Isozymes, and Prostate Cancer-Related Genes in Plucked Hair from Young Men with Androgenic Alopecia. Int J Mol Sci 2023; 24:17461. [PMID: 38139289 PMCID: PMC10744313 DOI: 10.3390/ijms242417461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Androgenic alopecia (AGA) is the most prevalent type of progressive hair loss and has psychological repercussions. Nevertheless, the effectiveness of current pharmacological treatments remains limited, in part because the molecular basis of the disease has not been fully elucidated. Our group previously highlighted the important roles of aromatase and 5α-reductase (5α-R) in alopecia in young women with female pattern hair loss. Additionally, an association has been proposed between AGA and prostate cancer (PCa), suggesting that genes implicated in PCa would also be involved in AGA. A low-invasive, sensitive, and precise method was used to determine mRNA levels of aromatase, 5α-R isozymes, and 84 PCa-related genes in samples of plucked hair from young men with AGA and controls. Samples were obtained with a trichogram from the vertex scalp, and mRNA levels were quantified using real-time RT-PCR. The men with AGA had significantly higher 5α-R2 mRNA levels in comparison to controls; interestingly, some of them also showed markedly elevated mRNA levels of 5α-R1 or 5α-R3 or of both, which may explain the varied response to 5α-R inhibitor treatments. The men with AGA also showed significant changes versus controls in 6 out of the 84 genes implicated in PCa. This study contributes greater knowledge of the molecular bases of AGA, facilitating early selection of the most appropriate pharmacological therapy and opening the way to novel treatments.
Collapse
Affiliation(s)
- Pilar Sánchez
- Department of Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; (P.S.); (S.M.R.)
| | | | - Sergio Martínez Rodríguez
- Department of Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; (P.S.); (S.M.R.)
| | - Jesús M. Torres
- Department of Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; (P.S.); (S.M.R.)
| | - Salvio Serrano
- Department of Dermatology, Faculty of Medicine, University of Granada, 18012 Granada, Spain;
| | - Esperanza Ortega
- Department of Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; (P.S.); (S.M.R.)
| |
Collapse
|
4
|
VARISLI LOKMAN, TOLAN VEYSEL, CEN JIYANH, VLAHOPOULOS SPIROS, CEN OSMAN. Dissecting the effects of androgen deprivation therapy on cadherin switching in advanced prostate cancer: A molecular perspective. Oncol Res 2023; 30:137-155. [PMID: 37305018 PMCID: PMC10208071 DOI: 10.32604/or.2022.026074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Prostate cancer is one of the most often diagnosed malignancies in males and its prevalence is rising in both developed and developing countries. Androgen deprivation therapy has been used as a standard treatment approach for advanced prostate cancer for more than 80 years. The primary aim of androgen deprivation therapy is to decrease circulatory androgen and block androgen signaling. Although a partly remediation is accomplished at the beginning of treatment, some cell populations become refractory to androgen deprivation therapy and continue to metastasize. Recent evidences suggest that androgen deprivation therapy may cause cadherin switching, from E-cadherin to N-cadherin, which is the hallmark of epithelial-mesenchymal transition. Diverse direct and indirect mechanisms are involved in this switching and consequently, the cadherin pool changes from E-cadherin to N-cadherin in the epithelial cells. Since E-cadherin represses invasive and migrative behaviors of the tumor cells, the loss of E-cadherin disrupts epithelial tissue structure leading to the release of tumor cells into surrounding tissues and circulation. In this study, we review the androgen deprivation therapy-dependent cadherin switching in advanced prostate cancer with emphasis on its molecular basis especially the transcriptional factors regulated through TFG-β pathway.
Collapse
Affiliation(s)
- LOKMAN VARISLI
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, 21280, Turkey
- Cancer Research Center, Dicle University, Diyarbakir, 21280, Turkey
| | - VEYSEL TOLAN
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, 21280, Turkey
| | - JIYAN H. CEN
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL, 61801, USA
| | - SPIROS VLAHOPOULOS
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - OSMAN CEN
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Natural Sciences and Engineering, John Wood College, Quincy, IL, 62305, USA
| |
Collapse
|
5
|
Zhang J, Wang Y, Yuan B, Qin H, Wang Y, Yu H, Teng X, Yang Y, Zou J, Zhang M, Huang W, Wang Y. Identifying key transcription factors and immune infiltration in non-small-cell lung cancer using weighted correlation network and Cox regression analyses. Front Oncol 2023; 13:1112020. [PMID: 37197420 PMCID: PMC10183566 DOI: 10.3389/fonc.2023.1112020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/12/2023] [Indexed: 05/19/2023] Open
Abstract
Introduction Lung cancer is one of the most common cancers and a significant cause of cancer-related deaths. Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases. Therefore, it is crucial to identify effective diagnostic and therapeutic methods. In addition, transcription factors are essential for eukaryotic cells to regulate their gene expression, and aberrant expression transcription factors are an important step in the process of oncogenesis in NSCLC. Methods Differentially expressed transcription factors between NSCLC and normal tissues by analyzing mRNA profiling from The Cancer Genome Atlas (TCGA) database program were identified. Weighted correlation network analysis (WGCNA) and line plot of least absolute shrinkage and selection operator (LASSO) were performed to find prognosis-related transcription factors. The cellular functions of transcription factors were performed by 5-ethynyl-2'-deoxyuridine (EdU) assay, wound healing assay, cell invasion assay in lung cancer cells. Results We identified 725 differentially expressed transcription factors between NSCLC and normal tissues. Three highly related modules for survival were discovered, and transcription factors highly associated with survival were obtained by using WGCNA. Then line plot of LASSO was applied to screen transcription factors related to prognosis and build a prognostic model. Consequently, SETDB2, SNAI3, SCML4, and ZNF540 were identified as prognosis-related transcription factors and validated in multiple databases. The low expression of these hub genes in NSCLC was associated with poor prognosis. The deletions of both SETDB2 and SNAI3 were found to promote proliferation, invasion, and stemness in lung cancer cells. Furthermore, there were significant differences in the proportions of 22 immune cells between the high- and low-score groups. Discussion Therefore, our study identified the transcription factors involved in regulating NSCLC, and we constructed a panel for the prediction of prognosis and immune infiltration to inform the clinical application of transcription factor analysis in the prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Jingyao Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinuo Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Baowen Yuan
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Qin
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong Wang
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hefen Yu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xu Teng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yunkai Yang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zou
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Zhang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- *Correspondence: Wei Huang, ; Yan Wang,
| | - Yan Wang
- Key Laboratory of Cancer and Microbiome, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Wei Huang, ; Yan Wang,
| |
Collapse
|
6
|
Khatiwada P, Rimal U, Malla M, Han Z, Shemshedini L. Peptides disrupting TM4SF3 interaction with AR or AR-V7 block prostate cancer cell proliferation. ENDOCRINE ONCOLOGY (BRISTOL, ENGLAND) 2023; 3:e230010. [PMID: 37822366 PMCID: PMC10563598 DOI: 10.1530/eo-23-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/29/2023] [Indexed: 10/13/2023]
Abstract
Androgen receptor (AR) plays a vital role in the development and progression of prostate cancer from the primary stage to the usually lethal stage known as castration-resistant prostate cancer (CRPC). Constitutively active AR splice variants (AR-Vs) lacking the ligand-binding domain are partially responsible for the abnormal activation of AR and may be involved in resistance to AR-targeting drugs occurring in CRPC. There is increasing consensus on the potential of drugs targeting protein-protein interactions. Our lab has recently identified transmembrane 4 superfamily 3 (TM4SF3) as a critical interacting partner for AR and AR-V7 and mapped the minimal interaction regions. Thus, we hypothesized that these interaction domains can be used to design peptides that can disrupt the AR/TM4SF3 interaction and kill prostate cancer cells. Peptides TA1 and AT1 were designed based on the TM3SF3 or AR interaction domain, respectively. TA1 or AT1 was able to decrease AR/TM4SF3 protein interaction and protein stability. Peptide TA1 reduced the recruitment of AR and TM4SF3 to promoters of androgen-regulated genes and subsequent activation of these AR target genes. Peptides TA1 and AT1 were strongly cytotoxic to prostate cancer cells that express AR and/or AR-V7. Peptide TA1 inhibited the growth and induced apoptosis of both enzalutamide-sensitive and importantly enzalutamide-resistant prostate cancer cells. TA1 also blocked the migration and malignant transformation of prostate cancer cells. Our data clearly demonstrate that using peptides to target the important interaction AR has with TM4SF3 provides a novel method to kill enzalutamide-resistant prostate cancer cells that can potentially lead to new more effective therapy for CRPC.
Collapse
Affiliation(s)
- Prabesh Khatiwada
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
- Center for Translational Immunology, Columbia University, New York, NY, USA
| | - Ujjwal Rimal
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Mamata Malla
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Zhengyang Han
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| | - Lirim Shemshedini
- Department of Biological Sciences, University of Toledo, Toledo, Ohio, USA
| |
Collapse
|
7
|
Han Z, Rimal U, Khatiwada P, Brandman J, Zhou J, Hussain M, Viola RE, Shemshedini L. Dual-Acting Peptides Target EZH2 and AR: A New Paradigm for Effective Treatment of Castration-Resistant Prostate Cancer. Endocrinology 2022; 164:6775160. [PMID: 36288553 DOI: 10.1210/endocr/bqac180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Indexed: 01/16/2023]
Abstract
Prostate cancer starts as a treatable hormone-dependent disease, but often ends in a drug-resistant form called castration-resistant prostate cancer (CRPC). Despite the development of the antiandrogens enzalutamide and abiraterone for CRPC, which target the androgen receptor (AR), drug resistance usually develops within 6 months and metastatic CRPC (mCRPC) leads to lethality. EZH2, found with SUZ12, EED, and RbAP48 in Polycomb repressive complex 2 (PRC2), has emerged as an alternative target for the treatment of deadly mCRPC. Unfortunately, drugs targeting EZH2 have shown limited efficacy in mCRPC. To address these failures, we have developed novel, dual-acting peptide inhibitors of PRC2 that uniquely target the SUZ12 protein component, resulting in the inhibition of both PRC2 canonical and noncanonical functions in prostate cancer. These peptides were found to inhibit not only the EZH2 methylation activity, but also block its positive effect on AR gene expression in prostate cancer cells. Since the peptide effect on AR levels is transcriptional, the inhibitory peptides can block the expression of both full-length AR and its splicing variants including AR-V7, which plays a significant role in the development of drug resistance. This dual-mode action provides the peptides with the capability to kill enzalutamide-resistant CRPC cells. These peptides are also more cytotoxic to prostate cancer cells than the combination of enzalutamide and an EZH2 inhibitory drug, which was recently suggested to be an effective treatment of mCRPC disease. Our data show that such a dual-acting therapeutic approach can be more effective than the existing front-line drug therapies for treating deadly mCRPC.
Collapse
Affiliation(s)
- Zhengyang Han
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Ujjwal Rimal
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Prabesh Khatiwada
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Jacob Brandman
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Jun Zhou
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Muhammad Hussain
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Ronald E Viola
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| | - Lirim Shemshedini
- Department of Biological Sciences and Department of Chemistry and Biochemistry, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
8
|
Zhang T, Wang Y, Xie M, Ji X, Luo X, Chen X, Zhang B, Liu D, Feng Y, Sun M, Huang W, Xia L. HGF-mediated elevation of ETV1 facilitates hepatocellular carcinoma metastasis through upregulating PTK2 and c-MET. J Exp Clin Cancer Res 2022; 41:275. [PMID: 36109787 PMCID: PMC9479266 DOI: 10.1186/s13046-022-02475-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Metastasis is a major determinant of death in patients with hepatocellular carcinoma (HCC). Dissecting key molecular mediators that promote this malignant feature may help yield novel therapeutic insights. Here, we investigated the role of E-twenty-six transformation-specific variant 1 (ETV1), a member of the E-twenty-six transformation-specific (ETS) family, in HCC metastasis. Methods The clinical significance of ETV1 and its target genes in two independent cohorts of HCC patients who underwent curative resection were assessed by Kaplan–Meier analysis and Multivariate Cox proportional hazards model. Luciferase reporter assay and chromatin immunoprecipitation assay were used to detect the transcriptional regulation of target gene promoters by ETV1. The effect of ETV1 on invasiveness and metastasis of HCC were detected by transwell assays and the orthotopically metastatic model. Results ETV1 expression was frequently elevated in human HCC specimens. Increased ETV1 expression was associated with the malignant biological characteristics and poor prognosis of HCC patients. ETV1 facilitated invasion and metastasis of HCC cells in vitro and in vivo. Mechanistically, ETV1 promoted HCC metastasis via upregulating metastasis-related genes, including protein tyrosine kinase 2 (PTK2) and MET. Down-regulated the expression of PTK2 or tyrosine protein kinase Met (c-MET) decreased ETV1-mediated HCC metastasis. Hepatocyte growth factor (HGF) upregulated ETV1 expression through activating c-MET-ERK1/2-ELK1 pathway. Notably, in two independent cohorts, patients with positive coexpression of ETV1/PTK2 or ETV1/c-MET had worse prognosis. Furthermore, the combination of PTK2 inhibitor defactinib and c-MET inhibitor capmatinib significantly suppressed HCC metastasis induced by ETV1. Conclusion This study uncovers functional and prognostic roles for ETV1 in HCC and exposes a positive feedback loop of HGF-ERK1/2-ETV1-c-MET. Targeting this pathway may provide a potential therapeutic intervention for ETV1-overexpressing HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02475-2.
Collapse
|
9
|
MicroRNA-149 suppresses osteogenic differentiation of mesenchymal stem cells via inhibition of AKT1-dependent Twist1 phosphorylation. Cell Death Dis 2022; 8:2. [PMID: 35013126 PMCID: PMC8748629 DOI: 10.1038/s41420-021-00618-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 11/08/2022]
Abstract
Osteogenic differentiation is a vital process for growth, repair, and remodeling of bones. Accumulating evidence have suggested that microRNAs (miRNAs or miRs) play a crucial role in osteogenic differentiation of mesenchymal stem cells (MSCs). Hence, the current study set out to elucidate the role of miR-149 in osteogenic differentiation of MSCs and the underlying mechanism. First, rat models of bone differentiation were established using the Masquelet-induced membrane technique, and MSCs were isolated. The expression of miR-149 and AKT1 in the rats and cells was detected with RT-qPCR and western blot analysis. The relationships among miR-149, AKT1, and Twist1 were further predicted by online bioinformatics prediction and verified using dual luciferase reporter gene assay. Alteration of miR-149, AKT1, or Twist1 was performed to further explore their effect on osteogenic differentiation of MSCs. miR-149 was poorly expressed in the process of osteogenic differentiation of MSCs, while AKT1 was highly expressed. miR-149 negatively regulated the expression of AKT1, which in turn diminished the protein levels of Twist1 and promoted the phosphorylation levels of Twist1. Lastly, miR-149 acted as an inhibitor of osteogenic differentiation of MSCs, which could be reversed by AKT1. To sum up, miR-149 silencing promoted osteogenic differentiation of MSCs by enhancing Twist1 degradation through AKT1 upregulation, representing a new method for bone repair treatment.
Collapse
|
10
|
You B, Sun Y, Luo J, Wang K, Liu Q, Fang R, Liu B, Chou F, Wang R, Meng J, Huang CP, Yeh S, Chang C, Xu W. Androgen receptor promotes renal cell carcinoma (RCC) vasculogenic mimicry (VM) via altering TWIST1 nonsense-mediated decay through lncRNA-TANAR. Oncogene 2021; 40:1674-1689. [PMID: 33510354 PMCID: PMC7932923 DOI: 10.1038/s41388-020-01616-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/30/2020] [Accepted: 12/10/2020] [Indexed: 01/30/2023]
Abstract
While the androgen receptor (AR) may influence the progression of clear cell renal cell carcinoma (ccRCC), its role to impact vasculogenic mimicry (VM) to alter the ccRCC progression and metastasis remains obscure. Here, we demonstrated that elevated AR expression was positively correlated with tumor-originated vasculogenesis in ccRCC patients. Consistently, in vitro research revealed AR promoted VM formation in ccRCC cell lines via modulating lncRNA-TANAR/TWIST1 signals. Mechanism dissection showed that AR could increase lncRNA-TANAR (TANAR) expression through binding to the androgen response elements (AREs) located in its promoter region. Moreover, we found that TANAR could impede nonsense-mediated mRNA decay (NMD) of TWIST1 mRNA by direct interaction with TWIST1 5'UTR. A preclinical study using in vivo mouse model with orthotopic xenografts of ccRCC cells further confirmed the in vitro data. Together, these results illustrated that AR-mediated TANAR signals might play a crucial role in ccRCC VM formation and metastasis, and targeting this newly identified AR/TANAR/TWIST1 signaling may help in the development of a novel anti-angiogenesis therapy to better suppress the ccRCC progression.
Collapse
Affiliation(s)
- Bosen You
- Department of Urology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Department of Urology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Yin Sun
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Jie Luo
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Keliang Wang
- Department of Urology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Qing Liu
- Department of Urology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Ruizhe Fang
- Department of Urology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Bingmei Liu
- Department of Pathology and Cutaneous Oncology, Heilongjiang Provincial Hospital, Harbin, 150001, China
| | - Fuju Chou
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Ronghao Wang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Jialin Meng
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Chi-Ping Huang
- Sex Hormone Research Center and Departments of Urology, China Medical University/Hospital, Taichung, 404, Taiwan
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Pathology and Urology, and The Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14646, USA.
- Sex Hormone Research Center and Departments of Urology, China Medical University/Hospital, Taichung, 404, Taiwan.
| | - Wanhai Xu
- Department of Urology, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
11
|
Yin L, Li J, Wang J, Pu T, Wei J, Li Q, Wu BJ. MAOA promotes prostate cancer cell perineural invasion through SEMA3C/PlexinA2/NRP1-cMET signaling. Oncogene 2021; 40:1362-1374. [PMID: 33420365 DOI: 10.1038/s41388-020-01615-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/05/2023]
Abstract
Perineural invasion (PNI), a pathologic feature defined as cancer cell invasion in, around, and through nerves, is an indicator of poor prognosis and survival in prostate cancer (PC). Despite widespread recognition of the clinical significance of PNI, the molecular mechanisms are largely unknown. Here, we report that monoamine oxidase A (MAOA) is a clinically and functionally important mediator of PNI in PC. MAOA promotes PNI of PC cells in vitro and tumor innervation in an orthotopic xenograft model. Mechanistically, MAOA activates SEMA3C in a Twist1-dependent transcriptional manner, which in turn stimulates cMET to facilitate PNI via autocrine or paracrine interaction with coactivated PlexinA2 and NRP1. Furthermore, MAOA inhibitor treatment effectively reduces PNI of PC cells in vitro and tumor-infiltrating nerve fiber density along with suppressed xenograft tumor growth and progression in mice. Collectively, these findings characterize the contribution of MAOA to the pathogenesis of PNI and provide a rationale for using MAOA inhibitors as a targeted treatment for PNI in PC.
Collapse
Affiliation(s)
- Lijuan Yin
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.,Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.,Laboratory of Regeneromics, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Tianjie Pu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA
| | - Qinlong Li
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
12
|
Qi T, Qu Q, Li G, Wang J, Zhu H, Yang Z, Sun Y, Lu Q, Qu J. Function and regulation of the PEA3 subfamily of ETS transcription factors in cancer. Am J Cancer Res 2020; 10:3083-3105. [PMID: 33163259 PMCID: PMC7642666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/17/2020] [Indexed: 06/11/2023] Open
Abstract
The PEA3 subfamily is a subgroup of the E26 transformation-specific (ETS) family. Its members, ETV1, ETV4, and ETV5, have been found to be overexpressed in multiple cancers. The deregulation of ETV1, ETV4, and ETV5 induces cell growth, invasion, and migration in various tumor cells, leading to tumor progression, metastasis, and drug resistance. Therefore, exploring drugs or therapeutic targets that target the PEA3 subfamily may contribute to the clinical treatment of tumor patients. In this review, we introduce the structures and functions of the PEA3 subfamily members, systematically review their main roles in various tumor cells, analyze their prognostic and diagnostic value, and, finally, introduce several molecular targets and therapeutic drugs targeting ETV1, ETV4, and ETV5. We conclude that targeting a series of upstream regulators and downstream target genes of the PEA3 subfamily may be an effective strategy for the treatment of ETV1/ETV4/ETV5-overexpressing tumors.
Collapse
Affiliation(s)
- Tingting Qi
- Department of Pharmacy, The Second Xiangya Hospital, Central South UniversityChangsha 410011, PR China
- Institute of Clinical Pharmacy, Central South UniversityChangsha 410011, PR China
| | - Qiang Qu
- Department of Pharmacy, Xiangya Hospital, Central South UniversityChangsha 410007, PR China
| | - Guohua Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South UniversityChangsha 410011, PR China
- Institute of Clinical Pharmacy, Central South UniversityChangsha 410011, PR China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South UniversityChangsha 410011, PR China
- Institute of Clinical Pharmacy, Central South UniversityChangsha 410011, PR China
| | - Haihong Zhu
- Department of Pharmacy, The Second Xiangya Hospital, Central South UniversityChangsha 410011, PR China
- Institute of Clinical Pharmacy, Central South UniversityChangsha 410011, PR China
| | - Zhi Yang
- Department of General Surgery, Xiangya Hospital, Central South UniversityChangsha 410007, PR China
| | - Yuesheng Sun
- Department of General Surgery, The Third Clinical College of Wenzhou Medical University, Wenzhou People’s HospitalWenzhou 325000, PR China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South UniversityChangsha 410011, PR China
- Institute of Clinical Pharmacy, Central South UniversityChangsha 410011, PR China
| | - Jian Qu
- Department of Pharmacy, The Second Xiangya Hospital, Central South UniversityChangsha 410011, PR China
- Institute of Clinical Pharmacy, Central South UniversityChangsha 410011, PR China
| |
Collapse
|
13
|
Capicua in Human Cancer. Trends Cancer 2020; 7:77-86. [PMID: 32978089 DOI: 10.1016/j.trecan.2020.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
Abstract
Capicua (CIC) is a highly conserved transcriptional repressor that is differentially regulated through mitogen-activated protein kinase (MAPK) signaling or genetic alteration across human cancer. CIC contributes to tumor progression and metastasis through direct transcriptional control of effector target genes. Recent findings indicate that CIC dysregulation is mechanistically linked and restricted to specific cancer subtypes, yet convergence on key downstream transcriptional nodes are critical for CIC-regulated oncogenesis across these cancers. In this review, we focus on how differential regulation of CIC through functional and genetic mechanisms contributes to subtype-specific cancer phenotypes and we propose new therapeutic strategies to effectively target CIC-altered cancers.
Collapse
|