1
|
Franz MJ, Wenisch P, Wohlleben P, Rupprecht L, Chubanov V, Gudermann T, Kyheröinen S, Vartiainen MK, Heinrich MR, Muehlich S. Identification of novel inhibitors of the transcriptional coactivator MRTF-A for HCC therapy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200855. [PMID: 39262570 PMCID: PMC11387234 DOI: 10.1016/j.omton.2024.200855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
Myocardin-related transcription factor A (MRTF-A) is a coactivator of serum response factor (SRF), which regulates the expression of genes involved in cell proliferation, migration, and differentiation and has been implicated in hepatocellular carcinoma (HCC) progression. We recently established inhibition of the transcriptional activity of MRTF-A by NS8593 as a novel therapeutic approach for HCC therapy. NS8593 is a negative gating modulator of the transient receptor potential cation channel TRPM7. In this report, we identify an aminobenzimidazole that is highly potent in inhibiting TRPM7 and its interaction with RhoA, leading to decreased SRF transcriptional activity and enhanced nuclear export of MRTF-A, as determined by fluorescence loss in photobleaching (FLIP). This resulted in reduced expression of the MRTF/SRF target genes transforming growth factor β1 (TGF-β1) and tetraspanin 5 (TSPAN5), senescence induction, and growth arrest in HCC cells. Replacement of the tetraline core by a 3-aminophenyl substructure yielded inhibitor 10 with higher potency than inhibitor 5, and further structural modifications yielded highly potent inhibitors of SRF activity, 14 and 16. Both compounds were capable of inhibiting cell proliferation and inducing senescence in HCC cells with improved efficacy compared to NS8593. These inhibitors represent valuable tools for understanding the molecular basis of drug development targeting TRPM7 and MRTFs.
Collapse
Affiliation(s)
- Miriam Jasmin Franz
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Pia Wenisch
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Petra Wohlleben
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Laura Rupprecht
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336 München, Germany
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität München, Goethestraße 33, 80336 München, Germany
| | - Salla Kyheröinen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Viikinkaari 5d, 00790 Helsinki, Finland
| | | | - Markus R Heinrich
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- FAU NeW-Research Center for New Bioactive Compounds, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| | - Susanne Muehlich
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
- FAU NeW-Research Center for New Bioactive Compounds, Nikolaus-Fiebiger-Straße 10, 91058 Erlangen, Germany
| |
Collapse
|
2
|
Luo X, Liu P, Ye X, He J, Lai Y, Lv Y, Wu X, Liu Y, Zhang Q, Yang H, Wei W, Deng C, Kuang S, Wu S, Xue Y, Rao F. Curcumin improves atrial fibrillation susceptibility by regulating tsRNA expression in aging mouse atrium. PeerJ 2024; 12:e17495. [PMID: 39076782 PMCID: PMC11285363 DOI: 10.7717/peerj.17495] [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: 06/29/2023] [Accepted: 05/09/2024] [Indexed: 07/31/2024] Open
Abstract
Age is an independent risk factor for atrial fibrillation (AF), and curcumin can delay aging related disease through reducing oxidative stress and inflammation. However, its target in aging-related AF remains unclear. Transfer RNA-derived small RNA (tsRNA) is a novel short non-coding RNA (sncRNA), and exerts a potential regulatory function in aging. This study was to explore the therapeutic targets of curcumin in atrium of aged mice by PANDORA-seq. Aged mice (18 month) were treated with curcumin (100 mg/kg). Rapid transjugular atrial pacing was performed to observe AF inducibility. SA-β-gal staining, reactive oxygen species (ROS) detection and qRT-PCR were used to assess the degree of aging and oxidative stress/inflammation levels. PANDORA-seq was performed to reveal the differentially expressed sncRNAs in the atrium of mice. The results showed that curcumin reduced the susceptibility AF of aged mice by improving aging-related atrial fibrosis. Compared to young mice (5 month) group, aged mice yielded 473 significantly altered tsRNA sequences, while 947 tsRNA sequences were significantly altered after treated with curcumin. Enrichment analysis revealed that the target genes were mainly related to DNA damage and protein modification. Compared with the 5 month group, the expression levels of mature-mt_tRNA-Val-TAC_CCA_end, mature-mt_tRNA-Glu-TTC_CCA_end, and mature-tRNA-Asp-GTC_CCA_end were up-regulated in the 18 month group, while the expression of mature-mt_tRNA-Thr-TGT_5_end was down-regulated. This trend was reversed in the 18 month + curcumin group. Increased cellular ROS levels, inflammation expression and senescence in aged mice atrium were improved by the down-regulation of mature-mt_tRNA-Val-TAC_CCA_end. In conclusion, our findings identified mature-mt_tRNA-Val-TAC_CCA_end participated in the mechanism of aging-related atrial fibrosis, providing new intervention target of aging-related AF.
Collapse
Affiliation(s)
- Xueshan Luo
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Panyue Liu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Xingdong Ye
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Jintao He
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Yingyu Lai
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Yidong Lv
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Xiongbin Wu
- Jiangmen Central Hospital, Jiangmen, Guangdong, China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Qianhuan Zhang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Hui Yang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Wei Wei
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Chunyu Deng
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Sujuan Kuang
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Shulin Wu
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Yumei Xue
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| | - Fang Rao
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangzhou, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
- South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Zhou J, Sun X, Chen X, Liu H, Miao X, Guo Y, Fan Z, Li J, Xu Y, Li Z. Phosphatidic acid-enabled MKL1 contributes to liver regeneration: Translational implication in liver failure. Acta Pharm Sin B 2024; 14:256-272. [PMID: 38261867 PMCID: PMC10793099 DOI: 10.1016/j.apsb.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 01/25/2024] Open
Abstract
Liver regeneration following injury aids the restoration of liver mass and the recovery of liver function. In the present study we investigated the contribution of megakaryocytic leukemia 1 (MKL1), a transcriptional modulator, to liver regeneration. We report that both MKL1 expression and its nuclear translocation correlated with hepatocyte proliferation in cell and animal models of liver regeneration and in liver failure patients. Mice with MKL1 deletion exhibited defective regenerative response in the liver. Transcriptomic analysis revealed that MKL1 interacted with E2F1 to program pro-regenerative transcription. MAPKAPK2 mediated phosphorylation primed MKL1 for its interaction with E2F1. Of interest, phospholipase d2 promoted MKL1 nuclear accumulation and liver regeneration by catalyzing production of phosphatidic acid (PA). PA administration stimulated hepatocyte proliferation and enhanced survival in a MKL1-dependent manner in a pre-clinical model of liver failure. Finally, PA levels was detected to be positively correlated with expression of pro-regenerative genes and inversely correlated with liver injury in liver failure patients. In conclusion, our data reveal a novel mechanism whereby MKL1 contributes to liver regeneration. Screening for small-molecule compounds boosting MKL1 activity may be considered as a reasonable approach to treat acute liver failure.
Collapse
Affiliation(s)
- Jiawen Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Xinyue Sun
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Xuelian Chen
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Huimin Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
| | - Xiulian Miao
- Institute of Biomedical Research, Liaocheng University, Liaocheng 252200, China
| | - Yan Guo
- Institute of Biomedical Research, Liaocheng University, Liaocheng 252200, China
| | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital Affiliated with Nanjing University Medical School, Nanjing 210008, China
- Institute of Viruses and Infectious Diseases, Nanjing University, Nanjing 210008, China
| | - Yong Xu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
- Institute of Biomedical Research, Liaocheng University, Liaocheng 252200, China
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 211198, China
- Institute of Biomedical Research, Liaocheng University, Liaocheng 252200, China
| |
Collapse
|
4
|
Wang Y, Wang Y, Zhang J, Shi Z, Liu J. LncRNA NONHSAT227443.1 Confers Esophageal Squamous Cell Carcinoma Chemotherapy Resistance by Activating PI3K/AKT Signaling via Targeting MRTFB. Technol Cancer Res Treat 2024; 23:15330338241274369. [PMID: 39150441 PMCID: PMC11329966 DOI: 10.1177/15330338241274369] [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: 02/20/2024] [Revised: 06/04/2024] [Accepted: 07/08/2024] [Indexed: 08/17/2024] Open
Abstract
INTRODUCTION Esophageal cancer presents significant challenges due to limited treatment options and poor prognosis, particularly in advanced stages. Dysregulated long non-coding RNAs (lncRNAs) are implicated in cancer progression and treatment resistance. This study investigated the roles of dysregulated lncRNA NONHSAT227443.1, identified through lncRNA-seq, and its downstream target gene MRTFB in esophageal squamous cell carcinoma (ESCC). METHODS Dysregulated lncRNAs were identified through lncRNA-seq in esophageal cancer tissues with varying chemotherapy response. The regulatory interaction of overexpressed NONHSAT227443.1 was assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. Functional assays, including cell viability, cell proliferation, and flow cytometry analyses, were performed to comprehensively investigate the influence of NONHSAT227443.1 and its downstream molecules on ESCC. RESULTS NONHSAT227443.1 was significantly overexpressed in paclitaxel plus platinum chemotherapy non-responders and esophageal cancer cell lines. Chemotherapy exposure led to diminished NONHSAT227443.1 expression. NONHSAT227443.1 negatively regulated MRTFB expression, and their combined dysregulation correlated with increased cancer activity, proliferation, and suppressed apoptosis. Diminished MRTFB expression was associated with PI3K/AKT pathway activation. CONCLUSION Our study provides insights into NONHSAT227443.1 and MRTFB roles in esophageal cancer, contributing to aggressive traits and treatment resistance. NONHSAT227443.1 and MRTFB may serve as potential therapeutic targets to enhance the response to paclitaxel plus platinum chemotherapy in non-responsive cases.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yingying Wang
- Department of Functional Region of Diagnosis, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jinze Zhang
- Department of Thoracic Endoscopy Room, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhihua Shi
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Junfeng Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
5
|
Ma F, Ma Y, Liu K, Gao J, Li S, Sun X, Li G. Resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis. Food Funct 2023; 14:1484-1497. [PMID: 36651299 DOI: 10.1039/d2fo01188c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inducing cell senescence is widely regarded as a potent tumor suppression mechanism. Resveratrol has attracted increasing attention for its capacity to prevent and suppress cancer. However, the mechanism of resveratrol on the induction of cancer cell senescence has not been well clarified. Our results showed that resveratrol inhibited cell viability and colony formation and promoted cell senescence along with augmentation of SA-β-gal activity and modulation of senescence-associated molecular markers p53, p21 and LaminB protein in breast and liver cancer cells. The underlying mechanism was that resveratrol increased ROS generation to enhance tumor suppressor gene DLC1 expression, and DLC1 further inhibited the DYRK1A-EGFR axis to trigger DNA damage accompanied by up-regulation of the DNA double strand break marker protein γH2AX and down-regulation of the DNA repair related proteins p-BRCA1 and RAD51, eventually leading to cancer cell senescence. Resveratrol also effectively inhibited the volume of transplanted tumor with increased SA-β-gal activity and DLC1 level in a chicken embryo allantoic membrane xenograft tumor model. This is the first report to investigate whether resveratrol induces DNA damage-mediated cancer cell senescence through the DLC1-DYRK1A-EGFR axis, which could provide a solid base for resveratrol's application in cancer prevention and clinical treatment as a food additive or adjuvant therapies.
Collapse
Affiliation(s)
- Fengqiu Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Yan Ma
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Keke Liu
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Junying Gao
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Shasha Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Xiaowen Sun
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant Biology, School of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| |
Collapse
|
6
|
Konopa A, Meier MA, Franz MJ, Bernardinelli E, Voegele AL, Atreya R, Ribback S, Roessler S, Aigner A, Singer K, Singer S, Sarikas A, Muehlich S. LPA receptor 1 (LPAR1) is a novel interaction partner of Filamin A that promotes Filamin A phosphorylation, MRTF-A transcriptional activity and oncogene-induced senescence. Oncogenesis 2022; 11:69. [PMID: 36577757 PMCID: PMC9797565 DOI: 10.1038/s41389-022-00445-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
Myocardin-related transcription factors A and B (MRTFs) are coactivators of Serum Response Factor (SRF), which controls fundamental biological processes such as cell growth, migration, and differentiation. MRTF and SRF transcriptional activity play an important role in hepatocellular carcinoma (HCC) growth, which represents the second leading cause of cancer-related mortality in humans worldwide. We, therefore, searched for druggable targets in HCC that regulate MRTF/SRF transcriptional activity and can be exploited therapeutically for HCC therapy. We identified the G protein-coupled lysophosphatidic acid receptor 1 (LPAR1) as a novel interaction partner of MRTF-A and Filamin A (FLNA) using fluorescence resonance energy transfer-(FRET) and proximity ligation assay (PLA) in vitro in HCC cells and in vivo in organoids. We found that LPAR1 promotes FLNA phosphorylation at S2152 which enhances the complex formation of FLNA and MRTF-A, actin polymerization, and MRTF transcriptional activity. Pharmacological blockade or depletion of LPAR1 prevents FLNA phosphorylation and complex formation with MRTF-A, resulting in reduced MRTF/SRF target gene expression and oncogene-induced senescence. Thus, inhibition of the LPAR1-FLNA-MRTF-A interaction represents a promising strategy for HCC therapy.
Collapse
Affiliation(s)
- Andreas Konopa
- grid.5330.50000 0001 2107 3311Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Melanie A. Meier
- grid.5330.50000 0001 2107 3311Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Miriam J. Franz
- grid.5330.50000 0001 2107 3311Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Emanuele Bernardinelli
- grid.21604.310000 0004 0523 5263Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Anna-Lena Voegele
- grid.5330.50000 0001 2107 3311Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Raja Atreya
- grid.5330.50000 0001 2107 3311Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Silvia Ribback
- grid.5603.0Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Stephanie Roessler
- grid.7700.00000 0001 2190 4373Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Achim Aigner
- grid.9647.c0000 0004 7669 9786Rudolf Boehm Institute of Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Kerstin Singer
- grid.411544.10000 0001 0196 8249Department for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Stephan Singer
- grid.411544.10000 0001 0196 8249Department for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Antonio Sarikas
- grid.21604.310000 0004 0523 5263Institute of Pharmacology and Toxicology, Paracelsus Medical University, Salzburg, Austria
| | - Susanne Muehlich
- grid.5330.50000 0001 2107 3311Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
7
|
Wan C, Liu W, Jiang L, Dong S, Ma W, Wang S, Liu D. Knockdown of MKL1 ameliorates oxidative stress-induced chondrocyte apoptosis and cartilage matrix degeneration by activating TWIST1-mediated PI3K/AKT signaling pathway in rats. Autoimmunity 2022; 55:559-566. [PMID: 36046946 DOI: 10.1080/08916934.2022.2114466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Studies have reported that megakaryocytic leukemia 1 (MKL1) is closely related to the pathological process of a variety of inflammatory diseases, but its role in osteoarthritis (OA) needs to be clarified. This study aimed to investigate the regulatory role of MKL1 in oxidative stress-induced chondrocyte apoptosis and cartilage matrix degeneration. The expressions of target mRNAs and proteins were measured by using reverse transcription-quantitative polymerase chain reaction and western blotting. ELISA assay was used to measure the levels of IL-6, IL-8, and TNF-α in chondrocytes. And commercial kits based on different spectrophotometry or colorimetry methods were performed to validate oxidative stress. CCK-8 and apoptosis kits were used to determine cell viability and apoptosis. Rat OA model was established by anterior cruciate ligament transection (ACLT), and the expression of MKL1 was interfered by injecting sh-MKL1 lentiviral vector into caudal vein. The results showed that the expression of MKL1was induced by H2O2 in chondrocytes. Knockdown of MKL1 alleviated H2O2-induced inflammation and cell apoptosis, reduced H2O2-induced oxidative stress, and improved cartilage matrix degeneration of chondrocytes. Besides, inhibition of MKL1 regulated the activation of TWIST1-mediated PI3K/AKT signaling. Further studies have found that TWIST1-mediated PI3K/AKT signaling was involved in the regulation mechanism of MKL1 on chondrocyte apoptosis and cartilage matrix degeneration. Next, intervention with MKL1 inhibited the progression of OA in rats. These results demonstrated that MKL1 regulate the apoptosis and cartilage matrix degeneration of chondrocytes via TWIST1-mediated PI3K/AKT signaling.
Collapse
Affiliation(s)
- Chao Wan
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, China
| | - Wei Liu
- Department of Pathophysiology, Binzhou Medical University, Binzhou, Shandong, China
| | - Limin Jiang
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, China
| | - Shengjie Dong
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, China
| | - Weihua Ma
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, China
| | - Shijun Wang
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, China
| | - Dan Liu
- Department of the Joint and Bone Surgery, Yantaishan Hospital, Yantai, Shandong, China
| |
Collapse
|
8
|
Liu Y, Yang H, Liu Q, Pan M, Wang D, Pan S, Zhang W, Wei J, Zhao X, Ji J. Selenocystine-Derived Label-Free Fluorescent Schiff Base Nanocomplex for siRNA Delivery Synergistically Kills Cancer Cells. Molecules 2022; 27:1302. [PMID: 35209090 PMCID: PMC8878402 DOI: 10.3390/molecules27041302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Chemo and siRNA synergic treatments for tumors is a promising new therapeutic trend. Selenocystine, a selenium analog of cysteine, has been considered a potential antitumor agent due to its redox perturbing role. In this study, we developed a nanocarrier for siRNA based on a selenocystine analog engineered polyetherimide and achieved traceable siRNA delivery and the synergic killing of tumor cells. Notably, we applied the label-free Schiff base fluorescence mechanism, which enabled us to trace the siRNA delivery and to monitor the selenocystine analogs' local performance. A novel selenocystine-derived fluorescent Schiff base linker was used to crosslink the polyetherimide, thereby generating a traceable siRNA delivery vehicle with green fluorescence. Moreover, we found that this compound induced tumor cells to undergo senescence. Together with the delivery of a siRNA targeting the anti-apoptotic BCL-xl/w genes in senescent cells, it achieved a synergistic inhibition function by inducing both senescence and apoptosis of tumor cells. Therefore, this study provides insights into the development of label-free probes, prodrugs, and materials towards the synergic strategies for cancer therapy.
Collapse
Affiliation(s)
- Yang Liu
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou 310058, China;
| | - Haoying Yang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Qian Liu
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Mingming Pan
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Danli Wang
- Zhoushan Hospital of Zhejiang Province, Zhoushan 316004, China;
| | - Shiyuan Pan
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Weiran Zhang
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Jinfeng Wei
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Xiaowei Zhao
- Henan Key Laboratory of Brain Targeted Bio-Nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng 475004, China; (H.Y.); (Q.L.); (M.P.); (S.P.); (W.Z.)
| | - Junfeng Ji
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou 310058, China;
| |
Collapse
|
9
|
Fixing the GAP: the role of RhoGAPs in cancer. Eur J Cell Biol 2022; 101:151209. [DOI: 10.1016/j.ejcb.2022.151209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
|
10
|
Liu Z, Sun J, Li C, Xu L, Liu J. MKL1 regulates hepatocellular carcinoma cell proliferation, migration and apoptosis via the COMPASS complex and NF-κB signaling. BMC Cancer 2021; 21:1184. [PMID: 34742274 PMCID: PMC8571910 DOI: 10.1186/s12885-021-08185-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/14/2021] [Indexed: 12/18/2022] Open
Abstract
Background Histone modification plays essential roles in hepatocellular carcinoma (HCC) pathogenesis, but the regulatory mechanisms remain poorly understood. In this study, we aimed to analyze the roles of Megakaryoblastic leukemia 1 (MKL1) and its regulation of COMPASS (complex of proteins associated with Set1) in HCC cells. Methods MKL1 expression in clinical tissues and cell lines were detected by bioinformatics, qRT-PCR and western blot. MKL1 expression in HCC cells were silenced with siRNA, followed by cell proliferation evaluation via Edu staining and colony formation, migration and invasion using the Transwell system, and apoptosis by Hoechst staining. HCC cell tumorigenesis was assessed by cancer cell line-based xenograft model, combined with H&E staining and IHC assays. Results MKL1 expression was elevated in HCC cells and clinical tissues which was correlated with poor prognosis. MKL1 silencing significantly repressed proliferation, migration, invasion and colony formation but enhanced apoptosis in HepG2 and Huh-7 cells. MKL1 silencing also inhibited COMPASS components and p65 protein expression in HepG2 and Huh-7 cells. HepG2 cell tumorigenesis in nude mice was severely impaired by MKL1 knockdown, resulted into suppressed Ki67 expression and cell proliferation. Conclusion MKL1 promotes HCC pathogenesis by regulating hepatic cell proliferation, migration and apoptosis via the COMPASS complex and NF-κB signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08185-w.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiuzheng Sun
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanzhi Li
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Liyou Xu
- Department of Hepatobiliary and Pancreatic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jun Liu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China. .,Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
11
|
Schreyer L, Mittermeier C, Franz MJ, Meier MA, Martin DE, Maier KC, Huebner K, Schneider-Stock R, Singer S, Holzer K, Fischer D, Ribback S, Liebl B, Gudermann T, Aigner A, Muehlich S. Tetraspanin 5 (TSPAN5), a Novel Gatekeeper of the Tumor Suppressor DLC1 and Myocardin-Related Transcription Factors (MRTFs), Controls HCC Growth and Senescence. Cancers (Basel) 2021; 13:cancers13215373. [PMID: 34771537 PMCID: PMC8582588 DOI: 10.3390/cancers13215373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) ranks second among the leading causes of cancer-related death. Since current therapeutic options are very limited, a deeper understanding of the molecular mechanisms underlying the tumor onset and progression of HCC holds great potential for improved therapeutic options. Although it has been shown that deleted in liver cancer 1 (DLC1) acts as a tumor suppressor whose allele is lost in 50% of liver cancers, alterations in gene expression initiated by DLC1 loss have not yet been the primary focus of liver cancer research. To identify novel gene targets that allow for a personalized medicine approach for HCC therapy, we performed gene expression profiling for HepG2 cells stably expressing DLC1shRNA. We provide evidence that TSPAN5 is required for HCC growth, migration and invasion, and dissected the underlying molecular mechanisms involving myocardin-related transcription factors. Thus, TSPAN5 represents a novel therapeutic target for the treatment of HCC characterized by DLC1 loss. Abstract Human hepatocellular carcinoma (HCC) is among the most lethal and common cancers in the human population, and new molecular targets for therapeutic intervention are urgently needed. Deleted in liver cancer 1 (DLC1) was originally identified as a tumor suppressor gene in human HCC. DLC1 is a Rho-GTPase-activating protein (RhoGAP) which accelerates the return of RhoGTPases to an inactive state. We recently described that the restoration of DLC1 expression induces cellular senescence. However, this principle is not amenable to direct therapeutic targeting. We therefore performed gene expression profiling for HepG2 cells depleted of DLC1 to identify druggable gene targets mediating the effects of DLC1 on senescence induction. This approach revealed that versican (VCAN), tetraspanin 5 (TSPAN5) and N-cadherin (CDH2) were strongly upregulated upon DLC1 depletion in HCC cells, but only TSPAN5 affected the proliferation of HCC cells and human HCC. The depletion of TSPAN5 induced oncogene-induced senescence (OIS), mediated by the p16INK4a/pRb pathways. Mechanistically, silencing TSPAN5 reduced actin polymerization and thereby myocardin-related transcription factor A- filamin A (MRTF-A-FLNA) complex formation, resulting in decreased expression of MRTF/SRF-dependent target genes and senescence induction in vitro and in vivo. Our results identify TSPAN5 as a novel druggable target for HCC.
Collapse
Affiliation(s)
- Laura Schreyer
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (L.S.); (M.J.F.); (M.A.M.); (D.F.)
| | - Constanze Mittermeier
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Miriam J. Franz
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (L.S.); (M.J.F.); (M.A.M.); (D.F.)
| | - Melanie A. Meier
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (L.S.); (M.J.F.); (M.A.M.); (D.F.)
| | - Dietmar E. Martin
- Gene Center, Department of Chemistry and Pharmacy, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (D.E.M.); (K.C.M.)
| | - Kerstin C. Maier
- Gene Center, Department of Chemistry and Pharmacy, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; (D.E.M.); (K.C.M.)
| | - Kerstin Huebner
- Experimental Tumor Pathology, Institute of Pathology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (K.H.); (R.S.-S.)
| | - Regine Schneider-Stock
- Experimental Tumor Pathology, Institute of Pathology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (K.H.); (R.S.-S.)
| | - Stephan Singer
- Department for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.S.); (K.H.)
| | - Kerstin Holzer
- Department for Pathology, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.S.); (K.H.)
| | - Dagmar Fischer
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (L.S.); (M.J.F.); (M.A.M.); (D.F.)
| | - Silvia Ribback
- Institute for Pathology, University of Greifswald, 17475 Greifswald, Germany;
| | - Bernhard Liebl
- LGL Bayerisches Landesamt für Gesundheit und Lebensmittelsicherheit, 85764 Oberschleißheim, Germany;
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, 80336 Munich, Germany;
| | - Achim Aigner
- Rudolf Boehm Institute of Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, 04107 Leipzig, Germany;
| | - Susanne Muehlich
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany; (L.S.); (M.J.F.); (M.A.M.); (D.F.)
- Correspondence: ; Tel.: +49-(0)9131-8565665
| |
Collapse
|
12
|
Passi M, Zahler S. Mechano-Signaling Aspects of Hepatocellular Carcinoma. J Cancer 2021; 12:6411-6421. [PMID: 34659531 PMCID: PMC8489129 DOI: 10.7150/jca.60102] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
HCC is one of the leading causes of cancer related death worldwide and comprises about 90% of the cases of primary liver cancer. It is generally accompanied by chronic liver fibrosis characterised by deposition of collagen fibres, which, in turn, causes enhanced stiffness of the liver tissue. Changes of tissue stiffness give rise to alterations of signalling pathways that are associated to mechanical properties of the cells and the extracellular matrix, and that can be subsumed as "mechano-signaling pathways", like, e.g., the YAP/TAZ pathway, or the SRF pathway. Stiffness of the liver tissue modulates mechanical regulation of many genes involved in HCC progression. However, mechano-signaling is still rather underrepresented in our concepts of cancer in comparison to "classical" biochemical signalling pathways. This review aims to give an overview of various stiffness induced mechano-biological aspects of HCC.
Collapse
Affiliation(s)
- Mehak Passi
- Center for Drug Research, Ludwig-Maximilians-University, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Stefan Zahler
- Center for Drug Research, Ludwig-Maximilians-University, Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
13
|
Senescence in HBV-, HCV- and NAFLD- Mediated Hepatocellular Carcinoma and Senotherapeutics: Current Evidence and Future Perspective. Cancers (Basel) 2021; 13:cancers13184732. [PMID: 34572959 PMCID: PMC8468315 DOI: 10.3390/cancers13184732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023] Open
Abstract
Cell senescence constitutes a physiological process that serves as protection from malignant transformation of cells. However, recent scientific discoveries also identify cell senescence as pivotal in hepatocellular cancer (HCC) biology. The review herein aimed to accumulate evidence on senescence as a mediator of HCC occurrence in hepatitis B (HBV), C (HCV) virus infections, and non-alcoholic fatty liver disease (NAFLD). In HBV infection, the carcinogenic HBV X protein frequently mutates during chronic infection, and subsequently exhibits different effects on senescence. In HCV infection, senescent non-functional T-cells do not effectively clear pre-malignant hepatocytes. Furthermore, the HCV Core protein inhibits the occurrence of normal stress-induced hepatocyte senescence, allowing damaged cells to maintain their proliferative potential. In NAFLD-mediated HCC, current data point towards the gut microbiome and hepatic stellate cell senescence. Additionally, senescence contributes in the development of resistance in targeted therapies, such as sorafenib. Finally, the promising role of senotherapeutics in HCC was also explored. Overall, although we may still be at a primitive stage in fully unraveling the role of senescence in cancer, it seems that understanding and harnessing senescence may have the potential to revolutionize the way we treat hepatocellular cancer.
Collapse
|
14
|
Zhang Z, Chen B, Zhu Y, Zhang T, Yuan Y, Zhang X, Xu Y. The Jumonji Domain-Containing Histone Demethylase Homolog 1D/lysine Demethylase 7A (JHDM1D/KDM7A) Is an Epigenetic Activator of RHOJ Transcription in Breast Cancer Cells. Front Cell Dev Biol 2021; 9:664375. [PMID: 34249916 PMCID: PMC8262595 DOI: 10.3389/fcell.2021.664375] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
The small GTPase RHOJ is a key regulator of breast cancer metastasis by promoting cell migration and invasion. The prometastatic stimulus TGF-β activates RHOJ transcription via megakaryocytic leukemia 1 (MKL1). The underlying epigenetic mechanism is not clear. Here, we report that MKL1 deficiency led to disrupted assembly of the RNA polymerase II preinitiation complex on the RHOJ promoter in breast cancer cells. This could be partially explained by histone H3K9/H3K27 methylation status. Further analysis confirmed that the H3K9/H3K27 dual demethylase JHDM1D/KDM7A was essential for TGF-β-induced RHOJ transcription in breast cancer cells. MKL1 interacted with and recruited KDM7A to the RHOJ promoter to cooperatively activate RHOJ transcription. KDM7A knockdown attenuated migration and invasion of breast cancer cells in vitro and mitigated the growth and metastasis of breast cancer cells in nude mice. KDM7A expression level, either singularly or in combination with that of RHOJ, could be used to predict prognosis in breast cancer patients. Of interest, KDM7A appeared to be a direct transcriptional target of TGF-β signaling. A SMAD2/SMAD4 complex bound to the KDM7A promoter and mediated TGF-β-induced KDM7A transcription. In conclusion, our data unveil a novel epigenetic mechanism whereby TGF-β regulates the transcription of the prometastatic small GTPase RHOJ. Screening for small-molecule inhibitors of KDM7A may yield effective therapeutic solutions to treat malignant breast cancers.
Collapse
Affiliation(s)
- Ziyu Zhang
- Key Laboratory of Women's Reproductive Health of Jiangxi, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China.,Central Laboratory, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Baoyu Chen
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Tianyi Zhang
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yibiao Yuan
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xiaoling Zhang
- School of Medicine, Nanchang University, Nanchang, China.,Department of Gynecology, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Yong Xu
- Key Laboratory of Targeted Invention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
15
|
Gutsch D, Jenke R, Büch T, Aigner A. Inhibition of HER Receptors Reveals Distinct Mechanisms of Compensatory Upregulation of Other HER Family Members: Basis for Acquired Resistance and for Combination Therapy. Cells 2021; 10:272. [PMID: 33572976 PMCID: PMC7911202 DOI: 10.3390/cells10020272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/02/2022] Open
Abstract
Overexpression of members of the HER/erbB transmembrane tyrosine kinase family like HER2/erbB2/neu is associated with various cancers. Some heterodimers, especially HER2/HER3 heterodimers, are particularly potent inducers of oncogenic signaling. Still, from a clinical viewpoint their inhibition has yielded only moderate success so far, despite promising data from cell cultures. This suggests acquired resistance upon inhibitor therapy as one putative issue, requiring further studies in cell culture also aiming at rational combination therapies. In this paper, we demonstrate in ovarian carcinoma cells that the RNAi-mediated single knockdown of HER2 or HER3 leads to the rapid counter-upregulation of the respective other HER family member, thus providing a rational basis for combinatorial inhibition. Concomitantly, combined knockdown of HER2/HER3 exerts stronger anti-tumor effects as compared to single inhibition. In a tumor cell line xenograft mouse model, therapeutic intervention with nanoscale complexes based on polyethylenimine (PEI) for siRNA delivery, again reveals HER3 upregulation upon HER2 single knockdown and a therapeutic benefit from combination therapy. On the mechanistic side, we demonstrate that HER2 knockdown or inhibition reduces miR-143 levels with subsequent de-repression of HER3 expression, and validates HER3 as a direct target of miR-143. HER3 knockdown or inhibition, in turn, increases HER2 expression through the upregulation of the transcriptional regulator SATB1. These counter-upregulation processes of HER family members are thus based on distinct molecular mechanisms and may provide the basis for the rational combination of inhibitors.
Collapse
Affiliation(s)
- Daniela Gutsch
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, D-04107 Leipzig, Germany; (D.G.); (R.J.); (T.B.)
| | - Robert Jenke
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, D-04107 Leipzig, Germany; (D.G.); (R.J.); (T.B.)
- University Cancer Center Leipzig (UCCL), University Hospital Leipzig, D-04103 Leipzig, Germany
| | - Thomas Büch
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, D-04107 Leipzig, Germany; (D.G.); (R.J.); (T.B.)
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, University of Leipzig, D-04107 Leipzig, Germany; (D.G.); (R.J.); (T.B.)
| |
Collapse
|
16
|
Molecular Mechanisms to Target Cellular Senescence in Hepatocellular Carcinoma. Cells 2020; 9:cells9122540. [PMID: 33255630 PMCID: PMC7761055 DOI: 10.3390/cells9122540] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has emerged as a major cause of cancer-related death and is the most common type of liver cancer. Due to the current paucity of drugs for HCC therapy there is a pressing need to develop new therapeutic concepts. In recent years, the role of Serum Response Factor (SRF) and its coactivators, Myocardin-Related Transcription Factors A and B (MRTF-A and -B), in HCC formation and progression has received considerable attention. Targeting MRTFs results in HCC growth arrest provoked by oncogene-induced senescence. The induction of senescence acts as a tumor-suppressive mechanism and therefore gains consideration for pharmacological interventions in cancer therapy. In this article, we describe the key features and the functional role of senescence in light of the development of novel drug targets for HCC therapy with a focus on MRTFs.
Collapse
|
17
|
Kunz M, Brandl M, Bhattacharya A, Nobereit-Siegel L, Ewe A, Weirauch U, Hering D, Reinert A, Kalwa H, Guzman J, Weigelt K, Wach S, Taubert H, Aigner A. Nanoparticle-complexed antimiRs for inhibiting tumor growth and metastasis in prostate carcinoma and melanoma. J Nanobiotechnology 2020; 18:173. [PMID: 33228711 PMCID: PMC7685669 DOI: 10.1186/s12951-020-00728-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background MiRNAs act as negative regulators of gene expression through target mRNA degradation or inhibition of its translation. In cancer, several miRNAs are upregulated and play crucial roles in tumorigenesis, making the inhibition of these oncomiRs an interesting therapeutic approach. This can be achieved by directly complementary single-stranded anti-miRNA oligonucleotides (antimiRs). A major bottleneck in antimiR therapy, however, is their efficient delivery. The nanoparticle formation with polyethylenimine (PEI) may be particularly promising, based on the PEI’s ability to electrostatically interact with oligonucleotides. This leads to their protection and supports delivery. In the present study, we explore for the first time PEI for antimiR formulation and delivery. We use the branched low molecular weight PEI F25-LMW for the complexation of different antimiRs, and analyse tumor- and metastasis-inhibitory effects of PEI/antimiR complexes in different tumor models. Results In prostate carcinoma, transfection of antimiRs against miR-375 and miR-141 leads to tumor cell inhibition in 2D- and 3D-models. More importantly, an in vivo tumor therapy study in prostate carcinoma xenografts reveals anti-tumor effects of the PEI/antimiR complexes. In advanced melanoma and metastasis, we identify by a microRNA screen miR-150 as a particularly relevant oncomiR candidate, and validate this result in vitro and in vivo. Again, the systemic application of PEI/antimiR complexes inhibiting this miRNA, or the previously described antimiR-638, leads to profound tumor growth inhibition. These effects are associated with the upregulation of direct miRNA target genes. In a melanoma metastasis mouse model, anti-metastatic effects of PEI/antimiR treatment are observed as well. Conclusions We thus describe PEI-based complexes as efficient platform for antimiR therapy, as determined in two different tumor entities using in vivo models of tumor growth or metastasis. Our study also highlights the therapeutic relevance of miR-375, miR-141, miR-150 and miR-638 as target miRNAs for antimiR-mediated inhibition.![]()
Collapse
Affiliation(s)
- Manfred Kunz
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - Madeleine Brandl
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Haertelstrasse 16-18, 04107, Leipzig, Germany
| | - Animesh Bhattacharya
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, Leipzig, Germany.,Department of Hematology, Oncology and Tumor Immunology, Charité-University Medical Center, Virchow Campus, Berlin, Germany
| | - Lars Nobereit-Siegel
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, Leipzig, Germany.,Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Haertelstrasse 16-18, 04107, Leipzig, Germany
| | - Alexander Ewe
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Haertelstrasse 16-18, 04107, Leipzig, Germany
| | - Ulrike Weirauch
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Haertelstrasse 16-18, 04107, Leipzig, Germany
| | - Doreen Hering
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, Leipzig, Germany
| | - Anja Reinert
- Faculty of Veterinary Medicine, Institute of Anatomy, Histology and Embryology, Leipzig University, Leipzig, Germany
| | - Hermann Kalwa
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, University of Leipzig, Leipzig, Germany
| | - Juan Guzman
- Department of Urology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Katrin Weigelt
- Department of Urology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Sven Wach
- Department of Urology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Helge Taubert
- Department of Urology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Haertelstrasse 16-18, 04107, Leipzig, Germany.
| |
Collapse
|
18
|
Chen B, Yuan Y, Sun L, Chen J, Yang M, Yin Y, Xu Y. MKL1 Mediates TGF-β Induced RhoJ Transcription to Promote Breast Cancer Cell Migration and Invasion. Front Cell Dev Biol 2020; 8:832. [PMID: 32984327 PMCID: PMC7478007 DOI: 10.3389/fcell.2020.00832] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022] Open
Abstract
Differential regulation of gene transcription contributes to cancer metastasis. We investigated the involvement of a Rho GTPase (RhoJ) in breast cancer metastasis focusing on the mechanism underlying RhoJ trans-activation by pro-metastatic cues. We report that expression of RhoJ was up-regulated in malignant breast cancer cells compared to more benign ones. Higher RhoJ expression was also detected in human breast cancer biopsy specimens of advanced stages. RhoJ depletion attenuated breast cancer cell migration and invasion in vitro and metastasis in vivo. The pro-metastatic stimulus TGF-β activated RhoJ via megakaryocytic leukemia 1 (MKL1). MKL1 interacted with and was recruited by ETS-related gene 1 (ERG1) to the RhoJ promoter to activate transcription. In conclusion, our data delineate a novel transcriptional pathway that contributes to breast cancer metastasis. Targeting the ERG1-MKL1-RhoJ axis may be considered as a reasonable approach to treat malignant breast cancer.
Collapse
Affiliation(s)
- Baoyu Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysioloy and Laboratory Center for Experimental Medicine, Nanjing Medical University, Nanjing, China
| | - Yibiao Yuan
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysioloy and Laboratory Center for Experimental Medicine, Nanjing Medical University, Nanjing, China
| | - Lina Sun
- Department of Pathology and Pathophysiology, College of Life and Basic Medical Sciences, Soochow University, Suzhou, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| | - Junliang Chen
- Department of Pathophysiology, Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Mengzhu Yang
- Department of Oncology, First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysioloy and Laboratory Center for Experimental Medicine, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
19
|
Tong Z, Cui Q, Wang J, Zhou Y. TransmiR v2.0: an updated transcription factor-microRNA regulation database. Nucleic Acids Res 2020; 47:D253-D258. [PMID: 30371815 PMCID: PMC6323981 DOI: 10.1093/nar/gky1023] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are important post-transcriptional regulators of gene expression and play vital roles in various biological processes. It has been reported that aberrant regulation of miRNAs was associated with the development and progression of various diseases, but the underlying mechanisms are not fully deciphered. Here, we described our updated TransmiR v2.0 database for more comprehensive information about transcription factor (TF)-miRNA regulations. 3730 TF–miRNA regulations among 19 species from 1349 reports were manually curated by surveying >8000 publications, and more than 1.7 million tissue-specific TF–miRNA regulations were further incorporated based on ChIP-seq data. Besides, we constructed a ‘Predict’ module to query the predicted TF–miRNA regulations in human based on binding motifs of TFs. To facilitate the community, we provided a ‘Network’ module to visualize TF–miRNA regulations for each TF and miRNA, or for a specific disease. An ‘Enrichment analysis’ module was also included to predict TFs that are likely to regulate a miRNA list of interest. In conclusion, with improved data coverage and webserver functionalities, TransmiR v2.0 would be a useful resource for investigating the regulation of miRNAs. TransmiR v2.0 is freely accessible at http://www.cuilab.cn/transmir.
Collapse
Affiliation(s)
- Zhan Tong
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Juan Wang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yuan Zhou
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
20
|
Friedrich M, Wiedemann K, Reiche K, Puppel SH, Pfeifer G, Zipfel I, Binder S, Köhl U, Müller GA, Engeland K, Aigner A, Füssel S, Fröhner M, Peitzsch C, Dubrovska A, Rade M, Christ S, Schreiber S, Hackermüller J, Lehmann J, Toma MI, Muders MH, Sommer U, Baretton GB, Wirth M, Horn F. The Role of lncRNAs TAPIR-1 and -2 as Diagnostic Markers and Potential Therapeutic Targets in Prostate Cancer. Cancers (Basel) 2020; 12:E1122. [PMID: 32365858 PMCID: PMC7280983 DOI: 10.3390/cancers12051122] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 01/17/2023] Open
Abstract
In search of new biomarkers suitable for the diagnosis and treatment of prostate cancer, genome-wide transcriptome sequencing was carried out with tissue specimens from 40 prostate cancer (PCa) and 8 benign prostate hyperplasia patients. We identified two intergenic long non-coding transcripts, located in close genomic proximity, which are highly expressed in PCa. Microarray studies on a larger cohort comprising 155 patients showed a profound diagnostic potential of these transcripts (AUC~0.94), which we designated as tumor associated prostate cancer increased lncRNA (TAPIR-1 and -2). To test their therapeutic potential, knockdown experiments with siRNA were carried out. The knockdown caused an increase in the p53/TP53 tumor suppressor protein level followed by downregulation of a large number of cell cycle- and DNA-damage repair key regulators. Furthermore, in radiation therapy resistant tumor cells, the knockdown leads to a renewed sensitization of these cells to radiation treatment. Accordingly, in a preclinical PCa xenograft model in mice, the systemic application of nanoparticles loaded with siRNA targeting TAPIR-1 significantly reduced tumor growth. These findings point to a crucial role of TAPIR-1 and -2 in PCa.
Collapse
Affiliation(s)
- Maik Friedrich
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Karolin Wiedemann
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Kristin Reiche
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Sven-Holger Puppel
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Gabriele Pfeifer
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
| | - Ivonne Zipfel
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
| | - Stefanie Binder
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
| | - Ulrike Köhl
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Gerd A. Müller
- Molecular Oncology, Medical School University of Leipzig, Semmelweisstr. 14, D-04103 Leipzig, Germany; (G.A.M.); (K.E.)
- Department of Chemistry and Biochemistry, University of California at Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Kurt Engeland
- Molecular Oncology, Medical School University of Leipzig, Semmelweisstr. 14, D-04103 Leipzig, Germany; (G.A.M.); (K.E.)
| | - Achim Aigner
- Clinical Pharmacology, Rudolf-Boehm-Institute for Pharmacology and Toxicology, Faculty of Medicine, Leipzig University, Härtelstr. 16–18, D-04107 Leipzig, Germany;
| | - Susanne Füssel
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany; (S.F.); (M.F.); (M.W.)
| | - Michael Fröhner
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany; (S.F.); (M.F.); (M.W.)
- Zeisigwaldklinik BETHANIEN, Zeisigwaldstraße 101, D-09130 Chemnitz, Germany
| | - Claudia Peitzsch
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, D-01307 Dresden, Germany;
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
| | - Anna Dubrovska
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, D-01307 Dresden, Germany;
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiooncology—OncoRay, D-01328 Dresden, Germany
| | - Michael Rade
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Sabina Christ
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| | - Stephan Schreiber
- Helmholtz Centre for Environmental Research—UFZ, Young Investigators Group Bioinformatics & Transcriptomics, Permoserstr. 15, D-04318 Leipzig, Germany; (S.S.); (J.H.)
| | - Jörg Hackermüller
- Helmholtz Centre for Environmental Research—UFZ, Young Investigators Group Bioinformatics & Transcriptomics, Permoserstr. 15, D-04318 Leipzig, Germany; (S.S.); (J.H.)
| | - Jörg Lehmann
- Department of Therapy Validation, Fraunhofer Institute for Cell Therapy and Immunology, GLP Test Facility, Perlickstr. 1, D-04103 Leipzig, Germany;
| | - Marieta I. Toma
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (M.I.T.); (M.H.M.); (U.S.); (G.B.B.)
- Institute of Pathology, Universitätsklinikum Bonn, Venusberg-Campus 1, D-53127 Bonn, Germany
| | - Michael H. Muders
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (M.I.T.); (M.H.M.); (U.S.); (G.B.B.)
- Rudolf-Becker-Laboratory for Prostate Cancer Research, Institute of Pathology, Universitätsklinikum Bonn, Venusberg-Campus 1, D-53127 Bonn, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (M.I.T.); (M.H.M.); (U.S.); (G.B.B.)
| | - Gustavo B. Baretton
- Institute of Pathology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (M.I.T.); (M.H.M.); (U.S.); (G.B.B.)
| | - Manfred Wirth
- Department of Urology, University Hospital and Faculty of Medicine, Technische Universität Dresden, Fetscherstr. 74, D-01307 Dresden, Germany; (S.F.); (M.F.); (M.W.)
| | - Friedemann Horn
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Johannisallee 30, D-04103 Leipzig, Germany; (K.W.); (K.R.); (G.P.); (I.Z.); (S.B.); (U.K.); (F.H.)
- Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, RIBOLUTION Biomarker Center Perlickstr. 1, D-04103 Leipzig, Germany; (S.-H.P.); (M.R.); (S.C.)
| |
Collapse
|
21
|
Wu W, Shang Y, Dai S, Yu C, Wang J. Downregulation of miR‑142‑5p inhibits human aortic smooth muscle cell proliferation and migration by targeting MKL2. Mol Med Rep 2020; 22:277-285. [PMID: 32626937 PMCID: PMC7248461 DOI: 10.3892/mmr.2020.11093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 06/26/2019] [Indexed: 12/15/2022] Open
Abstract
The increased proliferation and migration of vascular smooth muscle cells (VSMCs) are critical in the progression of atherosclerosis (AS). Platelet‑derived growth factor type BB (PDGF‑BB) may induce VSMC proliferation and migration. miR‑142‑5p plays a critical role in various biological processes, including tumorigenesis, angiogenesis and inflammation. However, whether miR‑142‑5p is involved in regulating the pathological process of arteriosclerosis remains to be elucidated. Therefore, in this study, the role of miR‑142‑5p in PDGF‑BB‑induced human aortic smooth muscle cell (HSAMC) proliferation and migration was investigated. The results revealed that the expression level of miR‑142‑5p was enhanced in the serum of patients with AS, while that of its target gene, myocardin‑like protein 2 (MKL2) was decreased, compared with that in healthy volunteers. Moreover, there was a negative correlation between miR‑142‑5p and MKL2 expression in the serum of patients with AS. Furthermore, the downregulation of miR‑142‑5p inhibited PDGF‑BB‑induced HASMC proliferation and migration; however, the inhibition of HASMC proliferation and migration was reversed by co‑transfection with small interfering RNA (siRNA) against MKL2 (siRNA‑MKL2). In addition, transfection with miR‑142‑5p inhibitor significantly increased the expression levels of MKL2, and decreased those of matrix metalloproteinase (MMP)2 and 9, and these effects were reversed by transfection with siRNA‑MKL2. Finally, MKL2 was proven to be a target of miR‑142‑5p. On the whole, the findings of the present study demonstrate that the downregulation of miR‑142‑5p inhibits human aortic smooth muscle cell (HSAMC) proliferation and migration possibly by targeting MKL2. Hence, miR‑142‑5p may prove to be a novel therapeutic target in the treatment of AS.
Collapse
Affiliation(s)
- Wei Wu
- Department of Cardiothoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yuqiang Shang
- Department of Cardiothoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Shiling Dai
- Department of Cardiothoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Chunjun Yu
- Department of Cardiothoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jie Wang
- Department of Cardiothoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
22
|
Inhibition of TRPM7 blocks MRTF/SRF-dependent transcriptional and tumorigenic activity. Oncogene 2019; 39:2328-2344. [PMID: 31844251 DOI: 10.1038/s41388-019-1140-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/24/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022]
Abstract
Myocardin-related transcription factors A and B (MRTFs) are coactivators of Serum Response Factor (SRF) that mediates the expression of genes involved in cell proliferation, migration and differentiation. There is mounting evidence that MRTFs and SRF represent promising targets for hepatocellular carcinoma (HCC) growth. Since MRTF-A nuclear localization is a prerequisite for its transcriptional activity and oncogenic properties, we searched for pharmacologically active compounds able to redistribute MRTF-A to the cytoplasm. We identified NS8593, a negative gating modulator of the transient receptor potential cation channel TRPM7, as a novel inhibitor of MRTF-A nuclear localization and transcriptional activity. Using a pharmacological approach and targeted genome editing, we investigated the functional contribution of TRPM7, a unique ion channel containing a serine-threonine kinase domain, to MRTF transcriptional and tumorigenic activity. We found that TRPM7 function regulates RhoA activity and subsequently actin polymerization, MRTF-A-Filamin A complex formation and MRTF-A/SRF target gene expression. Mechanistically, TRPM7 signaling relies on TRPM7 channel-mediated Mg2+ influx and phosphorylation of RhoA by TRPM7 kinase. Pharmacological blockade of TRPM7 results in oncogene-induced senescence of hepatocellular carcinoma (HCC) cells in vitro and in vivo in HCC xenografts. Hence, inhibition of the TRPM7/MRTF axis emerges as a promising strategy to curb HCC growth.
Collapse
|
23
|
Zhang Y, Li G. A tumor suppressor DLC1: The functions and signal pathways. J Cell Physiol 2019; 235:4999-5007. [DOI: 10.1002/jcp.29402] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/27/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Yang Zhang
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life SciencesShandong Normal UniversityJinan China
| | - Guorong Li
- Shandong Provincial Key Laboratory of Animal Resistant, School of Life SciencesShandong Normal UniversityJinan China
| |
Collapse
|
24
|
Myoferlin, a Membrane Protein with Emerging Oncogenic Roles. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7365913. [PMID: 31828126 PMCID: PMC6885792 DOI: 10.1155/2019/7365913] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/02/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022]
Abstract
Myoferlin (MYOF), initially identified in muscle cells, is a member of the Ferlin family involved in membrane fusion, membrane repair, and membrane trafficking. Dysfunction of this protein is associated with muscular dysfunction. Recently, a growing body of studies have identified MYOF as an oncogenic protein. It is overexpressed in a variety of human cancers and promotes tumorigenesis, tumor cell motility, proliferation, migration, epithelial to mesenchymal transition, angiogenesis as well as metastasis. Clinically, MYOF overexpression is associated with poor outcome in various cancers. It can serve as a prognostic marker of human malignant disease. MYOF drives the progression of cancer in various processes, including surface receptor transportation, endocytosis, exocytosis, intercellular communication, fit mitochondrial structure maintenance and cell metabolism. Depletion of MYOF demonstrates significant antitumor effects both in vitro and in vivo, suggesting that targeting MYOF may produce promising clinical benefits in the treatment of malignant disease. In the present article, we reviewed the physiological function of MYOF as well as its role in cancer, thus providing a general understanding for further exploration of this protein.
Collapse
|
25
|
MRTFB suppresses colorectal cancer development through regulating SPDL1 and MCAM. Proc Natl Acad Sci U S A 2019; 116:23625-23635. [PMID: 31690663 DOI: 10.1073/pnas.1910413116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Myocardin-related transcription factor B (MRTFB) is a candidate tumor-suppressor gene identified in transposon mutagenesis screens of the intestine, liver, and pancreas. Using a combination of cell-based assays, in vivo tumor xenograft assays, and Mrtfb knockout mice, we demonstrate here that MRTFB is a human and mouse colorectal cancer (CRC) tumor suppressor that functions in part by inhibiting cell invasion and migration. To identify possible MRTFB transcriptional targets, we performed whole transcriptome RNA sequencing in MRTFB siRNA knockdown primary human colon cells and identified 15 differentially expressed genes. Among the top candidate tumor-suppressor targets were melanoma cell adhesion molecule (MCAM), a known tumor suppressor, and spindle apparatus coiled-coil protein 1 (SPDL1), which has no confirmed role in cancer. To determine whether these genes play a role in CRC, we knocked down the expression of MCAM and SPDL1 in human CRC cells and showed significantly increased invasion and migration of tumor cells. We also showed that Spdl1 expression is significantly down-regulated in Mrtfb knockout mouse intestine, while lower SPDL1 expression levels are significantly associated with reduced survival in CRC patients. Finally, we show that depletion of MCAM and SPDL1 in human CRC cells significantly increases tumor development in xenograft assays, further confirming their tumor-suppressive roles in CRC. Collectively, our findings demonstrate the tumor-suppressive role of MRTFB in CRC and identify several genes, including 2 tumor suppressors, that act downstream of MRTFB to regulate tumor growth and survival in CRC patients.
Collapse
|
26
|
Li JP, Liao XH, Xiang Y, Yao A, Fan LJ, Li H, Zhang ZJ, Huang F, Dai ZT, Zhang TC. MKL1/miR34a/FOXP3 axis regulates cell proliferation in gastric cancer. J Cell Biochem 2019; 120:7814-7824. [PMID: 30426547 DOI: 10.1002/jcb.28056] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Megakaryoblastic leukemia 1 (MKL1) was closely related to the pathogenesis of various human malignant cancers. MiR34a was reported to be closely related to cancer cell proliferation. Forkhead box protein 3 (FOXP3) was a transcription factor that played a different role in different cancer types. CDK6 was involved in cell cycle progression and was upregulated in several types of cancers. The present study investigated the effects of MKL1/miR34a/FOXP3 axis on cell proliferation in MGC803 gastric cancer cells. Our results demonstrated that overexpression of MKL1 promoted proliferation of MGC80-3 cells, MKL1 directly binding to the promoter of CDK6 to increase its expression. Knockdown of FOXP3 promoted proliferation of MGC80-3 cells and MKL1 inhibited the expression of FOXP3 via miR-34a. The finding can contribute to elucidating the regulatory mechanism involved in the cell cycle progression of gastric cancer cells and may aid in screening potential gene targets for the biological therapy of gastric cancer.
Collapse
Affiliation(s)
- Jia-Peng Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Yuan Xiang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Ao Yao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Li-Juan Fan
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Hui Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Zi-Jian Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Feng Huang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Zhou-Tong Dai
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Tong-Cun Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China.,Key Laboratory of Industrial Fermentation Microbiology, Minwastry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
27
|
Therapeutic Targeting of Stat3 Using Lipopolyplex Nanoparticle-Formulated siRNA in a Syngeneic Orthotopic Mouse Glioma Model. Cancers (Basel) 2019; 11:cancers11030333. [PMID: 30857197 PMCID: PMC6468565 DOI: 10.3390/cancers11030333] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/25/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM), WHO grade IV, is the most aggressive primary brain tumor in adults. The median survival time using standard therapy is only 12–15 months with a 5-year survival rate of around 5%. Thus, new and effective treatment modalities are of significant importance. Signal transducer and activator of transcription 3 (Stat3) is a key signaling protein driving major hallmarks of cancer and represents a promising target for the development of targeted glioblastoma therapies. Here we present data showing that the therapeutic application of siRNAs, formulated in nanoscale lipopolyplexes (LPP) based on polyethylenimine (PEI) and the phospholipid 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), represents a promising new approach to target Stat3 in glioma. We demonstrate that the LPP-mediated delivery of siRNA mediates efficient knockdown of Stat3, suppresses Stat3 activity and limits cell growth in murine (Tu2449) and human (U87, Mz18) glioma cells in vitro. In a therapeutic setting, intracranial application of the siRNA-containing LPP leads to knockdown of STAT3 target gene expression, decreased tumor growth and significantly prolonged survival in Tu2449 glioma-bearing mice compared to negative control-treated animals. This is a proof-of-concept study introducing PEI-based lipopolyplexes as an efficient strategy for therapeutically targeting oncoproteins with otherwise limited druggability.
Collapse
|
28
|
Ji S, Zheng Z, Liu S, Ren G, Gao J, Zhang Y, Li G. Resveratrol promotes oxidative stress to drive DLC1 mediated cellular senescence in cancer cells. Exp Cell Res 2018; 370:292-302. [DOI: 10.1016/j.yexcr.2018.06.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
|
29
|
Liu L, Wu X, Xu H, Yu L, Zhang X, Li L, Jin J, Zhang T, Xu Y. Myocardin-related transcription factor A (MRTF-A) contributes to acute kidney injury by regulating macrophage ROS production. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3109-3121. [PMID: 29908908 DOI: 10.1016/j.bbadis.2018.05.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/11/2018] [Accepted: 05/31/2018] [Indexed: 11/17/2022]
Abstract
A host of pathogenic factors induce acute kidney injury (AKI) leading to insufficiencies of renal function. In the present study we evaluated the role of myocardin-related transcription factor A (MRTF-A) in the pathogenesis of AKI. We report that systemic deletion of MRTF-A or inhibition of MRTF-A activity with CCG-1423 significantly attenuated AKI in mice induced by either ischemia-reperfusion or LPS injection. Of note, MRTF-A deficiency or suppression resulted in diminished renal ROS production in AKI models with down-regulation of NAPDH oxdiase 1 (NOX1) and NOX4 expression. In cultured macrophages, MRTF-A promoted NOX1 transcription in response to either hypoxia-reoxygenation or LPS treatment. Interestingly, macrophage-specific MRTF-A deletion ameliorated AKI in mice. Mechanistic analyses revealed that MRTF-A played a role in regulating histone H4K16 acetylation surrounding the NOX gene promoters by interacting with the acetyltransferase MYST1. MYST1 depletion repressed NOX transcription in macrophages. Finally, administration of a MYST1 inhibitor MG149 alleviated AKI in mice. Therefore, we data illustrate a novel epigenetic pathway that controls ROS production in macrophages contributing to AKI. Targeting the MRTF-A-MYST1-NOX axis may yield novel therapeutic strategies to combat AKI.
Collapse
Affiliation(s)
- Li Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xiaoyan Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Huihui Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Liming Yu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xinjian Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Luyang Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Jianliang Jin
- Department of Anatomy and Histology, Nanjing Medical University, Nanjing, China
| | - Tao Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Department of Renal Medicine, Jiangsu Remin Hospital affiliated to Nanjing Medical University, Nanjing, China.
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
30
|
Xiao P, Long X, Zhang L, Ye Y, Guo J, Liu P, Zhang R, Ning J, Yu W, Wei F, Yu J. Neurotensin/IL-8 pathway orchestrates local inflammatory response and tumor invasion by inducing M2 polarization of Tumor-Associated macrophages and epithelial-mesenchymal transition of hepatocellular carcinoma cells. Oncoimmunology 2018; 7:e1440166. [PMID: 29900041 DOI: 10.1080/2162402x.2018.1440166] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 01/31/2018] [Accepted: 02/06/2018] [Indexed: 12/12/2022] Open
Abstract
We previously demonstrated that neurotensin (NTS) induces local inflammation and promotes tumor invasion in hepatocellular carcinoma (HCC). However, the underlying molecular mechanisms are not clear. In this study, positive correlations between NTS and interleukin (IL)-8 were identified at both the mRNA and protein levels in 71 fresh HCC tissues and 100 paraffin-embedded HCC tissues. Furthermore, significant correlations were determined among the co-expression of NTS and IL-8, infiltration of inflammatory cells and enhanced epithelial-mesenchymal transition (EMT) of HCC cells. NTS-induced IL-8 production was associated with activation of the mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways rather than the protein kinase C (PKC) and phosphoinositide-3 kinase (PI3K) pathways, whose specific antagonists significantly inhibited activation of the NTS/IL-8 pathway. IL-8, which promoted EMT and HCC invasion both in vitro and in vivo, was produced by NTS-induced HCC cells and was effectively attenuated by blocking IL-8 receptors in vitro. Moreover, HCC-derived IL-8 attracted more CD68+ tumor-associated macrophages (TAMs) and CD66b+ polymorphonuclear neutrophils (PMNs) to the local microenvironment, displaying enhanced cytokine secretion and phagocytosis. IL-8 stimulated the M2 polarization of TAMs, which promoted the EMT and invasive potential of HCC cells. Blockage of the IL-8 receptor, NTR1 receptor or both significantly reduced HCC metastases in tumor-bearing mouse models via inhibiting EMT. In summary, aberrant activation of the NTS/IL-8 pathway in HCC dramatically stimulated the invasive potential of HCC cells. HCC-derived IL-8 promoted a pro-oncogenic inflammatory microenvironment by inducing M2-type TAMs and indirectly promoting EMT, which might be a valuable therapeutic target to prevent tumor progression.
Collapse
Affiliation(s)
- Pei Xiao
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Xinxin Long
- Department of Oncology, Tengzhou Central People's Hospital, Tengzhou, Shandong, P.R. China
| | - Lijie Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China
| | - Jincheng Guo
- Bioinformatics Research Group & Health Big-Data, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Pengpeng Liu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China
| | - Rui Zhang
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China
| | - Junya Ning
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Tianjin, P. R. China.,Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| |
Collapse
|
31
|
Seifert A, Posern G. Tightly controlled MRTF-A activity regulates epithelial differentiation during formation of mammary acini. Breast Cancer Res 2017; 19:68. [PMID: 28592291 PMCID: PMC5463372 DOI: 10.1186/s13058-017-0860-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/25/2017] [Indexed: 01/06/2023] Open
Abstract
Background Myocardin-related transcription factors (MRTF) A and B link actin dynamics and mechanotransduction to gene expression. In mice, MRTF-A is involved in mammary gland differentiation, but its role in human mammary epithelial cells remains unclear. Methods Three-dimensional cultures of human mammary epithelial MCF10A cells were used to model acinar morphogenesis. Stable MRTF-A knockdown, MRTF-A/B rescue and MRTF-A/B overexpression was established to characterize the functional role during morphogenesis using confocal microscopy and expression analysis. Breast cancer patient databases were analyzed for MRTF-A expression. Results We showed that a precise temporal control of MRTFs is required for normal morphogenesis of MCF10A mammary acini. MRTF transcriptional activity, but not their protein amounts, is transiently induced during 3D acini formation. MRTF-A knockdown dramatically reduces acini size and prevents lumen formation. These effects are rescued by re-expression of MRTF-A, and partially by MRTF-B. Conversely, overexpression of MRTF-A and MRTF-B increases acini size, resulting in irregular spheroids without lumen and defective apico-basal polarity. These phenotypes correlate with deregulated expression of cell cycle inhibitors p21/Waf1, p27/Kip1 and altered phosphorylation of retinoblastoma protein. In MRTF overexpressing spheroids, proliferation and apoptosis are simultaneously increased at late stages, whilst neither occurs in control acini. MRTFs interfere with anoikis of the inner cells and cause an integrin switch from α6 to α5, repression of E-cadherin and induction of mesenchymal markers vimentin, Snai2 and Zeb1. Moreover, MRTF-overexpressing spheroids are insensitive to alteration in matrix stiffness. In two breast cancer cohorts, high expression of MRTF-A and known target genes was associated with decreased patient survival. Conclusion MRTF-A is required for proliferation and formation of mammary acini from luminal epithelial cells. Conversely, elevated MRTF activity results in pre-malignant spheroid formation due to defective proliferation, polarity loss and epithelial-mesenchymal transition. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0860-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anja Seifert
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany.
| |
Collapse
|
32
|
Hermanns C, Hampl V, Holzer K, Aigner A, Penkava J, Frank N, Martin DE, Maier KC, Waldburger N, Roessler S, Goppelt-Struebe M, Akrap I, Thavamani A, Singer S, Nordheim A, Gudermann T, Muehlich S. The novel MKL target gene myoferlin modulates expansion and senescence of hepatocellular carcinoma. Oncogene 2017; 36:3464-3476. [DOI: 10.1038/onc.2016.496] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/23/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022]
|
33
|
Muehlich S, Rehm M, Ebenau A, Goppelt-Struebe M. Synergistic induction of CTGF by cytochalasin D and TGFβ-1 in primary human renal epithelial cells: Role of transcriptional regulators MKL1, YAP/TAZ and Smad2/3. Cell Signal 2016; 29:31-40. [PMID: 27721022 DOI: 10.1016/j.cellsig.2016.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/26/2016] [Accepted: 10/06/2016] [Indexed: 02/08/2023]
Abstract
Changes in cell morphology that involve alterations of the actin cytoskeleton are a hallmark of diseased renal tubular epithelial cells. While the impact of actin remodeling on gene expression has been analyzed in many model systems based on cell lines, this study investigated human primary tubular epithelial cells isolated from healthy parts of tumor nephrectomies. Latrunculin B (LatB) and cytochalasin D (CytoD) were used to modulate G-actin levels in a receptor-independent manner. Both compounds (at 0.5μM) profoundly altered F-actin structures in a Rho kinase-dependent manner, but only CytoD strongly induced the pro-fibrotic factor CTGF (connective tissue growth factor). CTGF induction was dependent on YAP as shown by transient downregulation experiments. However, CytoD did not alter the nuclear localization of either YAP or TAZ, whereas LatB reduced nuclear levels particularly of TAZ. CytoD modified MKL1, a coactivator of serum response factor (SRF) regulating CTGF induction, and promoted its nuclear localization. TGFβ-1 is one of the major factors involved in tubulointerstitial disease and an inducer of CTGF. Preincubation with CytoD but not LatB synergistically enhanced the TGFβ-1-stimulated synthesis of CTGF, both in cells cultured on plastic dishes as well as in polarized epithelial cells. CytoD had no direct effect on the phosphorylation of Smad2/3, but facilitated their phosphorylation and thus activation by TGFβ-1. Our present findings provide evidence that morphological alterations have a strong impact on cellular signaling of one of the major pro-fibrotic factors, TGFβ-1. However, our data also indicate that changes in cell morphology per se cannot predict those interactions which are critically dependent on molecular fine tuning of actin reorganization.
Collapse
Affiliation(s)
- Susanne Muehlich
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Goethestrasse 33, D-80336 München, Germany
| | - Margot Rehm
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Loschgestrasse 8, D-91054 Erlangen, Germany
| | - Astrid Ebenau
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Loschgestrasse 8, D-91054 Erlangen, Germany
| | - Margarete Goppelt-Struebe
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Loschgestrasse 8, D-91054 Erlangen, Germany.
| |
Collapse
|
34
|
Muehlich S, Hermanns C, Meier MA, Kircher P, Gudermann T. Unravelling a new mechanism linking actin polymerization and gene transcription. Nucleus 2016; 7:121-5. [PMID: 27104924 DOI: 10.1080/19491034.2016.1171433] [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] [Indexed: 10/21/2022] Open
Abstract
In the recent years, the role of actin and actin-binding proteins in gene transcription has received considerable attention. Nuclear monomeric and polymerized actin and several actin binding proteins have been detected in the mammalian cell nucleus, although their roles in transcription are just beginning to emerge. Our group recently reported that the actin-binding protein Filamin A interacts with the transcriptional coactivator MKL1 to link actin polymerization with transcriptional activity of Serum Response Factor. Here we summarize the regulation and function of MKL1, and highlight this novel mechanism of MKL1 regulation through binding to Filamin A and its implications for cell migration.
Collapse
Affiliation(s)
- Susanne Muehlich
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Constanze Hermanns
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Melanie A Meier
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Philipp Kircher
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| | - Thomas Gudermann
- a Walther Straub Institute of Pharmacology and Toxicology , Ludwig-Maximilians-University , Munich , Germany
| |
Collapse
|
35
|
Kircher P, Hermanns C, Nossek M, Drexler MK, Grosse R, Fischer M, Sarikas A, Penkava J, Lewis T, Prywes R, Gudermann T, Muehlich S. Filamin A interacts with the coactivator MKL1 to promote the activity of the transcription factor SRF and cell migration. Sci Signal 2015; 8:ra112. [PMID: 26554816 DOI: 10.1126/scisignal.aad2959] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Megakaryoblastic leukemia 1 (MKL1) is a coactivator of serum response factor (SRF) that promotes the expression of genes associated with cell proliferation, motility, adhesion, and differentiation-processes that also involve dynamic cytoskeletal changes in the cell. MKL1 is inactive when bound to monomeric globular actin (G-actin), but signals that activate the small guanosine triphosphatase RhoA cause actin polymerization and MKL1 dissociation from G-actin. We found a new mechanism of MKL1 activation that is mediated through its binding to filamin A (FLNA), a protein that binds filamentous actin (F-actin). The interaction of FLNA and MKL1 was required for the expression of MKL1 target genes in primary fibroblasts, melanoma, mammary and hepatocellular carcinoma cells. We identified the regions of interaction between MKL1 and FLNA, and cells expressing an MKL1 mutant that was unable to bind FLNA exhibited impaired cell migration and reduced expression of MKL1-SRF target genes. Induction and repression of MKL1-SRF target genes correlated with increased or decreased MKL1-FLNA interaction, respectively. Lysophosphatidic acid-induced RhoA activation in primary human fibroblasts promoted the association of endogenous MKL1 with FLNA, whereas exposure to an actin polymerization inhibitor dissociated MKL1 from FLNA and decreased MKL1-SRF target gene expression in melanoma cells. Thus, FLNA functions as a positive cellular transducer linking actin polymerization to MKL1-SRF activity, counteracting the known repressive complex of MKL1 and monomeric G-actin.
Collapse
Affiliation(s)
- Philipp Kircher
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Constanze Hermanns
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Maximilian Nossek
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Maria Katharina Drexler
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Robert Grosse
- Institute of Pharmacology, Biochemical-Pharmacological Center, University of Marburg, Marburg 35043, Germany
| | - Maximilian Fischer
- Institute of Pharmacology and Toxicology, Technical University Munich, Munich 80802, Germany
| | - Antonio Sarikas
- Institute of Pharmacology and Toxicology, Technical University Munich, Munich 80802, Germany
| | - Josef Penkava
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany
| | - Thera Lewis
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany. Comprehensive Pneumology Center Munich, German Center for Lung Research, Munich 81377, Germany. German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich 80802, Germany
| | - Susanne Muehlich
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich 80336, Germany.
| |
Collapse
|
36
|
Davis B, Shen Y, Poon CC, Luchman HA, Stechishin OD, Pontifex CS, Wu W, Kelly JJ, Blough MD. Comparative genomic and genetic analysis of glioblastoma-derived brain tumor-initiating cells and their parent tumors. Neuro Oncol 2015; 18:350-60. [PMID: 26245525 DOI: 10.1093/neuonc/nov143] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a fatal cancer that has eluded major therapeutic advances. Failure to make progress may reflect the absence of a human GBM model that could be used to test compounds for anti-GBM activity. In this respect, the development of brain tumor-initiating cell (BTIC) cultures is a step forward because BTICs appear to capture the molecular diversity of GBM better than traditional glioma cell lines. Here, we perform a comparative genomic and genetic analysis of BTICs and their parent tumors as preliminary evaluation of the BTIC model. METHODS We assessed single nucleotide polymorphisms (SNPs), genome-wide copy number variations (CNVs), gene expression patterns, and molecular subtypes of 11 established BTIC lines and matched parent tumors. RESULTS Although CNV differences were noted, BTICs retained the major genomic alterations characteristic of GBM. SNP patterns were similar between BTICs and tumors. Importantly, recurring SNP or CNV alterations specific to BTICs were not seen. Comparative gene expression analysis and molecular subtyping revealed differences between BTICs and GBMs. These differences formed the basis of a 63-gene expression signature that distinguished cells from tumors; differentially expressed genes primarily involved metabolic processes. We also derived a set of 73 similarly expressed genes; these genes were not associated with specific biological functions. CONCLUSIONS Although not identical, established BTIC lines preserve the core molecular alterations seen in their parent tumors, as well as the genomic hallmarks of GBM, without acquiring recurring BTIC-specific changes.
Collapse
Affiliation(s)
- Brad Davis
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Yaoqing Shen
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Candice C Poon
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - H Artee Luchman
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Owen D Stechishin
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Carly S Pontifex
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Wei Wu
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - John J Kelly
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | - Michael D Blough
- Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada (B.D., Y. S.); Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., J.J.K.); Clark Smith Brain Tumour Research Centre, Southern Alberta Cancer Research Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (C.C.P., C.S.P., W.W., J.J.K., M.D.B.); Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada (H.A.L., O.D.S.)
| | | |
Collapse
|
37
|
Giehl K, Keller C, Muehlich S, Goppelt-Struebe M. Actin-mediated gene expression depends on RhoA and Rac1 signaling in proximal tubular epithelial cells. PLoS One 2015; 10:e0121589. [PMID: 25816094 PMCID: PMC4376694 DOI: 10.1371/journal.pone.0121589] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 02/14/2015] [Indexed: 12/29/2022] Open
Abstract
Morphological alterations of cells can lead to modulation of gene expression. An essential link is the MKL1-dependent activation of serum response factor (SRF), which translates changes in the ratio of G- and F-actin into mRNA transcription. SRF activation is only partially characterized in non-transformed epithelial cells. Therefore, the impact of GTPases of the Rho family and changes in F-actin structures were analyzed in renal proximal tubular epithelial cells. Activation of SRF signaling was compared to the regulation of a known MKL1/SRF target gene, connective tissue growth factor (CTGF). In the human proximal tubular cell line HKC-8 overexpression of two actin mutants either favoring or preventing the formation of F-actin fibers regulated SRF-mediated transcription as well as CTGF expression. Only overexpression of constitutively active RhoA activated SRF-dependent gene expression whereas no effect was detected upon overexpression of Rac1 mutants. To elucidate the functional role of Rho kinases as downstream mediators of RhoA, pharmacological inhibition and genetic inhibition by transient siRNA knock down were compared. Upon stimulation with lysophosphatidic acid (LPA) Rho kinase inhibitors partially suppressed SRF-mediated transcription, whereas interference with Rho kinase expression by siRNA reduced activation of SRF, but barely affected CTGF expression. Together with the partial inhibition of CTGF expression by the pharmacological inhibitors Y27432 and H1154, Rho kinases seem to be less important in mediating RhoA signaling related to CTGF expression in HKC-8 epithelial cells. Short term pharmacological inhibition of Rac1 activity by EHT1864 reduced SRF-dependent CTGF expression in HKC-8 cells, but was overcome by a stimulatory effect after prolonged incubation after 4-6 h. Similarly, human primary cells of proximal but not of distal tubular origin showed inhibitory as well as stimulatory effects of Rac1 inhibition. Thus, RhoA signaling activates MKL1-SRF-mediated CTGF expression in proximal tubular cells, whereas Rac1 signaling is more complex with adaptive cellular responses.
Collapse
Affiliation(s)
- Klaudia Giehl
- Signal Transduction of Cellular Motility, Internal Medicine V, Justus-Liebig-University Giessen, Giessen, Germany
| | - Christof Keller
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Muehlich
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Margarete Goppelt-Struebe
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
38
|
Cheng X, Yang Y, Fan Z, Yu L, Bai H, Zhou B, Wu X, Xu H, Fang M, Shen A, Chen Q, Xu Y. MKL1 potentiates lung cancer cell migration and invasion by epigenetically activating MMP9 transcription. Oncogene 2015; 34:5570-81. [PMID: 25746000 DOI: 10.1038/onc.2015.14] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/17/2014] [Accepted: 01/07/2015] [Indexed: 12/24/2022]
Abstract
Malignant tumors are exemplified by excessive proliferation and aggressive migration/invasion contributing to increased mortality of cancer patients. Matrix metalloproteinase 9 (MMP9) expression is positively correlated with lung cancer malignancy. The mechanism underlying an elevated MMP9 expression is not clearly defined. We demonstrate here that the transcriptional modulator megakaryocytic leukemia 1 (MKL1) was activated by hypoxia and transforming growth factor (TGF-β), two prominent pro-malignancy factors, in cultured lung cancer cells. MKL1 levels were also increased in more invasive types of lung cancer in humans. Depletion of MKL1 in lung cancer cells attenuated migration and invasion both in vitro and in vivo. Overexpression of MKL1 potentiated the induction of MMP9 transcription by hypoxia and TGF-β, whereas MKL1 silencing diminished MMP9 expression. Of interest, MKL1 knockdown eliminated histone H3K4 methylation surrounding the MMP9 promoter. Further analyses revealed that MKL1 recruited ASH2, a component of the H3K4 methyltransferase complex, to activate MMP9 transcription. Depletion of ASH2 ameliorated cancer cell migration and invasion in an MMP9-dependent manner. Together our data indicate that MKL1 potentiates lung cancer cell migration and invasion by epigenetically activating MMP9 transcription.
Collapse
Affiliation(s)
- X Cheng
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Y Yang
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Z Fan
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - L Yu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - H Bai
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - B Zhou
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - X Wu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - H Xu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - M Fang
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China
| | - A Shen
- Department of Key Laboratory of Inflammation and Molecular Targets, Medical College, Nantong University, Nantong, China
| | - Q Chen
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Y Xu
- Key Laboratory of Cardiovascular Disease and Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
39
|
Ohrnberger S, Thavamani A, Braeuning A, Lipka DB, Kirilov M, Geffers R, Authenrieth SE, Römer M, Zell A, Bonin M, Schwarz M, Schütz G, Schirmacher P, Plass C, Longerich T, Nordheim A, Nordheim A. Dysregulated serum response factor triggers formation of hepatocellular carcinoma. Hepatology 2015; 61:979-89. [PMID: 25266280 PMCID: PMC4365683 DOI: 10.1002/hep.27539] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/25/2014] [Indexed: 12/19/2022]
Abstract
UNLABELLED The ubiquitously expressed transcriptional regulator serum response factor (SRF) is controlled by both Ras/MAPK (mitogen-activated protein kinase) and Rho/actin signaling pathways, which are frequently activated in hepatocellular carcinoma (HCC). We generated SRF-VP16iHep mice, which conditionally express constitutively active SRF-VP16 in hepatocytes, thereby controlling subsets of both Ras/MAPK- and Rho/actin-stimulated target genes. All SRF-VP16iHep mice develop hyperproliferative liver nodules that progresses to lethal HCC. Some murine (m)HCCs acquire Ctnnb1 mutations equivalent to those in human (h)HCC. The resulting transcript signatures mirror those of a distinct subgroup of hHCCs, with shared activation of oncofetal genes including Igf2, correlating with CpG hypomethylation at the imprinted Igf2/H19 locus. CONCLUSION SRF-VP16iHep mHCC reveal convergent Ras/MAPK and Rho/actin signaling as a highly oncogenic driver mechanism for hepatocarcinogenesis. This suggests simultaneous inhibition of Ras/MAPK and Rho/actin signaling as a treatment strategy in hHCC therapy.
Collapse
Affiliation(s)
- Stefan Ohrnberger
- Department for Molecular Biology, Interfaculty Institute of Cell Biology, Tuebingen UniversityGermany
| | - Abhishek Thavamani
- Department for Molecular Biology, Interfaculty Institute of Cell Biology, Tuebingen UniversityGermany,German Cancer Consortium (DKTK) and DKFZHeidelberg, Germany
| | | | - Daniel B Lipka
- German Cancer Consortium (DKTK) and DKFZHeidelberg, Germany,Division of Epigenomics and Cancer Risk Factors, DKFZHeidelberg, Germany
| | | | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection ResearchBraunschweig, Germany
| | | | | | | | - Michael Bonin
- Institute of Medical Genetics and Applied Genomics, UKTTuebingen, Germany
| | | | | | - Peter Schirmacher
- Institute of Pathology, University Hospital HeidelbergHeidelberg, Germany
| | - Christoph Plass
- German Cancer Consortium (DKTK) and DKFZHeidelberg, Germany,Division of Epigenomics and Cancer Risk Factors, DKFZHeidelberg, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital HeidelbergHeidelberg, Germany
| | - Alfred Nordheim
- Department for Molecular Biology, Interfaculty Institute of Cell Biology, Tuebingen UniversityGermany,German Cancer Consortium (DKTK) and DKFZHeidelberg, Germany
| | | |
Collapse
|
40
|
Popescu NC, Goodison S. Deleted in liver cancer-1 (DLC1): an emerging metastasis suppressor gene. Mol Diagn Ther 2015; 18:293-302. [PMID: 24519699 DOI: 10.1007/s40291-014-0086-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
While significant progress continues to be made in the early detection and therapeutic management of primary tumors, the incidence of metastatic disease remains the major cause of mortality. Accordingly, the development of novel effective therapies that can ameliorate dissemination and secondary tumor growth are a clinical priority. The identification of genetic and functional alterations in cancer cells that affect factors implicated in the metastatic process is critical for designing preventive and therapeutic strategies. Evidence implicating the protein deleted in liver cancer-1 (DLC1), a Rho GTPase activator, in metastasis has accumulated to a point where DLC1 may be considered as a metastasis suppressor gene. This review presents evidence supporting an anti-metastatic role for DLC1 in several human cancers and discusses the mechanisms contributing to its inhibitory effects. In addition, promising opportunities for therapeutic interventions based on DLC1 function and downstream pathways involved in the metastatic process are considered.
Collapse
Affiliation(s)
- Nicholas C Popescu
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, Building 37, Room 4140, 37 Convent Dr., MSC 4262, Bethesda, MD, 20892-4262, USA,
| | | |
Collapse
|
41
|
Myocardin related transcription factor A programs epigenetic activation of hepatic stellate cells. J Hepatol 2015; 62:165-74. [PMID: 25109772 DOI: 10.1016/j.jhep.2014.07.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/17/2014] [Accepted: 07/23/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Activation of hepatic stellate cells (HSCs) represents a key process in liver injury and, in the absence of intervention, leads to irreversible cirrhosis contributing significantly to the mortality of patients with liver disease. A missing link in the current understanding of HSC activation is the involvement of the epigenetic machinery. We investigated the role of the myocardin related transcription factor A (MRTF-A) in HSC activation. METHODS Liver fibrosis was induced in wild type (WT) and MRTF-A deficient (KO) mice by CCl4 injection. Expression of mRNA and protein was measured by real-time PCR, Western blotting, and immunohistochemistry. Protein binding to DNA was assayed by chromatin immunoprecipitation (ChIP). Knockdown of endogenous proteins was mediated by either small interfering RNA (siRNA) or short hairpin RNA (shRNA), carried by lentiviral particles. RESULTS KO mice exhibited resistance to CCl4-induced liver fibrosis compared to WT littermates. The expression of activated HSC signature genes was suppressed in the absence of MRTF-A. ChIP assays revealed that MRTF-A deficiency led to the erasure of key histone modifications, associated with transcriptional activation, such as H3K4 di- and tri-methylation, on the promoter regions of fibrogenic genes. Mechanistically, MRTF-A recruited a histone methyltransferase complex (COMPASS) to the promoters of fibrogenic genes to activate transcription. Silencing of individual COMPASS components dampened transactivation of fibrogenic genes in vitro and blocked liver fibrosis in mice. Oestradiol suppressed HSC activation by dampening the expression and binding activity of COMPASS. CONCLUSIONS Our data illustrate a novel mechanism that connects MRTF-A dependent histone H3K4 methylation to HSC activation.
Collapse
|
42
|
Affiliation(s)
- Susanne Muehlich
- Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Germany
| | | |
Collapse
|