1
|
Zhao Z, He K, Liu B, Nie W, Luo X, Liu J. Intrarenal pH-Responsive Self-Assembly of Luminescent Gold Nanoparticles for Diagnosis of Early Kidney Injury. Angew Chem Int Ed Engl 2024; 63:e202406016. [PMID: 38703020 DOI: 10.1002/anie.202406016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/06/2024]
Abstract
Metabolic acidosis-induced kidney injury (MAKI) is asymptomatic and lack of clinical biomarkers in early stage, but rapidly progresses to severe renal fibrosis and ultimately results in end-stage kidney failure. Therefore, developing rapid and noninvasive strategies direct responsive to renal tubular acidic microenvironment rather than delayed biomarkers are essential for timely renoprotective interventions. Herein, we develop pH-responsive luminescent gold nanoparticles (p-AuNPs) in the second near-infrared emission co-coated with 2,3-dimethylaleic anhydride conjugated β-mercaptoethylamine and cationic 2-diethylaminoethanethiol hydrochloride, which showed sensitive pH-induced charge reversal and intrarenal self-assembly for highly sensitive and long-time (~24 h) imaging of different stages of MAKI. By integrating advantages of pH-induced intrarenal self-assembly and enhanced interactions between pH-triggered positively charged p-AuNPs and renal tubular cells, the early- and late-stage MAKI could be differentiated rapidly within 10 min post-injection (p.i.) with contrast index (CI) of 3.5 and 4.3, respectively. The corresponding maximum CI could reach 5.1 and 9.2 at 12 h p.i., respectively. Furthermore, p-AuNPs were demonstrated to effectively real-time monitor progressive recovery of kidney injury in MAKI mice after therapy, and also exhibit outstanding capabilities for drug screening. This pH-responsive strategy showed great promise for feedback on kidney dysfunction progression, opening new possibilities for early-stage diagnosis of pH-related diseases.
Collapse
Affiliation(s)
- Zhipeng Zhao
- State Key Laboratory of Pulp and Paper Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Kui He
- State Key Laboratory of Pulp and Paper Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Ben Liu
- State Key Laboratory of Pulp and Paper Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Wenyan Nie
- State Key Laboratory of Pulp and Paper Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Xiaoxi Luo
- State Key Laboratory of Pulp and Paper Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| | - Jinbin Liu
- State Key Laboratory of Pulp and Paper Engineering, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, P. R. China
| |
Collapse
|
2
|
Chen Y, Li J, Ma J, Bao Y. ZNF143 facilitates the growth and migration of glioma cells by regulating KPNA2-mediated Hippo signalling. Sci Rep 2023; 13:11097. [PMID: 37423952 DOI: 10.1038/s41598-023-38158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/04/2023] [Indexed: 07/11/2023] Open
Abstract
The disordered expression of ZNF143 is closely related to the malignant progression of tumours. However, the basic control mechanism of ZNF143 in glioma has not yet been clarified. Therefore, we tried to find a new pathway to illustrate the function of ZNF143 in glioma. To explore the function of KPNA2 in the development of glioma, we used survival analysis by the Kaplan‒Meier method to assess the overall survival (OS) of patients with low and high KPNA2 expression in the TCGA and CGGA cohorts. Western blotting assays and RT‒PCR assays were utilized to determine the expression level of KPNA2 in glioma cells. The interaction between ZNF143 and KPNA2 was confirmed by ChIP assays. Proliferation was assessed by CCK-8 assays, and migration was evaluated by wound healing and Transwell assays. Apoptosis was determined by flow cytometry, and the expression level of YAP/TAZ was visualized using an immunofluorescence assay. The expression levels of LATS1, LATS2, YAP1, and p-YAP1 were determined. Patients with low KPNA2 expression showed a better prognosis than those with high KPNA2 expression. KPNA2 was found to be upregulated in human glioma cells. ZNF143 can bind to the promoter region of KPNA2. Downregulation of ZNF143 and KPNA2 can activate the Hippo signalling pathway and reduce YAP/TAZ expression in human glioma cells, thus inducing apoptosis of human glioma cells and weakening their proliferation, migration and invasion. In conclusion, ZNF143 mediates the Hippo/YAP signalling pathway and inhibits the growth and migration of glioma cells by regulating KPNA2.
Collapse
Affiliation(s)
- Yan Chen
- Department of Neurosurgery, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, People's Republic of China
| | - Jitao Li
- Department of Oncology, Shengli Oilfield Central Hospital, Dongying, 257034, People's Republic of China
| | - Jiangchun Ma
- Department of Neurosurgery, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China.
| | - Yizhong Bao
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310013, People's Republic of China.
| |
Collapse
|
3
|
Muzyka L, Goff NK, Choudhary N, Koltz MT. Systematic Review of Molecular Targeted Therapies for Adult-Type Diffuse Glioma: An Analysis of Clinical and Laboratory Studies. Int J Mol Sci 2023; 24:10456. [PMID: 37445633 DOI: 10.3390/ijms241310456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Gliomas are the most common brain tumor in adults, and molecularly targeted therapies to treat gliomas are becoming a frequent topic of investigation. The current state of molecular targeted therapy research for adult-type diffuse gliomas has yet to be characterized, particularly following the 2021 WHO guideline changes for classifying gliomas using molecular subtypes. This systematic review sought to characterize the current state of molecular target therapy research for adult-type diffuse glioma to better inform scientific progress and guide next steps in this field of study. A systematic review was conducted in accordance with PRISMA guidelines. Studies meeting inclusion criteria were queried for study design, subject (patients, human cell lines, mice, etc.), type of tumor studied, molecular target, respective molecular pathway, and details pertaining to the molecular targeted therapy-namely the modality, dose, and duration of treatment. A total of 350 studies met the inclusion criteria. A total of 52 of these were clinical studies, 190 were laboratory studies investigating existing molecular therapies, and 108 were laboratory studies investigating new molecular targets. Further, a total of 119 ongoing clinical trials are also underway, per a detailed query on clinicaltrials.gov. GBM was the predominant tumor studied in both ongoing and published clinical studies as well as in laboratory analyses. A few studies mentioned IDH-mutant astrocytomas or oligodendrogliomas. The most common molecular targets in published clinical studies and clinical trials were protein kinase pathways, followed by microenvironmental targets, immunotherapy, and cell cycle/apoptosis pathways. The most common molecular targets in laboratory studies were also protein kinase pathways; however, cell cycle/apoptosis pathways were the next most frequent target, followed by microenvironmental targets, then immunotherapy pathways, with the wnt/β-catenin pathway arising in the cohort of novel targets. In this systematic review, we examined the current evidence on molecular targeted therapy for adult-type diffuse glioma and discussed its implications for clinical practice and future research. Ultimately, published research falls broadly into three categories-clinical studies, laboratory testing of existing therapies, and laboratory identification of novel targets-and heavily centers on GBM rather than IDH-mutant astrocytoma or oligodendroglioma. Ongoing clinical trials are numerous in this area of research as well and follow a similar pattern in tumor type and targeted pathways as published clinical studies. The most common molecular targets in all study types were protein kinase pathways. Microenvironmental targets were more numerous in clinical studies, whereas cell cycle/apoptosis were more numerous in laboratory studies. Immunotherapy pathways are on the rise in all study types, and the wnt/β-catenin pathway is increasingly identified as a novel target.
Collapse
Affiliation(s)
- Logan Muzyka
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Nicolas K Goff
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Nikita Choudhary
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| | - Michael T Koltz
- Department of Neurosurgery, Dell Medical School, The University of Texas at Austin, 1501 Red River Street, Austin, TX 78712, USA
| |
Collapse
|
4
|
Wang X, Zhang Y, Wu Y, Cheng H, Wang X. The role of E3 ubiquitin ligases and deubiquitinases in bladder cancer development and immunotherapy. Front Immunol 2023; 14:1202633. [PMID: 37215134 PMCID: PMC10196180 DOI: 10.3389/fimmu.2023.1202633] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Bladder cancer is one of the common malignant urothelial tumors. Post-translational modification (PTMs), including ubiquitination, acetylation, methylation, and phosphorylation, have been revealed to participate in bladder cancer initiation and progression. Ubiquitination is the common PTM, which is conducted by E1 ubiquitin-activating enzyme, E2 ubiquitin-conjugating enzyme and E3 ubiquitin-protein ligase. E3 ubiquitin ligases play a key role in bladder oncogenesis and progression and drug resistance in bladder cancer. Therefore, in this review, we summarize current knowledge regarding the functions of E3 ubiquitin ligases in bladder cancer development. Moreover, we provide the evidence of E3 ubiquitin ligases in regulation of immunotherapy in bladder cancer. Furthermore, we mention the multiple compounds that target E3 ubiquitin ligases to improve the therapy efficacy of bladder cancer. We hope our review can stimulate researchers and clinicians to investigate whether and how targeting E3 ubiquitin ligases acts a novel strategy for bladder cancer therapy.
Collapse
|
5
|
Luo M, Xu Y, Chen H, Wu Y, Pang A, Hu J, Dong X, Che J, Yang H. Advances of targeting the YAP/TAZ-TEAD complex in the hippo pathway for the treatment of cancers. Eur J Med Chem 2022; 244:114847. [DOI: 10.1016/j.ejmech.2022.114847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/03/2022]
|
6
|
Wang X, Zhou Y, Wang Y, Wang X, Zhang Y, Mao Y, Zhang L, Qi J, Zhang Y, Lyu F, Gu L, Yu R, Zhou X. SU4312 Represses Glioma Progression by Inhibiting YAP and Inducing Sensitization to the Effect of Temozolomide. J Clin Med 2022; 11:jcm11164765. [PMID: 36013004 PMCID: PMC9410026 DOI: 10.3390/jcm11164765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/31/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
SU4312, initially designed as a multi-target tyrosine kinase inhibitor, is consequently reported to inhibit tumor angiogenesis by blocking VEGFR. However, although SU4312 can penetrate the brain–blood barrier, its potential to inhibit glioma growth is unknown. In this study, we report that SU4312 inhibited glioma cell proliferation and down-regulated yes-associated protein (YAP), the key effector of the hippo pathway. The exogenous over-expression of YAP partially restored the inhibitory effect of SU4312 on glioma progression. Interestingly, SU4312 sensitized the antitumor effect of temozolomide, both in vitro and in vivo. Moreover, SU4312 decreased the M2tumor-associated macrophages and enhanced anti-tumor immunity by down-regulating the YAP-CCL2 axis. In conclusion, our results suggest that SU4312 represses glioma progression by down-regulating YAP transcription and consequently CCL2 secretion. SU4312 may be synergistic with temozolomide for glioma treatment.
Collapse
Affiliation(s)
- Xu Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Yi Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Yan Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
| | - Xiang Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
| | - Yufei Mao
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Long Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Ji Qi
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Yining Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Feng Lyu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Linbo Gu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221004, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
- Correspondence: (R.Y.); (X.Z.)
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou 221002, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou 221002, China
- Correspondence: (R.Y.); (X.Z.)
| |
Collapse
|
7
|
Park JH, Feroze AH, Emerson SN, Mihalas AB, Keene CD, Cimino PJ, de Lomana ALG, Kannan K, Wu WJ, Turkarslan S, Baliga NS, Patel AP. A single-cell based precision medicine approach using glioblastoma patient-specific models. NPJ Precis Oncol 2022; 6:55. [PMID: 35941215 PMCID: PMC9360428 DOI: 10.1038/s41698-022-00294-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/22/2022] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma (GBM) is a heterogeneous tumor made up of cell states that evolve over time. Here, we modeled tumor evolutionary trajectories during standard-of-care treatment using multi-omic single-cell analysis of a primary tumor sample, corresponding mouse xenografts subjected to standard of care therapy, and recurrent tumor at autopsy. We mined the multi-omic data with single-cell SYstems Genetics Network AnaLysis (scSYGNAL) to identify a network of 52 regulators that mediate treatment-induced shifts in xenograft tumor-cell states that were also reflected in recurrence. By integrating scSYGNAL-derived regulatory network information with transcription factor accessibility deviations derived from single-cell ATAC-seq data, we developed consensus networks that modulate cell state transitions across subpopulations of primary and recurrent tumor cells. Finally, by matching targeted therapies to active regulatory networks underlying tumor evolutionary trajectories, we provide a framework for applying single-cell-based precision medicine approaches to an individual patient in a concurrent, adjuvant, or recurrent setting.
Collapse
Affiliation(s)
| | - Abdullah H Feroze
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Samuel N Emerson
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Anca B Mihalas
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Patrick J Cimino
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | | | - Wei-Ju Wu
- Institute for Systems Biology, Seattle, WA, USA
| | | | - Nitin S Baliga
- Institute for Systems Biology, Seattle, WA, USA.
- Departments of Microbiology, Biology, and Molecular Engineering Sciences, University of Washington, Seattle, WA, USA.
| | - Anoop P Patel
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA.
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
- Brotman-Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA.
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, USA.
| |
Collapse
|
8
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15. [DOI: https:/doi.org/10.3389/fnmol.2022.910543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
|
9
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15:910543. [PMID: 35935338 PMCID: PMC9354928 DOI: 10.3389/fnmol.2022.910543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
Affiliation(s)
- Hao Wu
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Min Wei
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Yuping Li
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Qiang Ma
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Hengzhu Zhang
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
- *Correspondence: Hengzhu Zhang,
| |
Collapse
|
10
|
Mechanical Properties of the Extracellular Environment of Human Brain Cells Drive the Effectiveness of Drugs in Fighting Central Nervous System Cancers. Brain Sci 2022; 12:brainsci12070927. [PMID: 35884733 PMCID: PMC9313046 DOI: 10.3390/brainsci12070927] [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: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
The evaluation of nanomechanical properties of tissues in health and disease is of increasing interest to scientists. It has been confirmed that these properties, determined in part by the composition of the extracellular matrix, significantly affect tissue physiology and the biological behavior of cells, mainly in terms of their adhesion, mobility, or ability to mutate. Importantly, pathophysiological changes that determine disease development within the tissue usually result in significant changes in tissue mechanics that might potentially affect the drug efficacy, which is important from the perspective of development of new therapeutics, since most of the currently used in vitro experimental models for drug testing do not account for these properties. Here, we provide a summary of the current understanding of how the mechanical properties of brain tissue change in pathological conditions, and how the activity of the therapeutic agents is linked to this mechanical state.
Collapse
|
11
|
Zhang Y, Wang X, Zhou X. Functions of Yes-association protein (YAP) in cancer progression and anticancer therapy resistance. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The Hippo pathway, a highly conserved kinase cascade, regulates cell proliferation, apoptosis, organ size, and tissue homeostasis. Dysregulation of this pathway reportedly plays an important role in the progression of various human cancers. Yes-association protein (YAP), the Hippo pathway’s core effector, is considered a marker for cancer therapy and patient prognosis. In addition, studies have indicated that YAP is involved in promoting anticancer drug resistance. This review summarizes current knowledge on YAP’s role in cancer progression, anticancer drug resistance, and advances in the development of YAP-targeting drugs. A thorough understanding of the complex interactions among molecular, cellular, and environmental factors concerning YAP function in cancer progression may provide new insight into the underlying mechanism of anticancer drug resistance. It might lead to improved prognosis through novel combined therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- These authors contributed equally to this work
| | - Xiang Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- These authors contributed equally to this work
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
12
|
Hsu SC, Lin CY, Lin YY, Collins CC, Chen CL, Kung HJ. TEAD4 as an Oncogene and a Mitochondrial Modulator. Front Cell Dev Biol 2022; 10:890419. [PMID: 35602596 PMCID: PMC9117765 DOI: 10.3389/fcell.2022.890419] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
TEAD4 (TEA Domain Transcription Factor 4) is well recognized as the DNA-anchor protein of YAP transcription complex, which is modulated by Hippo, a highly conserved pathway in Metazoa that controls organ size through regulating cell proliferation and apoptosis. To acquire full transcriptional activity, TEAD4 requires co-activator, YAP (Yes-associated protein) or its homolog TAZ (transcriptional coactivator with PDZ-binding motif) the signaling hub that relays the extracellular stimuli to the transcription of target genes. Growing evidence suggests that TEAD4 also exerts its function in a YAP-independent manner through other signal pathways. Although TEAD4 plays an essential role in determining that differentiation fate of the blastocyst, it also promotes tumorigenesis by enhancing metastasis, cancer stemness, and drug resistance. Upregulation of TEAD4 has been reported in several cancers, including colon cancer, gastric cancer, breast cancer, and prostate cancer and serves as a valuable prognostic marker. Recent studies show that TEAD4, but not other members of the TEAD family, engages in regulating mitochondrial dynamics and cell metabolism by modulating the expression of mitochondrial- and nuclear-encoded electron transport chain genes. TEAD4’s functions including oncogenic activities are tightly controlled by its subcellular localization. As a predominantly nuclear protein, its cytoplasmic translocation is triggered by several signals, such as osmotic stress, cell confluency, and arginine availability. Intriguingly, TEAD4 is also localized in mitochondria, although the translocation mechanism remains unclear. In this report, we describe the current understanding of TEAD4 as an oncogene, epigenetic regulator and mitochondrial modulator. The contributing mechanisms will be discussed.
Collapse
Affiliation(s)
- Sheng-Chieh Hsu
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ching-Yu Lin
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yi Lin
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Colin C. Collins
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Chia-Lin Chen
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Chia-Lin Chen, ; Hsing-Jien Kung,
| | - Hsing-Jien Kung
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States
- *Correspondence: Chia-Lin Chen, ; Hsing-Jien Kung,
| |
Collapse
|
13
|
Xu G, Seng Z, Zhang M, Qu J. Angiomotin-like 1 plays a tumor-promoting role in glioma by enhancing the activation of YAP1 signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:2500-2511. [PMID: 34480788 DOI: 10.1002/tox.23363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 06/13/2023]
Abstract
Angiomotin-like 1 (AMOTL1) is reportedly a pivotal tumor-associated protein that is strongly associated with the tumorigenesis of multiple malignant tumors. However, the issue of whether AMOTL1 plays a role in the tumorigenesis of glioma remains unclear. The aim of this work was to explore the possible relationship between AMOTL1 and glioma progression. Results demonstrated that high levels of AMOTL1 in glioma tissues were associated with a reduced survival rate in patients with glioma. Cellular functional assays revealed that silencing of AMOTL1 in glioma cell lines substantially decreased cell proliferation and invasion and increased cell apoptosis. Further investigation revealed that silencing of AMOTL1 inhibited the activation of yes-associated protein 1 (YAP1) and decreased the expression of YAP1 target genes. Reactivation of YAP1 reversed AMOTL1-silencing-induced antitumor effects, whereas inhibition of YAP1 abolished AMOTL1-overexpression-induced tumor-promoting effects in glioma cells. Silencing of AMOTL1 also retarded the growth of glioma cell-derived xenograft tumors in vivo. In conclusion, these findings suggested that AMOTL1 may exert a tumor-promoting function in glioma by enhancing the activation of YAP1 signaling. This work suggested AMOTL1 as a potential target for the development of antiglioma therapy.
Collapse
Affiliation(s)
- Gang Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhiyuan Seng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jianqiang Qu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
14
|
Barrette AM, Ronk H, Joshi T, Mussa Z, Mehrotra M, Bouras A, Nudelman G, Jesu Raj JG, Bozec D, Lam W, Houldsworth J, Yong R, Zaslavsky E, Hadjipanayis CG, Birtwistle MR, Tsankova NM. Anti-invasive efficacy and survival benefit of the YAP-TEAD inhibitor Verteporfin in preclinical glioblastoma models. Neuro Oncol 2021; 24:694-707. [PMID: 34657158 DOI: 10.1093/neuonc/noab244] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) remains a largely incurable disease as current therapy fails to target the invasive nature of GBM growth in disease progression and recurrence. Here we use the FDA-approved drug and small molecule Hippo inhibitor Verteporfin to target YAP-TEAD activity, known to mediate convergent aspects of tumor invasion/metastasis, and assess the drug's efficacy and survival benefit in GBM models. METHODS Up to eight low-passage patient-derived GBM cell lines with distinct genomic drivers, including three primary/recurrent pairs, were treated with Verteporfin or vehicle to assess in-vitro effects on proliferation, migration, YAP-TEAD activity, and transcriptomics. Patient-derived orthotopic xenograft models (PDX) were used to assess Verteporfin's brain penetrance and effects on tumor burden and survival. RESULTS Verteporfin treatment disturbed YAP/TAZ-TEAD activity; disrupted transcriptome signatures related to invasion, epithelial-to-mesenchymal, and proneural-to-mesenchymal transition, phenocopying TEAD1-knockout effects; and impaired tumor migration/invasion dynamics across primary and recurrent GBM lines. In an aggressive orthotopic PDX GBM model, short-term Verteporfin treatment consistently diminished core and infiltrative tumor burden, which was associated with decreased tumor expression of Ki67, nuclear YAP, TEAD1, and TEAD-associated targets EGFR, CDH2 and ITGB1. Finally, long-term Verteporfin treatment appeared non-toxic and conferred survival benefit compared to vehicle in two PDX models: as monotherapy in primary (de-novo) GBM and in combination with Temozolomide chemoradiation in recurrent GBM, where VP treatment associated with increased MGMT methylation. CONCLUSIONS We demonstrate combined anti-invasive and anti-proliferative efficacy for Verteporfin with survival benefit in preclinical GBM models, indicating potential therapeutic value of this already FDA-approved drug if repurposed for glioblastoma patients.
Collapse
Affiliation(s)
- Anne Marie Barrette
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Halle Ronk
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tanvi Joshi
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zarmeen Mussa
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meenakshi Mehrotra
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Bouras
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - German Nudelman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joe G Jesu Raj
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dominique Bozec
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William Lam
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jane Houldsworth
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raymund Yong
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Zaslavsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina, USA
| | - Nadejda M Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
15
|
Luo T, Ding K, Ji J, Zhang X, Yang X, Chen A, Huang B, Zhang D, Wang J, Li X. Cytoskeleton-associated protein 4 (CKAP4) promotes malignant progression of human gliomas through inhibition of the Hippo signaling pathway. J Neurooncol 2021; 154:275-283. [PMID: 34476666 DOI: 10.1007/s11060-021-03831-6] [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] [Received: 04/02/2021] [Accepted: 08/16/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE Gliomas are the most common and aggressive malignant brain tumors and are associated with high mortality and incidence in humans. Despite rigorous multi-modal therapy, including surgery, chemotherapy and radiotherapy, patients with malignant glioma survive an average of 12-15 months following primary diagnosis. Therefore, new molecular biomarkers are urgently needed for diagnosis and targeted therapy. Here, we find that suppression of CKAP4 might inhibit glioma growth through regulation of Hippo signaling. METHODS We examined the expression levels of CKAP4 through analysis of RNA sequencing data from GEPIA and CGGA databases. Then, Lentivirus was used to construct stable cell lines with knockout or overexpression of CKAP4. Next, the function of CKAP4 on glioma was investigated in vitro and in an orthotopic brain tumor model in mice. Lastly, luciferase reporter assay, immunofluorescence and immunoblotting were performed to explore the potential mechanism of how CKAP4 affects gliomas. RESULTS CKAP4 is highly upregulated in glioma and high CKAP4 expressing tumors were associated with poor patient survival. And CKAP4 promotes malignant progression of gliomas via inhibiting Hippo signaling. CONCLUSION CKAP4 has potential as a promising biomarker and can predict the prognosis of patients with gliomas. And targeting CKAP4 expression may be an effective therapeutic strategy for the treatment of human gliomas.
Collapse
Affiliation(s)
- Tao Luo
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Kaikai Ding
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Xiaobing Yang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China.,School of Medicine, Shandong University, Shandong, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China.,Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009, Bergen, Norway.,Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Cheeloo College of Medicine, Shandong University, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China. .,Key Laboratory of Brain Functional Remodeling, Shandong, 250012, Jinan, China. .,Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Key Laboratory of Brain Functional Remodeling, 107# Wenhua Xi Road, Shandong, 250012, Jinan, China.
| |
Collapse
|
16
|
Strepkos D, Markouli M, Papavassiliou KA, Papavassiliou AG, Piperi C. Emerging roles for the YAP/TAZ transcriptional regulators in brain tumour pathology and targeting options. Neuropathol Appl Neurobiol 2021; 48:e12762. [PMID: 34409639 DOI: 10.1111/nan.12762] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 12/23/2022]
Abstract
The transcriptional co-activators Yes-associated protein 1/transcriptional co-activator with PDZ-binding motif (YAP/TAZ) have emerged as significant regulators of a wide variety of cellular and organ functions with impact in early embryonic development, especially during the expansion of the neural progenitor cell pool. YAP/TAZ signalling regulates organ size development, tissue homeostasis, wound healing and angiogenesis by participating in a complex network of various pathways. However, recent evidence suggests an association of these physiologic regulatory effects of YAP/TAZ with pro-oncogenic activities. Herein, we discuss the physiological functions of YAP/TAZ as well as the extensive network of signalling pathways that control their expression and activity, leading to brain tumour development and progression. Furthermore, we describe current targeting approaches and drug options including direct YAP/TAZ and YAP-TEA domain transcription factor (TEAD) interaction inhibitors, G-protein coupled receptors (GPCR) signalling modulators and kinase inhibitors, which may be used to successfully attack YAP/TAZ-dependent tumours.
Collapse
Affiliation(s)
- Dimitrios Strepkos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Mariam Markouli
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas A Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Zheng XH, Wang LL, Zheng MZ, Zhong JJ, Chen YY, Shen YL. RGFP966 inactivation of the YAP pathway attenuates cardiac dysfunction induced by prolonged hypothermic preservation. J Zhejiang Univ Sci B 2021; 21:703-715. [PMID: 32893527 PMCID: PMC7519627 DOI: 10.1631/jzus.b2000026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022]
Abstract
Oxidative stress and apoptosis are the key factors that limit the hypothermic preservation time of donor hearts to within 4-6 h. The aim of this study was to investigate whether the histone deacetylase 3 (HDAC3) inhibitor RGFP966 could protect against cardiac injury induced by prolonged hypothermic preservation. Rat hearts were hypothermically preserved in Celsior solution with or without RGFP966 for 12 h followed by 60 min of reperfusion. Hemodynamic parameters during reperfusion were evaluated. The expression and phosphorylation levels of mammalian STE20-like kinase-1 (Mst1) and Yes-associated protein (YAP) were determined by western blotting. Cell apoptosis was measured by the terminal deoxynucleotidyl-transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) method. Addition of RGFP966 in Celsior solution significantly inhibited cardiac dysfunction induced by hypothermic preservation. RGFP966 inhibited the hypothermic preservation-induced increase of the phosphorylated (p)-Mst1/Mst1 and p-YAP/YAP ratios, prevented a reduction in total YAP protein expression, and increased the nuclear YAP protein level. Verteporfin (VP), a small molecular inhibitor of YAP-transcriptional enhanced associate domain (TEAD) interaction, partially abolished the protective effect of RGFP966 on cardiac function, and reduced lactate dehydrogenase activity and malondialdehyde content. RGFP966 increased superoxide dismutase, catalase, and glutathione peroxidase gene and protein expression, which was abolished by VP. RGFP966 inhibited hypothermic preservation-induced overexpression of B-cell lymphoma protein 2 (Bcl-2)-associated X (Bax) and cleaved caspase-3, increased Bcl-2 mRNA and protein expression, and reduced cardiomyocyte apoptosis. The antioxidant and anti-apoptotic effects of RGFP966 were cancelled by VP. The results suggest that supplementation of Celsior solution with RGFP966 attenuated prolonged hypothermic preservation-induced cardiac dysfunction. The mechanism may involve inhibition of oxidative stress and apoptosis via inactivation of the YAP pathway.
Collapse
Affiliation(s)
- Xiao-he Zheng
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lin-lin Wang
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Orthopaedics, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Ming-zhi Zheng
- Department of Pharmacology, Hangzhou Medical College, Hangzhou 310053, China
| | - Jin-jie Zhong
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Obstetrics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ying-ying Chen
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
- Department of Obstetrics, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yue-liang Shen
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
18
|
O’Connor SA, Feldman HM, Arora S, Hoellerbauer P, Toledo CM, Corrin P, Carter L, Kufeld M, Bolouri H, Basom R, Delrow J, McFaline‐Figueroa JL, Trapnell C, Pollard SM, Patel A, Paddison PJ, Plaisier CL. Neural G0: a quiescent-like state found in neuroepithelial-derived cells and glioma. Mol Syst Biol 2021; 17:e9522. [PMID: 34101353 PMCID: PMC8186478 DOI: 10.15252/msb.20209522] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/30/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
Single-cell RNA sequencing has emerged as a powerful tool for resolving cellular states associated with normal and maligned developmental processes. Here, we used scRNA-seq to examine the cell cycle states of expanding human neural stem cells (hNSCs). From these data, we constructed a cell cycle classifier that identifies traditional cell cycle phases and a putative quiescent-like state in neuroepithelial-derived cell types during mammalian neurogenesis and in gliomas. The Neural G0 markers are enriched with quiescent NSC genes and other neurodevelopmental markers found in non-dividing neural progenitors. Putative glioblastoma stem-like cells were significantly enriched in the Neural G0 cell population. Neural G0 cell populations and gene expression are significantly associated with less aggressive tumors and extended patient survival for gliomas. Genetic screens to identify modulators of Neural G0 revealed that knockout of genes associated with the Hippo/Yap and p53 pathways diminished Neural G0 in vitro, resulting in faster G1 transit, down-regulation of quiescence-associated markers, and loss of Neural G0 gene expression. Thus, Neural G0 represents a dynamic quiescent-like state found in neuroepithelial-derived cells and gliomas.
Collapse
Affiliation(s)
- Samantha A O’Connor
- School of Biological and Health Systems EngineeringArizona State UniversityTempeAZUSA
| | - Heather M Feldman
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Sonali Arora
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Pia Hoellerbauer
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
- Molecular and Cellular Biology ProgramUniversity of WashingtonSeattleWAUSA
| | - Chad M Toledo
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
- Molecular and Cellular Biology ProgramUniversity of WashingtonSeattleWAUSA
| | - Philip Corrin
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Lucas Carter
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Megan Kufeld
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Hamid Bolouri
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Ryan Basom
- Genomics and Bioinformatics Shared ResourcesFred Hutchinson Cancer Research CenterSeattleWAUSA
| | - Jeffrey Delrow
- Genomics and Bioinformatics Shared ResourcesFred Hutchinson Cancer Research CenterSeattleWAUSA
| | | | - Cole Trapnell
- Department of Genome SciencesUniversity of WashingtonSeattleWAUSA
| | - Steven M Pollard
- Edinburgh CRUK Cancer Research CentreMRC Centre for Regenerative MedicineThe University of EdinburghEdinburghUK
| | - Anoop Patel
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
- Department of NeurosurgeryUniversity of WashingtonSeattleWAUSA
| | - Patrick J Paddison
- Human Biology DivisionFred Hutchinson Cancer Research CenterSeattleWAUSA
- Molecular and Cellular Biology ProgramUniversity of WashingtonSeattleWAUSA
| | | |
Collapse
|
19
|
Curry RN, Glasgow SM. The Role of Neurodevelopmental Pathways in Brain Tumors. Front Cell Dev Biol 2021; 9:659055. [PMID: 34012965 PMCID: PMC8127784 DOI: 10.3389/fcell.2021.659055] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Disruptions to developmental cell signaling pathways and transcriptional cascades have been implicated in tumor initiation, maintenance and progression. Resurgence of aberrant neurodevelopmental programs in the context of brain tumors highlights the numerous parallels that exist between developmental and oncologic mechanisms. A deeper understanding of how dysregulated developmental factors contribute to brain tumor oncogenesis and disease progression will help to identify potential therapeutic targets for these malignancies. In this review, we summarize the current literature concerning developmental signaling cascades and neurodevelopmentally-regulated transcriptional programs. We also examine their respective contributions towards tumor initiation, maintenance, and progression in both pediatric and adult brain tumors and highlight relevant differentiation therapies and putative candidates for prospective treatments.
Collapse
Affiliation(s)
- Rachel N. Curry
- Department of Neuroscience, Baylor College of Medicine, Center for Cell and Gene Therapy, Houston, TX, United States
- Integrative Molecular and Biomedical Sciences, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX, United States
| | - Stacey M. Glasgow
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States
- Neurosciences Graduate Program, University of California, San Diego, San Diego, CA, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
20
|
Huang R, Dong R, Wang N, He Y, Zhu P, Wang C, Lan B, Gao Y, Sun L. Adaptive Changes Allow Targeting of Ferroptosis for Glioma Treatment. Cell Mol Neurobiol 2021; 42:2055-2074. [PMID: 33893939 DOI: 10.1007/s10571-021-01092-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 04/16/2021] [Indexed: 12/16/2022]
Abstract
Ferroptosis is a type of regulated cell death that plays an essential role in various brain diseases, including cranial trauma, neuronal diseases, and brain tumors. It has been reported that cancer cells rely on their robust antioxidant capacity to escape ferroptosis. Therefore, ferroptosis exploitation could be an effective strategy to prevent tumor proliferation and invasion. Glioma is a common malignant craniocerebral tumor exhibiting complicated drug resistance and survival mechanisms, resulting in a high mortality rate and short survival time. Recent studies have determined that metabolic alterations in glioma offer exploitable therapeutic targets. These metabolic alterations allow targeted therapy to achieve some initial efficacy but have failed to inhibit glioma growth, invasion, and drug resistance effectively. It has been proposed that the reason for the high malignancy and drug resistance observed with glioma is that these tumors can effectively evade ferroptosis. Ferroptosis-inducing drugs were found to exert a positive effect by targeting this particular characteristic of glioma cells. Moreover, gliomas develop enhanced drug resistance through anti-ferroptosis mechanisms. In this study, we provided an overview of the mechanisms by which glioma aggressiveness and drug resistance are mediated by the evasion of ferroptosis. This information might provide new targets for glioma therapy as well as new insights and ideas for future research.
Collapse
Affiliation(s)
- Renxuan Huang
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Rui Dong
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Nan Wang
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Yichun He
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Peining Zhu
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Chong Wang
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Beiwu Lan
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China
| | - Yufei Gao
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, China.
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
21
|
Multiregional Sequencing of IDH-WT Glioblastoma Reveals High Genetic Heterogeneity and a Dynamic Evolutionary History. Cancers (Basel) 2021; 13:cancers13092044. [PMID: 33922652 PMCID: PMC8122908 DOI: 10.3390/cancers13092044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Glioblastoma is the most common and aggressive primary brain malignancy in adults. In addition to extensive inter-patient heterogeneity, glioblastoma shows intra-tumor extensive cellular and molecular heterogeneity, both spatially and temporally. This heterogeneity is one of the main reasons for the poor prognosis and overall survival. Moreover, it raises the important question of whether the molecular characterization of a single biopsy sample, as performed in standard diagnostics, actually represents the entire lesion. In this study, we sequenced the whole exome of nine spatially different cancer regions of three primary glioblastomas. We characterized their mutational profiles and copy number alterations, with implications for our understanding of tumor biology in relation to clonal architecture and evolutionary dynamics, as well as therapeutically relevant alterations. Abstract Glioblastoma is one of the most common and lethal primary neoplasms of the brain. Patient survival has not improved significantly over the past three decades and the patient median survival is just over one year. Tumor heterogeneity is thought to be a major determinant of therapeutic failure and a major reason for poor overall survival. This work aims to comprehensively define intra- and inter-tumor heterogeneity by mapping the genomic and mutational landscape of multiple areas of three primary IDH wild-type (IDH-WT) glioblastomas. Using whole exome sequencing, we explored how copy number variation, chromosomal and single loci amplifications/deletions, and mutational burden are spatially distributed across nine different tumor regions. The results show that all tumors exhibit a different signature despite the same diagnosis. Above all, a high inter-tumor heterogeneity emerges. The evolutionary dynamics of all identified mutations within each region underline the questionable value of a single biopsy and thus the therapeutic approach for the patient. Multiregional collection and subsequent sequencing are essential to try to address the clinical challenge of precision medicine. Especially in glioblastoma, this approach could provide powerful support to pathologists and oncologists in evaluating the diagnosis and defining the best treatment option.
Collapse
|
22
|
Kim Y, Varn FS, Park SH, Yoon BW, Park HR, Lee C, Verhaak RGW, Paek SH. Perspective of mesenchymal transformation in glioblastoma. Acta Neuropathol Commun 2021; 9:50. [PMID: 33762019 PMCID: PMC7992784 DOI: 10.1186/s40478-021-01151-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
Despite aggressive multimodal treatment, glioblastoma (GBM), a grade IV primary brain tumor, still portends a poor prognosis with a median overall survival of 12–16 months. The complexity of GBM treatment mainly lies in the inter- and intra-tumoral heterogeneity, which largely contributes to the treatment-refractory and recurrent nature of GBM. By paving the road towards the development of personalized medicine for GBM patients, the cancer genome atlas classification scheme of GBM into distinct transcriptional subtypes has been considered an invaluable approach to overcoming this heterogeneity. Among the identified transcriptional subtypes, the mesenchymal subtype has been found associated with more aggressive, invasive, angiogenic, hypoxic, necrotic, inflammatory, and multitherapy-resistant features than other transcriptional subtypes. Accordingly, mesenchymal GBM patients were found to exhibit worse prognosis than other subtypes when patients with high transcriptional heterogeneity were excluded. Furthermore, identification of the master mesenchymal regulators and their downstream signaling pathways has not only increased our understanding of the complex regulatory transcriptional networks of mesenchymal GBM, but also has generated a list of potent inhibitors for clinical trials. Importantly, the mesenchymal transition of GBM has been found to be tightly associated with treatment-induced phenotypic changes in recurrence. Together, these findings indicate that elucidating the governing and plastic transcriptomic natures of mesenchymal GBM is critical in order to develop novel and selective therapeutic strategies that can improve both patient care and clinical outcomes. Thus, the focus of our review will be on the recent advances in the understanding of the transcriptome of mesenchymal GBM and discuss microenvironmental, metabolic, and treatment-related factors as critical components through which the mesenchymal signature may be acquired. We also take into consideration the transcriptomic plasticity of GBM to discuss the future perspectives in employing selective therapeutic strategies against mesenchymal GBM.
Collapse
|
23
|
Zhao M, Zhang Y, Jiang Y, Wang K, Wang X, Zhou D, Wang Y, Yu R, Zhou X. YAP promotes autophagy and progression of gliomas via upregulating HMGB1. J Exp Clin Cancer Res 2021; 40:99. [PMID: 33726796 PMCID: PMC7968184 DOI: 10.1186/s13046-021-01897-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Due to the hypoxia and nutrient deficiency microenvironment, glioblastoma (GBM) exhibits high autophagy activity and autophagy plays an important role in the progression of GBM. However, the molecular mechanism of autophagy in GBM progression remains unclear. The aim of this study is to delve out the role and mechanism of yes-associated protein (YAP) in GBM autophagy and progression. METHODS The level of autophagy or autophagy flux were assessed by using western blotting, GFP-LC3 puncta (Live) imaging, transmission electron microscopy and GFP-RFP-LC3 assay. The GBM progression was detected by using CCK8, EdU, nude mouse xenograft and Ki67 staining. Isobaric tags for relative and absolute quantification (iTraq) quantitative proteomics was used to find out the mediator of YAP in autophagy. Expression levels of YAP and HMGB1 in tissue samples from GBM patients were examined by Western blotting, tissue microarray and immunohistochemistry. RESULTS YAP over-expression enhanced glioma cell autophagy under basal and induced conditions. In addition, blocking autophagy by chloroquine abolished the promoting effect of YAP on glioma growth. Mechanistically, YAP over-expression promoted the transcription and translocation of high mobility group box 1(HMGB1), a well-known regulator of autophagy, from nucleus to cytoplasm. Down-regulation of HMGB1 abolished the promoting effect of YAP on autophagy and glioma growth. Furthermore, the expression of YAP and HMGB1 were positively associated with each other and suggested poor prognosis for clinical GBM. CONCLUSION YAP promoted glioma progression by enhancing HMGB1-mediated autophagy, indicating that YAP-HMGB1 axis was a feasible therapeutic target for GBM. Our study revealed a clinical opportunity involving the combination of chemo-radiotherapy with pharmacological autophagy inhibition for treating GBM patients with YAP high expression.
Collapse
Affiliation(s)
- Min Zhao
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Yang Jiang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Present address: Clinical Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Kai Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Xiang Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Ding Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Yan Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
24
|
Targeting the YAP-TEAD interaction interface for therapeutic intervention in glioblastoma. J Neurooncol 2021; 152:217-231. [PMID: 33511508 DOI: 10.1007/s11060-021-03699-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Recent studies have suggested that dysregulated Hippo pathway signaling may contribute to glioblastoma proliferation and invasive characteristics. The downstream effector of the pathway, the Yes-associated protein (YAP) oncoprotein, has emerged as a promising target in glioblastoma multiforme (GBM). METHODS Utilizing a high-throughput yeast two-hybrid based screen, a small molecule was identified which inhibits the association of the co-transcriptional activator YAP1 and the TEA domain family member 1 (TEAD1) transcription factor protein-protein interaction interface. This candidate inhibitor, NSC682769, a novel benzazepine compound, was evaluated for its ability to affect Hippo/YAP axis signaling and potential anti-glioblastoma properties. RESULTS NSC682769 potently blocked association of YAP and TEAD in vitro and in GBM cells treated with submicromolar concentrations. Moreover, inhibitor-coupled bead pull down and surface plasmon resonance analyses demonstrate that NSC682769 binds to YAP. NSC682769 treatment of GBM lines and patient derived cells resulted in downregulation of YAP expression levels resulting in curtailed YAP-TEAD transcriptional activity. In GBM cell models, NSC682769 inhibited proliferation, colony formation, migration, invasiveness and enhanced apoptosis. In tumor xenograft and genetically engineered mouse models, NSC682769 exhibited marked anti-tumor responses and resulted in increased overall survival and displayed significant blood-brain barrier penetration. CONCLUSIONS These results demonstrate that blockade of YAP-TEAD association is a viable therapeutic strategy for glioblastoma. On the basis of these favorable preclinical studies further clinical studies are warranted.
Collapse
|
25
|
MiR-429 Involves in the Pathogenesis of Colorectal Cancer via Directly Targeting LATS2. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5316276. [PMID: 33414893 PMCID: PMC7769676 DOI: 10.1155/2020/5316276] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/18/2020] [Accepted: 12/10/2020] [Indexed: 11/17/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death around the world whose recurrence and metastasis rate is high. Due to the underlying unclear pathogenesis, it is hard so far to predict the tumorigenesis and prevent its recurrence. YAP/TAZ has been reported to be activated and functioned as a potential oncogene in multiple cancer types and proved to be essential for the carcinogenesis of most solid tumors. In the present study, we found that YAP/TAZ was markedly upregulated in CRC tissues comparing with the adjacent noncancerous tissues due to the downregulation of LATS2, the main upstream regulator. We further identified miR-429 as a direct regulator of LATS2-YAP/TAZ activation, suggesting that the miR-429-LATS2-YAP/TAZ might be novel effective diagnostic axis and therapeutic targets for CRC.
Collapse
|
26
|
Wang JJ, Wang H, Zhu BL, Wang X, Qian YH, Xie L, Wang WJ, Zhu J, Chen XY, Wang JM, Ding ZL. Development of a prognostic model of glioma based on immune-related genes. Oncol Lett 2020; 21:116. [PMID: 33376548 PMCID: PMC7751470 DOI: 10.3892/ol.2020.12377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Glioma is the most common type of primary brain cancer, and the prognosis of most patients with glioma, and particularly that of patients with glioblastoma, is poor. Tumor immunity serves an important role in the development of glioma. However, immunotherapy for glioma has not been completely successful, and thus, comprehensive examination of the immune-related genes (IRGs) of glioma is required. In the present study, differentially expressed genes (DEGs) and differentially expressed IRGs (DEIRGs) were identified using the edgeR package. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was used for functional enrichment analysis of DEIRGs. Survival-associated IRGs were selected via univariate Cox regression analysis. A The Cancer Genome Atlas prognostic model and GSE43378 validation model were established using lasso-penalized Cox regression analysis. Based on the median risk score value, patients were divided into high-risk and low-risk groups for clinical analysis. Receiver operating characteristic curve and nomogram analyses were used to assess the accuracy of the models. Reverse transcription-quantitative PCR was performed to measure the expression levels of relevant genes, such as cyclin-dependent kinase 4 (CDK4), interleukin 24 (IL24), NADPH oxidase 4 (NOX4), bone morphogenetic protein 2 (BMP2) and baculoviral IAP repeat containing 5 (BIRC5). A total of 3,238 DEGs, including 1,950 upregulated and 1,288 downregulated DEGs, and 97 DEIRGs, including 60 upregulated and 37 downregulated DEIRGs, were identified. ‘Neuroactive ligand-receptor interaction’ and ‘Cytokine-cytokine receptor interaction’ were the most significantly enriched pathways according to KEGG pathway analysis. A prognostic model and a validation prognostic model were created for glioma, including 15 survival-associated IRGs (FCER1G, NOX4, TRIM5, SOCS1, APOBEC3C, BIRC5, VIM, TNC, BMP2, CMTM3, IL24, JAG1, CALCRL, HNF4G and CDK4). Furthermore, multivariate Cox regression analysis results suggested that age, high WHO Grade by histopathology, wild type isocitrate dehydrogenase 1 and high risk score were independently associated with poor overall survival. The infiltration of B cells, CD8+ T cells, dendritic cells, macrophages and neutrophils was positively associated with the prognostic risk score. In the present study, several clinically significant survival-associated IRGs were identified, and a prognosis evaluation model of glioma was established.
Collapse
Affiliation(s)
- Jing-Jing Wang
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Han Wang
- Department of Oncology, Jining Cancer Hospital, Jining, Shandong 272000, P.R. China
| | - Bao-Long Zhu
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Xiang Wang
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Yong-Hong Qian
- Department of Radio-Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Lei Xie
- Department of Oncology, Taizhou Hospital of Traditional Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Wen-Jie Wang
- Department of Radio-Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Jie Zhu
- Department of Oncology, Changzhou Traditional Chinese Medical Hospital, Changzhou, Jiangsu, 213003, P.R. China
| | - Xing-Yu Chen
- Department of General Surgery, Taizhou Fourth People's Hospital, Taizhou, Jiangsu 225300, P.R. China
| | - Jing-Mei Wang
- Department of Geriatrics, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310002, P.R. China
| | - Zhi-Liang Ding
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
27
|
Vigneswaran K, Boyd NH, Oh SY, Lallani S, Boucher A, Neill SG, Olson JJ, Read RD. YAP/TAZ Transcriptional Coactivators Create Therapeutic Vulnerability to Verteporfin in EGFR-mutant Glioblastoma. Clin Cancer Res 2020; 27:1553-1569. [PMID: 33172899 DOI: 10.1158/1078-0432.ccr-20-0018] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 08/04/2020] [Accepted: 11/06/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Glioblastomas (GBMs), neoplasms derived from glia and neuroglial progenitor cells, are the most common and lethal malignant primary brain tumors diagnosed in adults, with a median survival of 14 months. GBM tumorigenicity is often driven by genetic aberrations in receptor tyrosine kinases, such as amplification and mutation of EGFR. EXPERIMENTAL DESIGN Using a Drosophila glioma model and human patient-derived GBM stem cells and xenograft models, we genetically and pharmacologically tested whether the YAP and TAZ transcription coactivators, effectors of the Hippo pathway that promote gene expression via TEA domain (TEAD) cofactors, are key drivers of GBM tumorigenicity downstream of oncogenic EGFR signaling. RESULTS YAP and TAZ are highly expressed in EGFR-amplified/mutant human GBMs, and their knockdown in EGFR-amplified/mutant GBM cells inhibited proliferation and elicited apoptosis. Our results indicate that YAP/TAZ-TEAD directly regulates transcription of SOX2, C-MYC, and EGFR itself to create a feedforward loop to drive survival and proliferation of human GBM cells. Moreover, the benzoporphyrin derivative verteporfin, a disruptor of YAP/TAZ-TEAD-mediated transcription, preferentially induced apoptosis of cultured patient-derived EGFR-amplified/mutant GBM cells, suppressed expression of YAP/TAZ transcriptional targets, including EGFR, and conferred significant survival benefit in an orthotopic xenograft GBM model. Our efforts led us to design and initiate a phase 0 clinical trial of Visudyne, an FDA-approved liposomal formulation of verteporfin, where we used intraoperative fluorescence to observe verteporfin uptake into tumor cells in GBM tumors in human patients. CONCLUSIONS Together, our data suggest that verteporfin is a promising therapeutic agent for EGFR-amplified and -mutant GBM.
Collapse
Affiliation(s)
| | - Nathaniel H Boyd
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Se-Yeong Oh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Shoeb Lallani
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Andrew Boucher
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| | - Stewart G Neill
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia.,Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Renee D Read
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia. .,Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
28
|
Rao HC, Wu ZK, Wei SD, Jiang Y, Guo QX, Wang JW, Chen CX, Yang HY. MiR-25-3p Serves as an Oncogenic MicroRNA by Downregulating the Expression of Merlin in Osteosarcoma. Cancer Manag Res 2020; 12:8989-9001. [PMID: 33061594 PMCID: PMC7522417 DOI: 10.2147/cmar.s262245] [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: 05/11/2020] [Accepted: 08/13/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Moesin-ezrin-radixin-like protein (Merlin) has been identified as a tumor suppressor in several types of cancers. However, the biological function of Merlin in osteosarcoma remains unclear. MicroRNAs (miRNAs) can influence cancer progression by targeting oncogenes or anti-oncogenes. In this study, we sought to evaluate the regulation of Merlin expression by miR-25-3p and the role of the miR-25-3p/Merlin axis in osteosarcoma progression, with the aim of identifying a potential therapeutic target for osteosarcoma. MATERIALS AND METHODS TCGA (The Cancer Genome Atlas) database was used to analyze the correlation between Merlin expression and prognosis. RT-qPCR and Western blotting analyses were performed to compare Merlin expression between normal and malignant cells. A dual-luciferase reporter assay was performed to evaluate the direct targeting of Merlin by miR-25-3p. We overexpressed miR-25-3p, or/and Merlin, in U-2 OS and 143B cells, and studied their cellular functions in vitro. MTT and colony formation assays were performed to determine the effects on cell growth. EdU and cell cycle assays were performed to analyze the effects in cell replication. We used annexin V-fluorescein isothiocyanate and propidium iodide to stain apoptotic cells, and analyzed the cells using flow cytometry. The effects on cell metastasis were studied in wound healing and transwell assays. Lastly, the underlying mechanism was determined in RT-qPCR and Western blotting experiments. RESULTS Low Merlin expression was linked to poor prognosis. miR-25-3p was observed to directly target Merlin and downregulate its expression. miR-25-3p promoted cell growth, migration, and invasion, and inhibited apoptosis induced by cisplatin. Moreover, the overexpression of Merlin reversed the abovementioned effects of miR-25-3p. Further, the miR-25-3p/Merlin axis was observed to play an important role in the Hippo pathway, and regulated the expression of genes such as BIRC5, CTGF, and CYR61. CONCLUSION miR-25-3p functions as an oncogenic microRNA in osteosarcoma by targeting Merlin, and may serve as a potential therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Hua-Chun Rao
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Zhao-Ke Wu
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Si-da Wei
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Yun Jiang
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Qing-Xin Guo
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Jia-Wen Wang
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Chang-Xian Chen
- Quanzhou Orthopedic-Traumatological Hospital, Fengze District, Quanzhou, Fujian, People's Republic of China
| | - Hui-Yong Yang
- School of Medicine, Institute of Molecular Medicine, Huaqiao University, Quanzhou, People's Republic of China
| |
Collapse
|
29
|
Xue J, Sang W, Su LP, Gao HX, Cui WL, Abulajiang G, Wang Q, Zhang J, Zhang W. Proteomics reveals protein phosphatase 1γ as a biomarker associated with Hippo signal pathway in glioma. Pathol Res Pract 2020; 216:153187. [PMID: 32919304 DOI: 10.1016/j.prp.2020.153187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/12/2022]
Abstract
Hub proteins related with Hippo signal pathway in glioma were investigated using proteomics methods (Tandem Mass Tag, TMT) to determine the differentially expressed proteins in glioblastoma (GBM). Ingenuity Pathway Analysis (IPA) was performed to complement proteomic findings by identifying the top canonical pathways as well as to suggest novel proteins for the targeted therapy of glioma. A total of 222 formalin-fixed paraffin-embedded (FFPE) glioma tissue samples were used to verify the expression of protein phosphatase 1γ (PP1γ), Yes-associated protein 1 (YAP1), and SOX2 via immunohistochemistry. Bioinformatics analysis revealed these proteins as crucial in the Hippo signaling pathway in GBM. Spearman correlation was performed to analyze the relationship of these three proteins, and survival analysis was conducted to investigate their effects on prognosis. Among the 5808 proteins identified by TMT with the standard of P-value < 0.05 and fold change (FC) of>1.2 or <0.83, 1398 upregulated and 1060 downregulated differentially expressed proteins were found. IPA revealed that the Hippo signaling was activated in the top 10 canonical pathways, and PP1γ was activated in the Hippo signaling. Immunohistochemistry analysis indicated that PP1γ, YAP1, and SOX2 were highly and positively expressed in glioma. PP1γ expression was related to WHO grade (p = 0.003) and ki-67 expression (p = 0.012). Low PP1γ expression was associated with IDH1-mut in low-grade glioma (LGG; WHO grades II and III) (p = 0.037). PP1γ was positively correlated with YAP1 (p < 0.001; r = 0.259) and SOX2 (p = 0.009; r = 0.175). In survival analysis, age, WHO grade, ki-67 expression, and PP1γ expression independently predicted a short OS in total cohort (p < 0.05). Therefore, PP1γ is a hub protein associated with Hippo signal pathway in glioma, and its expression indicates poor prognosis in patients with glioma. Therefore, PP1γ may be a promising prognostic biomarker and a therapeutic target in glioma.
Collapse
Affiliation(s)
- Jing Xue
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China; Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830011, PR China; Department of Pathology, Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, No. 116 Huanghe Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830000, PR China
| | - Wei Sang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China
| | - Li-Ping Su
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China
| | - Hai-Xia Gao
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China; Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830011, PR China
| | - Wen-Li Cui
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China
| | - Gulinaer Abulajiang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China
| | - Qian Wang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China; Xinjiang Medical University, No. 393 Xinyi Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830011, PR China
| | - Jing Zhang
- Department of Pathology, Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, No. 116 Huanghe Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830000, PR China
| | - Wei Zhang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan Southern Road, Urumqi, The Xinjiang Uygur Autonomous Region of China, 830054, PR China.
| |
Collapse
|
30
|
Shang Y, Yan Y, Chen B, Zhang J, Zhang T. Over-expressed MST1 impaired spatial memory via disturbing neural oscillation patterns in mice. GENES BRAIN AND BEHAVIOR 2020; 19:e12678. [PMID: 32468668 DOI: 10.1111/gbb.12678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 11/28/2022]
Abstract
The activated mammalian Ste20-like serine/threonine kinases 1 (MST1) was found in the central nervous system diseases, such as cerebral ischemia, stroke and ALS, which were related with cognitions. The aim of this study was to examine the effect of elevated MST1 on memory functions in C57BL/6J mice. We also explored the underlying mechanism about the pattern alteration of neural oscillations, closely associated with cognitive dysfunctions, at different physiological rhythms, which were related to a wide range of basic and higher-level cognitive activities. A mouse model of the adeno-associated virus (AAV)-mediated overexpression of MST1 was established. The behavioral experiments showed that spatial memory was significantly damaged in MST1 mice. The distribution of either theta or gamma power was clearly disturbed in MST1 animals. Moreover, the synchronization in both theta and gamma rhythms, and theta-gamma cross-frequency coupling were significantly weakened in MST1 mice. In addition, the expressions of GABAA receptor, GAD67 and parvalbumin (PV) were obviously increased in MST1 mice. Meanwhile, blocking MST1 activity could inhibit the activation of FOXO3a and YAP. The above data suggest that MST1-overexpression may induce memory impairments via disturbing the patterns of neural activities, which is possibly associated with the abnormal GABAergic expression level.
Collapse
Affiliation(s)
- Yingchun Shang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China
| | - Yuxing Yan
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China
| | - Bin Chen
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China
| | - Jianhai Zhang
- School of Computer Science & Technology, and Key Laboratory of Brain Machine Collaborative Intelligence of Zhejiang Province, Hangzhou Dianzi University, Hangzhou, China
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China
| |
Collapse
|
31
|
Kierulf-Vieira KS, Sandberg CJ, Waaler J, Lund K, Skaga E, Saberniak BM, Panagopoulos I, Brandal P, Krauss S, Langmoen IA, Vik-Mo EO. A Small-Molecule Tankyrase Inhibitor Reduces Glioma Stem Cell Proliferation and Sphere Formation. Cancers (Basel) 2020; 12:cancers12061630. [PMID: 32575464 PMCID: PMC7352564 DOI: 10.3390/cancers12061630] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/19/2022] Open
Abstract
Evidence suggests that the growth and therapeutic resistance of glioblastoma (GBM) may be enabled by a population of glioma stem cells (GSCs) that are regulated by typical stem cell pathways, including the WNT/β-catenin signaling pathway. We wanted to explore the effect of treating GSCs with a small-molecule inhibitor of tankyrase, G007-LK, which has been shown to be a potent modulator of the WNT/β-catenin and Hippo pathways in colon cancer. Four primary GSC cultures and two primary adult neural stem cell cultures were treated with G007-LK and subsequently evaluated through the measurement of growth characteristics, as well as the expression of WNT/β-catenin and Hippo signaling pathway-related proteins and genes. Treatment with G007-LK decreased in vitro proliferation and sphere formation in all four primary GSC cultures in a dose-dependent manner. G007-LK treatment altered the expression of key downstream WNT/β-catenin and Hippo signaling pathway-related proteins and genes. Finally, cotreatment with the established GBM chemotherapeutic compound temozolomide (TMZ) led to an additive reduction in sphere formation, suggesting that WNT/β-catenin signaling may contribute to TMZ resistance. These observations suggest that tankyrase inhibition may serve as a supplement to current GBM therapy, although more work is needed to determine the exact downstream mechanisms involved.
Collapse
Affiliation(s)
- Kirsten Strømme Kierulf-Vieira
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (C.J.S.); (E.S.); (B.M.S.); (I.A.L.); (E.O.V.-M.)
- Norwegian Stem Cell Center, Oslo University Hospital, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Correspondence:
| | - Cecilie Jonsgar Sandberg
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (C.J.S.); (E.S.); (B.M.S.); (I.A.L.); (E.O.V.-M.)
- Norwegian Stem Cell Center, Oslo University Hospital, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
| | - Jo Waaler
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (J.W.); (K.L.); (S.K.)
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 OSLO, Norway
| | - Kaja Lund
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (J.W.); (K.L.); (S.K.)
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 OSLO, Norway
| | - Erlend Skaga
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (C.J.S.); (E.S.); (B.M.S.); (I.A.L.); (E.O.V.-M.)
- Norwegian Stem Cell Center, Oslo University Hospital, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
| | - Birthe Mikkelsen Saberniak
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (C.J.S.); (E.S.); (B.M.S.); (I.A.L.); (E.O.V.-M.)
- Norwegian Stem Cell Center, Oslo University Hospital, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
| | - Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, P.O. Box 49534 Nydalen, 0424 Oslo, Norway; (I.P.); (P.B.)
| | - Petter Brandal
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, P.O. Box 49534 Nydalen, 0424 Oslo, Norway; (I.P.); (P.B.)
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, P.O. Box 49534 Nydalen, 0424 Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
| | - Stefan Krauss
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (J.W.); (K.L.); (S.K.)
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 OSLO, Norway
| | - Iver Arne Langmoen
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (C.J.S.); (E.S.); (B.M.S.); (I.A.L.); (E.O.V.-M.)
- Norwegian Stem Cell Center, Oslo University Hospital, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway
| | - Einar Osland Vik-Mo
- Vilhelm Magnus Laboratory for Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway; (C.J.S.); (E.S.); (B.M.S.); (I.A.L.); (E.O.V.-M.)
- Norwegian Stem Cell Center, Oslo University Hospital, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, P.O. Box 1112 Blindern, 0317 Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
32
|
Ouyang T, Meng W, Li M, Hong T, Zhang N. Recent Advances of the Hippo/YAP Signaling Pathway in Brain Development and Glioma. Cell Mol Neurobiol 2020; 40:495-510. [PMID: 31768921 DOI: 10.1007/s10571-019-00762-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/16/2019] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway is highly conserved from Drosophila melanogaster to mammals and plays a crucial role in organ size control, tissue regeneration, and tumor suppression. The Yes-associated protein (YAP) is an important transcriptional co-activator that is negatively regulated by the Hippo signaling pathway. The Hippo signaling pathway is also regulated by various upstream regulators, such as cell polarity, adhesion proteins, and other signaling pathways (the Wnt/β-catenin, Notch, and MAPK pathways). Recently, accumulated evidence suggests that the Hippo/YAP signaling pathway plays important roles in central nervous system development and brain tumor, including glioma. In this review, we summarize the results of recent studies on the physiological effect of the Hippo/YAP signaling pathway in neural stem cells, neural progenitor cells, and glial cells. In particular, we also focus on the expression of MST1/2, LATS1/2, and the downstream effector YAP, in glioma, and offer a review of the latest research of the Hippo/YAP signaling pathway in glioma pathogenesis. Finally, we also present future research directions and potential therapeutic strategies for targeting the Hippo/YAP signaling in glioma.
Collapse
Affiliation(s)
- Taohui Ouyang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi Province, China
| | - Wei Meng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi Province, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi Province, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Jiangxi Province, China
| | - Na Zhang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Jiangxi Province, No.17, Yongwai Street, Nanchang, 336000, China.
| |
Collapse
|
33
|
Qi Y, Sun D, Yang W, Xu B, Lv D, Han Y, Sun M, Jiang S, Hu W, Yang Y. Mammalian Sterile 20-Like Kinase (MST) 1/2: Crucial Players in Nervous and Immune System and Neurological Disorders. J Mol Biol 2020; 432:3177-3190. [PMID: 32198112 DOI: 10.1016/j.jmb.2020.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/25/2020] [Accepted: 03/09/2020] [Indexed: 12/28/2022]
Abstract
As central components of the Hippo signaling pathway in mammals, the mammalian sterile 20-like kinase 1 (MST1) and MST2 protein kinases regulate cell proliferation, survival, and death and are involved in the homeostasis of many tissues. Recent studies have elucidated the roles of MST1 and MST2 in the nervous system and immune system, particularly in neurological disorders, which are influenced by aging. In this review, we provide a comprehensive overview of these research areas. First, the activation mechanisms and roles of MST1 and MST2 in neurons, non-neuronal cells, and immune cells are introduced. The roles of MST1 and MST2 in neurological disorders, including brain tumors, cerebrovascular diseases, neurodegenerative disorders, and neuromuscular disorders, are then presented. Finally, the existing obstacles for further research are discussed. Collectively, the information compiled herein provides a common framework for the function of MST1 and MST2 in the nervous system, should contribute to the design of further experiments, and sheds light on potential treatments for aging associated neurological disorders.
Collapse
Affiliation(s)
- Yating Qi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Wenwen Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Baoping Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Dewen Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Yuehu Han
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Meng Sun
- Department of Cardiology, The First Hospital of Shanxi Medical University, 85 Jiefang South Road, Taiyuan 030001, China
| | - Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Wei Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| |
Collapse
|
34
|
Baglo Y, Sorrin AJ, Liang BJ, Huang HC. Harnessing the Potential Synergistic Interplay Between Photosensitizer Dark Toxicity and Chemotherapy. Photochem Photobiol 2020; 96:636-645. [PMID: 31856423 DOI: 10.1111/php.13196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022]
Abstract
The combination of photodynamic therapy and taxol- or platinum-based chemotherapy (photochemotherapy) is an effective and promising cancer treatment. While the mechanisms of action of photochemotherapy are actively studied, relatively little is known about the cytotoxicity and molecular alterations induced by the combination of chemotherapy and photosensitizers without light activation in cancer cells. This study investigates the interplay between the photosensitizer benzoporphyrin derivative (BPD) without light activation and cisplatin or paclitaxel in two glioblastoma lines, U87 and U251. The combination effect of BPD and cisplatin in U87 cells is slightly synergistic (combination index, CI = 0.93), showing 1.8- to 2.6-fold lower half-maximal inhibitory concentrations (IC50 ) compared to those of individual drugs. In contrast, combining BPD and paclitaxel is slightly antagonistic (CI = 1.14) in U87 cells. In U251 cells, the combinations of BPD and cisplatin or paclitaxel are both antagonistic (CI = 1.24 and 1.34, respectively). Western blotting was performed to investigate changes in the expression levels of YAP, TAZ, Bcl-2 and EGFR in U87 and U251 cells treated with BPD, cisplatin and paclitaxel, both as monotherapies and in combination. Our study provides insights into the molecular alterations in two glioma lines caused by each monotherapy and the combinations, in order to inform the design of effective treatments.
Collapse
Affiliation(s)
- Yan Baglo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
| | - Aaron J Sorrin
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
| | - Barry J Liang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD.,Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
35
|
PABPC1-induced stabilization of BDNF-AS inhibits malignant progression of glioblastoma cells through STAU1-mediated decay. Cell Death Dis 2020; 11:81. [PMID: 32015336 PMCID: PMC6997171 DOI: 10.1038/s41419-020-2267-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/29/2022]
Abstract
Glioblastoma is the most common and malignant form of primary central nervous tumor in adults. Long noncoding RNAs (lncRNAs) have been reported to play a pivotal role in modulating gene expression and regulating human tumor’s malignant behaviors. In this study, we confirmed that lncRNA brain-derived neurotrophic factor antisense (BDNF-AS) was downregulated in glioblastoma tissues and cells, interacted and stabilized by polyadenylate-binding protein cytoplasmic 1 (PABPC1). Overexpression of BDNF-AS inhibited the proliferation, migration, and invasion, as well as induced the apoptosis of glioblastoma cells. In the in vivo study, PABPC1 overexpression combined with BDNF-AS overexpression produced the smallest tumor and the longest survival. Moreover, BDNF-AS could elicit retina and anterior neural fold homeobox 2 (RAX2) mRNA decay through STAU1-mediated decay (SMD), and thereby regulated the malignant behaviors glioblastoma cells. Knockdown of RAX2 produced tumor-suppressive function in glioblastoma cells and increased the expression of discs large homolog 5 (DLG5), leading to the activation of the Hippo pathway. In general, this study elucidated that the PABPC1-BDNF-AS-RAX2-DLG5 mechanism may contribute to the anticancer potential of glioma cells and may provide potential therapeutic targets for human glioma.
Collapse
|
36
|
Ji J, Ding K, Luo T, Xu R, Zhang X, Huang B, Chen A, Zhang D, Miletic H, Bjerkvig R, Thorsen F, Wang J, Li X. PMEPA1 isoform a drives progression of glioblastoma by promoting protein degradation of the Hippo pathway kinase LATS1. Oncogene 2020; 39:1125-1139. [PMID: 31605013 PMCID: PMC6989403 DOI: 10.1038/s41388-019-1050-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/20/2019] [Accepted: 09/26/2019] [Indexed: 11/08/2022]
Abstract
The Hippo signaling pathway controls organ development and is also known, in cancer, to have a tumor suppressing role. Within the Hippo pathway, we here demonstrate, in human gliomas, a functional interaction of a transmembrane protein, prostate transmembrane protein, androgen induced 1 (PMEPA1) with large tumor suppressor kinase 1 (LATS1). We show that PMEPA1 is upregulated in primary human gliomas. The PMEPA1 isoform PMEPA1a was predominantly expressed in glioma specimens and cell lines, and ectopic expression of the protein promoted glioma growth and invasion in vitro and in an orthotopic xenograft model in nude mice. In co-immunoprecipitation experiments, PMEPA1a associated with the Hippo tumor suppressor kinase LATS1. This interaction led to a proteasomal degradation of LATS1 through recruitment of the ubiquitin ligase, neural precursor cell expressed, developmentally downregulated 4 (NEDD4), which led to silencing of Hippo signaling. Alanine substitution in PMEPA1a at PY motifs resulted in failed LATS1 degradation. Targeting of a downstream component in the Hippo signaling pathway, YAP, with shRNA, interfered with the growth promoting activities of PMEPA1a in vitro and in vivo. In conclusion, the presented work shows that PMEPA1a contributes to glioma progression by a dysregulation of the Hippo signaling pathway and thus represents a promising target for the treatment of gliomas.
Collapse
Affiliation(s)
- Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Kaikai Ding
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Tao Luo
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Ran Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- School of Medicine, Shandong University, Jinan, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Hrvoje Miletic
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Jonas Lies vei 65, 5021, Bergen, Norway
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
- Department of Oncology, Luxembourg Institute of Health, 84, Val Fleuri, Luxembourg, L-1526, Luxembourg
| | - Frits Thorsen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
- The Molecular Imaging Center, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.
| |
Collapse
|
37
|
Xu X, Chen Y, Wang X, Mu X. Role of Hippo/YAP signaling in irradiation-induced glioma cell apoptosis. Cancer Manag Res 2019; 11:7577-7585. [PMID: 31496812 PMCID: PMC6693089 DOI: 10.2147/cmar.s210825] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/06/2019] [Indexed: 12/22/2022] Open
Abstract
Background Although Hippo/Yes-associated protein (YAP) signaling plays crucial roles in radiation sensitivity and resistance of multiple kinds of cancers, its role in the radiation sensitivity of glioma cells remains unclear. The present study aimed to reveal Hippo/YAP role in the radiation sensitivity of glioma cells. Methods Glioma U251 cells were administrated with different doses of irradiation. Cell Counting Kit-8 (CCK-8) and flow cytometry assays were used to assess cell viability and apoptosis. Co-immunoprecipitation (co-IP) assay was used to assess the interactions between proteins. Results The results showed that irradiation exposure significantly inhibited cell viability and induced cell apoptosis in a dose-dependent manner, as well as decreased YAP1 expression via enhancing RCHY1-mediated YAP1 protein degradation. In addition, we observed that downregulation of YAP1 or RCHY1 weakened the role of irradiation exposure in cell viability inhibition and apoptosis promotion. Conclusion Collectively, this study emphasizes the vital role of Hippo/YAP signaling in radiation sensitivity of glioma, that RCHY1-mediated YAP1 protein downregulation is a main mechanism accounting for radiation-induced glioma cell apoptosis. Our study may enrich the theoretical basis of Hippo/YAP signaling as a new target for improving radiation sensitivity in glioma.
Collapse
Affiliation(s)
- Xiaofei Xu
- Department of Radiology, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| | - Yan Chen
- Department of Neurosurgery, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| | - Xi Wang
- Department of Radiology, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| | - Xingguo Mu
- Department of Radiology, The Second Hospital of Jilin University, Chang Chun 130041, People's Republic of China
| |
Collapse
|
38
|
Wang J, Cai C, Nie D, Song X, Sun G, Zhi T, Li B, Qi J, Zhang J, Chen H, Shi Q, Yu R. FRK suppresses human glioma growth by inhibiting ITGB1/FAK signaling. Biochem Biophys Res Commun 2019; 517:588-595. [PMID: 31395336 DOI: 10.1016/j.bbrc.2019.07.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
Abstract
Fyn-related kinase (FRK), a member of the Src-related tyrosine kinase family, functions as a tumor suppressor in several malignancies. We previously showed that FRK overexpression inhibited the growth of glioma cells. However, it is unknown whether FRK is equally effective against intracranial glioma in vivo, and the mechanism by which FRK influences glioma cell growth remains unclear. In this study, we found that tumor volume was reduced by about one-third in mice with FRK overexpression, which showed improved survival relative to controls. Immunofluorescence analysis revealed that FRK overexpression inhibited glioma cell proliferation and induced their apoptosis. Importantly, in vitro we further found that FRK decreased the expression of integrin subunit β1 (ITGB1) at both the mRNA and protein levels. FRK also inhibited transactivation by ITGB1, resulting in the suppression of its target proteins AKT and focal adhesion kinase (FAK). ITGB1 overexpression promoted glioma cell growth and partially reduced FRK-induced growth suppression. These results indicate that FRK inhibits human glioma growth via regulating ITGB1/FAK signaling and provide a potential therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, PR China; Department of Neurosurgery, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, PR China
| | - Chang Cai
- Department of Neurosurgery, Suqian First Hospital, 120 Su Zhi Road, Suqian, 223800, Jiangsu, PR China
| | - Dekang Nie
- Department of Neurosurgery, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, PR China
| | - Xu Song
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, PR China
| | - Guan Sun
- Department of Neurosurgery, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, PR China
| | - Tongle Zhi
- Department of Neurosurgery, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, PR China
| | - Bing Li
- Department of Neurosurgery, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, PR China
| | - Juxing Qi
- Department of Neurosurgery, The First People's Hospital of Yancheng, The Forth Affiliated Hospital of Nantong University, Yancheng, 224006, Jiangsu, PR China
| | - Jianyong Zhang
- Department of Neurosurgery, Suqian First Hospital, 120 Su Zhi Road, Suqian, 223800, Jiangsu, PR China
| | - Honglin Chen
- Department of Neurosurgery, Suqian First Hospital, 120 Su Zhi Road, Suqian, 223800, Jiangsu, PR China
| | - Qiong Shi
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, PR China.
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, PR China; Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, PR China.
| |
Collapse
|
39
|
Ding K, Ji J, Zhang X, Huang B, Chen A, Zhang D, Li X, Wang X, Wang J. RNA splicing factor USP39 promotes glioma progression by inducing TAZ mRNA maturation. Oncogene 2019; 38:6414-6428. [PMID: 31332287 PMCID: PMC6756117 DOI: 10.1038/s41388-019-0888-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 06/06/2019] [Accepted: 07/03/2019] [Indexed: 01/01/2023]
Abstract
Increasing evidence demonstrates that ubiquitin specific protease 39 (USP39) plays an oncogenic role in various human tumors. Here, using expression analysis of the publicly available Oncomine database, clinical glioma patient samples, and glioma cells, we found that USP39 was overexpressed in human gliomas. Knockdown of USP39 in glioma cells demonstrated that the protein promoted cell growth, invasion and migration in vitro and in a tumor model in nude mice. To identify mediators of USP39 growth-promoting properties, we used luciferase reporter constructs under transcriptional control of various promoters specific to seven canonical cancer-associated pathways. Luciferase activity from a synthetic TEAD-dependent YAP/TAZ-responsive reporter, as a direct readout of the Hippo signaling pathway, was decreased by 92% in cells with USP39 knockdown, whereas the luciferase activities from the other six cancer pathways, including MAPK/ERK, MAPK/JNK, NFκB, Notch, TGFβ, and Wnt, remained unchanged. TAZ protein expression however was decreased independent of canonical Hippo signaling. Immunohistochemistry revealed a positive correlation between USP39 and TAZ proteins in orthotopic xenografts derived from modified glioma cells expressing USP39 shRNAs and primary human glioma samples (p < 0.05). Finally, loss of USP39 decreased TAZ pre-mRNA splicing efficiency in glioma cells in vitro, which led to reduced levels of TAZ protein. In summary, USP39 has oncogenic properties that increase TAZ protein levels by inducing maturation of its mRNA. USP39 therefore provides a novel therapeutic target for the treatment of human glioma.
Collapse
Affiliation(s)
- Kaikai Ding
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Jianxiong Ji
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China.,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China
| | - Xinyu Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China. .,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China.
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, 250012, Jinan, PR China. .,Shandong Key Laboratory of Brain Function Remodeling, 250012, Jinan, PR China. .,Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
| |
Collapse
|
40
|
Lu QR, Qian L, Zhou X. Developmental origins and oncogenic pathways in malignant brain tumors. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e342. [PMID: 30945456 DOI: 10.1002/wdev.342] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/20/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022]
Abstract
Brain tumors such as adult glioblastomas and pediatric high-grade gliomas or medulloblastomas are among the leading causes of cancer-related deaths, exhibiting poor prognoses with little improvement in outcomes in the past several decades. These tumors are heterogeneous and can be initiated from various neural cell types, contributing to therapy resistance. How such heterogeneity arises is linked to the tumor cell of origin and their genetic alterations. Brain tumorigenesis and progression recapitulate key features associated with normal neurogenesis; however, the underlying mechanisms are quite dysregulated as tumor cells grow and divide in an uncontrolled manner. Recent comprehensive genomic, transcriptomic, and epigenomic studies at single-cell resolution have shed new light onto diverse tumor-driving events, cellular heterogeneity, and cells of origin in different brain tumors. Primary and secondary glioblastomas develop through different genetic alterations and pathways, such as EGFR amplification and IDH1/2 or TP53 mutation, respectively. Mutations such as histone H3K27M impacting epigenetic modifications define a distinct group of pediatric high-grade gliomas such as diffuse intrinsic pontine glioma. The identification of distinct genetic, epigenomic profiles and cellular heterogeneity has led to new classifications of adult and pediatric brain tumor subtypes, affording insights into molecular and lineage-specific vulnerabilities for treatment stratification. This review discusses our current understanding of tumor cells of origin, heterogeneity, recurring genetic and epigenetic alterations, oncogenic drivers and signaling pathways for adult glioblastomas, pediatric high-grade gliomas, and medulloblastomas, the genetically heterogeneous groups of malignant brain tumors. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Signaling Pathways > Cell Fate Signaling.
Collapse
Affiliation(s)
- Q Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lily Qian
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xianyao Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China.,Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Eales KL, Wilkinson EA, Cruickshank G, Tucker JHR, Tennant DA. Verteporfin selectively kills hypoxic glioma cells through iron-binding and increased production of reactive oxygen species. Sci Rep 2018; 8:14358. [PMID: 30254296 PMCID: PMC6156578 DOI: 10.1038/s41598-018-32727-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022] Open
Abstract
Gliomas are highly malignant brain tumours characterised by extensive areas of poor perfusion which subsequently leads to hypoxia and reduced survival. Therapies that address the hypoxic microenvironment are likely to significantly improve patient outcomes. Verteporfin, a benzoporphyrin-like drug, has been suggested to target the Yes-associated protein (YAP). Increased YAP expression and transcriptional activity has been proposed in other tumour types to promote malignant cell survival and thus YAP-inhibitor, verteporfin, may be predicted to impact glioma cell growth and viability. Due to the extensive hypoxic nature of gliomas, we investigated the effect of hypoxia on YAP expression and found that YAP transcription is increased under these conditions. Treatment of both primary and immortalised glioblastoma cell lines with verteporfin resulted in a significant decrease in viability but strikingly only under hypoxic conditions (1% O2). We discovered that cell death occurs through a YAP-independent mechanism, predominately involving binding of free iron and likely through redox cycling, contributes to production of reactive oxygen species. This results in disruption of normal cellular processes and death in cells already under oxidative stress - such as those in hypoxia. We suggest that through repurposing verteporfin, it represents a novel means of treating highly therapy-resistant, hypoxic cells in glioma.
Collapse
Affiliation(s)
- Katherine L Eales
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Edward A Wilkinson
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Garth Cruickshank
- Department of Neurosurgery, University Hospitals Birmingham, NHS Foundation Trust, Birmingham, UK
| | - James H R Tucker
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
42
|
Guichet PO, Masliantsev K, Tachon G, Petropoulos C, Godet J, Larrieu D, Milin S, Wager M, Karayan-Tapon L. Fatal correlation between YAP1 expression and glioma aggressiveness: clinical and molecular evidence. J Pathol 2018; 246:205-216. [PMID: 30009411 DOI: 10.1002/path.5133] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/21/2018] [Accepted: 06/28/2018] [Indexed: 12/23/2022]
Abstract
During the last decade, large-scale genomic analyses have clarified the somatic alterations in gliomas, providing new molecular classification based on IDH1/2 mutations and 1p19q codeletion with more accurate patient prognostication. The Hippo pathway downstream effectors, YAP1 and TAZ, have recently emerged as major determinants of malignancy by inducing proliferation, chemoresistance, and metastasis in solid tumors. In this study, we investigated the expression of YAP1 in 117 clinical samples of glioma described according to the WHO 2016 classification. We showed for the first time that YAP1 was tightly associated with glioma molecular subtypes and patient outcome. We validated our results in an independent cohort from the TCGA database. More interestingly, we found that YAP1 may have prognostic significance for predicting patient survival, especially in low-grade gliomas. Using patient-derived glioblastoma stem cell cultures, we demonstrated that YAP1 was activated and that it controlled cell proliferation. Transcriptome analysis revealed lower expression of YAP1 in the proneural GBM subtype. Furthermore, we found that overexpression of YAP1 was sufficient to inhibit the OLIG2 proneural marker, suggesting its involvement in maintenance of the GBM phenotype. Taken together, our results showed that YAP1 could be a relevant prognostic biomarker and a potential therapeutic target in glioma. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Pierre-Olivier Guichet
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.,Université de Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, France
| | - Konstantin Masliantsev
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.,Université de Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, France
| | - Gaëlle Tachon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.,Université de Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, France
| | - Christos Petropoulos
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.,Université de Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, France
| | - Julie Godet
- CHU de Poitiers, Service d'Anatomo-Cytopathologie, Poitiers, France
| | - Delphine Larrieu
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, France
| | - Serge Milin
- CHU de Poitiers, Service d'Anatomo-Cytopathologie, Poitiers, France
| | - Michel Wager
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.,Université de Poitiers, France.,CHU de Poitiers, Service de Neurochirurgie, Poitiers, France
| | - Lucie Karayan-Tapon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, Poitiers, France.,Université de Poitiers, France.,CHU de Poitiers, Laboratoire de Cancérologie Biologique, Poitiers, France
| |
Collapse
|
43
|
Zhou X, Xie S, Wu S, Qi Y, Wang Z, Zhang H, Lu D, Wang X, Dong Y, Liu G, Yang D, Shi Q, Bian W, Yu R. Golgi phosphoprotein 3 promotes glioma progression via inhibiting Rab5-mediated endocytosis and degradation of epidermal growth factor receptor. Neuro Oncol 2018; 19:1628-1639. [PMID: 28575494 DOI: 10.1093/neuonc/nox104] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Golgi phosphoprotein 3 (GOLPH3) is associated with worse prognosis of gliomas, but its role and mechanism in glioma progression remain largely unknown. This study aimed to explore the role and mechanism of GOLPH3 in glioma progression. Methods The expression of GOLPH3 in glioma tissues was detected by quantitative PCR, immunoblotting, and immunohistochemistry. GOLPH3's effect on glioma progression was examined using cell growth assays and an intracranial glioma model. The effect of GOLPH3 on epidermal growth factor receptor (EGFR) stability, endocytosis, and degradation was examined by immunoblotting and immunofluorescence. The activity of Rab5 was checked by glutathione S-transferase pulldown assay. Results GOLPH3 was upregulated in gliomas, and its downregulation inhibited glioma cell proliferation both in vitro and in vivo. Furthermore, GOLPH3 depletion dampened EGFR signaling by enhancing EGFR endocytosis, driving EGFR into late endosome and promoting lysosome-mediated degradation. Interestingly, GOLPH3 bound to Rab5 and GOLPH3 downregulation promoted the activation of Rab5. In addition, Rab5 depletion abolished the effect of GOLPH3 on EGFR endocytosis and degradation. Conclusion Our results imply that GOLPH3 promotes glioma cell proliferation via inhibiting Rab5-mediated endocytosis and degradation of EGFR, thereby activating the phosphatidylinositol-3 kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling pathway. We find a new mechanism by which GOLPH3 promotes tumor progression through regulating cell surface receptor trafficking. Extensive and intensive understanding of the role of GOLPH3 in glioma progression may provide an opportunity to develop a novel molecular therapeutic target for gliomas.
Collapse
Affiliation(s)
- Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shao Xie
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shishuang Wu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanhua Qi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhaohao Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hao Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dong Lu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Dong
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guanzheng Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongxu Yang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiong Shi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenbin Bian
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
44
|
Chen S, Fang Y, Xu S, Reis C, Zhang J. Mammalian Sterile20-like Kinases: Signalings and Roles in Central Nervous System. Aging Dis 2018; 9:537-552. [PMID: 29896440 PMCID: PMC5988607 DOI: 10.14336/ad.2017.0702] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/02/2017] [Indexed: 12/25/2022] Open
Abstract
Mammalian Sterile20-like (MST) kinases are located upstream in the mitogen-activated protein kinase pathway, and play an important role in cell proliferation, differentiation, renewal, polarization and migration. Generally, five MST kinases exist in mammalian signal transduction pathways, including MST1, MST2, MST3, MST4 and YSK1. The central nervous system (CNS) is a sophisticated entity that takes charge of information reception, integration and response. Recently, accumulating evidence proposes that MST kinases are critical in the development of disease in different systems involving the CNS. In this review, we summarized the signal transduction pathways and interacting proteins of MST kinases. The potential biological function of each MST kinase and the commonly reported MST-related diseases in the neural system are also reviewed. Further investigation of MST kinases and their interaction with CNS diseases would provide the medical community with new therapeutic targets for human diseases.
Collapse
Affiliation(s)
- Sheng Chen
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuanjian Fang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shenbin Xu
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cesar Reis
- 2Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA.,3Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- 1Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,4Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
45
|
Ji J, Xu R, Zhang X, Han M, Xu Y, Wei Y, Ding K, Wang S, Bin Huang, Chen A, Di Zhang, Jiang Z, Xu S, Zhang Q, Li W, Ni S, Wang J, Li X. Actin like-6A promotes glioma progression through stabilization of transcriptional regulators YAP/TAZ. Cell Death Dis 2018; 9:517. [PMID: 29725063 PMCID: PMC5938705 DOI: 10.1038/s41419-018-0548-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/20/2022]
Abstract
Increased Actin-like 6A (ACTL6A) expression has been implicated in the development of diverse cancers and recently associated with the Hippo signaling pathway, which is known to regulate biological properties, including proliferation, tissue regeneration, stem cell biology, as well as tumorigenesis. Here we first show that ACTL6A is upregulated in human gliomas and its expression is associated with glioma patient survival. ACTL6A promotes malignant behaviors of glioma cells in vitro and in orthotopic xenograft model. In co-immunoprecipitation assays, we discover that ACTL6A physically associated with YAP/TAZ and furthermore disrupts the interaction between YAP and β-TrCP E3 ubiquitin ligase, which promotes YAP protein degradation. Moreover, effects of ACTL6A on glioma cells proliferation, migration, and invasion could be mediated by YAP/TAZ. These data indicate that ACTL6A may contribute to cancer progression by stabilizing YAP/TAZ and therefore provide a novel therapeutic target for the treatment of human gliomas.
Collapse
MESH Headings
- Actins/antagonists & inhibitors
- Actins/genetics
- Actins/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Aged
- Animals
- Brain Neoplasms/genetics
- Brain Neoplasms/metabolism
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Chromosomal Proteins, Non-Histone/antagonists & inhibitors
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- DNA-Binding Proteins/antagonists & inhibitors
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Progression
- Female
- Gene Expression Regulation, Neoplastic
- Glioblastoma/genetics
- Glioblastoma/metabolism
- Glioblastoma/mortality
- Glioblastoma/pathology
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Nude
- Middle Aged
- Neoplasm Invasiveness
- Phosphoproteins/genetics
- Phosphoproteins/metabolism
- Protein Binding
- Protein Stability
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Survival Analysis
- Trans-Activators
- Transcription Factors
- Transcriptional Coactivator with PDZ-Binding Motif Proteins
- Tumor Burden
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
- beta-Transducin Repeat-Containing Proteins/genetics
- beta-Transducin Repeat-Containing Proteins/metabolism
Collapse
Affiliation(s)
- Jianxiong Ji
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Ran Xu
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
- Brain and Mind Centre, and Faculty of Health Sciences, University of Sydney, Camperdown, NSW 2050, Australia
| | - Xin Zhang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Mingzhi Han
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yangyang Xu
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yuzhen Wei
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
- Department of Neurosurgery, Jining No.1 People's Hospital, Jiankang Road, Jining, 272011, China
| | - Kaikai Ding
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shuai Wang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Bin Huang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Anjing Chen
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Di Zhang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Zheng Jiang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shuo Xu
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Qing Zhang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Wenjie Li
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Shilei Ni
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Jian Wang
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China.
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen, 5009, Norway.
| | - Xingang Li
- Key Laboratory of Brain Functional Remodeling, Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
46
|
Yang C, Tan J, Zhu J, Wang S, Wei G. YAP promotes tumorigenesis and cisplatin resistance in neuroblastoma. Oncotarget 2018; 8:37154-37163. [PMID: 28415761 PMCID: PMC5514898 DOI: 10.18632/oncotarget.16209] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/06/2017] [Indexed: 02/06/2023] Open
Abstract
The transcriptional co-activator Yes-associated protein (YAP) is essential for Hippo pathway-driven tumorigenesis in various cancers. However, the expression and function of YAP in neuroblastoma remains elusive. Here, we show that YAP was highly expressed in Neuroblastoma (NB) and expression levels correlated with advanced tumor staging. Knockdown of YAP significantly impaired neuroblastoma proliferation, tumorigenesis, and invasion in vitro. Injection of the YAP inhibitor, Peptide 17, dramatically prevented neuroblastoma subcutaneous tumor growth by efficiently downregulating YAP expression in tumors. Additionally, less proliferative and more apoptotic cells were found in the Peptide 17 treatment group. Furthermore, YAP inhibition significantly inhibited cisplatin-resistant neuroblastoma proliferation, tumorigenesis, and invasion in vitro. The combination of Peptide 17 with low-dose cisplatin efficiently impaired cisplatin-resistant NB subcutaneous tumor growth, being as effective as high-dose cisplatin. Notably, the combination therapy caused lesser liver toxicity in mice compared to the high-dose cisplatin treatment group. Collectively, this work identifies YAP as a novel regulator of neuroblastoma proliferation, tumorigenesis, and invasion and indicates that YAP is a potential therapeutic target for cisplatin-resistant neuroblastoma.
Collapse
Affiliation(s)
- Chao Yang
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Juan Tan
- Clinical Department of Children's Hospital of Chongqing Medical University, Lijia Campus, Chongqing, China
| | - Jun Zhu
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Department of Pathology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Shan Wang
- Department of Pediatric Surgical Oncology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Guanghui Wei
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Department of Urology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| |
Collapse
|
47
|
Zhang Y, Xie P, Wang X, Pan P, Wang Y, Zhang H, Dong Y, Shi Y, Jiang Y, Yu R, Zhou X. YAP Promotes Migration and Invasion of Human Glioma Cells. J Mol Neurosci 2018; 64:262-272. [PMID: 29306996 DOI: 10.1007/s12031-017-1018-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Previously, we have reported that Yes-associated protein (YAP) is upregulated in human glioma tissues and its level is positively correlated with patient prognosis. However, the role and mechanism of YAP in the highly invasive nature of human gliomas were largely unknown. In this study, examined by wound healing assay, transwell assay, or live-imaging, we found that YAP downregulation inhibited glioma cell migration and invasion, while YAP over-expression promoted them. Interestingly, the above effect of YAP on immortalized glioma cells was recapitulated in cultured primary glioma cells. In addition, the protein level of N-cadherin and Twist, two important proteins involved in tumor invasion, increased after YAP over-expression. Meanwhile, YAP over-expression significantly increased the F-actin level and changed the distribution of F-actin, leading to cytoskeletal reorganization, which plays an important role in cell motility. Furthermore, the promotion effect of YAP over-expression on glioma cell migration and invasion was partially abolished by Twist downregulation. Taken together, our findings show that YAP contributes to glioma cell migration and invasion by regulating N-cadherin and Twist, as well as cytoskeletal reorganization.
Collapse
Affiliation(s)
- Yu Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng Xie
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Neurosurgery, Huaian Second People's Hospital, Huaian, 223002, China
| | - Xu Wang
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Peng Pan
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan Wang
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hao Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Dong
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yi Shi
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Jiang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Xiuping Zhou
- Brain Hospital, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
48
|
Wang Y, Pan P, Wang Z, Zhang Y, Xie P, Geng D, Jiang Y, Yu R, Zhou X. β-catenin-mediated YAP signaling promotes human glioma growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:136. [PMID: 28962630 PMCID: PMC5622484 DOI: 10.1186/s13046-017-0606-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 09/22/2017] [Indexed: 02/07/2023]
Abstract
Background Hippo/YAP pathway is known to be important for development, growth and organogenesis, and dysregulation of this pathway leads to tumor progression.We and others find that YAP is up-regulated in human gliomas and associated with worse prognosis of patients. However, the role and mechanism of YAP in glioma progression is largely unknown. Methods The expression of YAP in glioma tissues was detected by quantitative polymerase chain reaction (qPCR) and immunoblotting. The effect of YAP on glioma progression was examined using cell growth assays and intracranial glioma model. The effect of YAP on β-catenin protein level, subcellular location and transcription activity was examined by immunoblotting, immunofluorescence and RT-PCR. Results Firstly, knockdown of YAP inhibited glioma cell proliferation in vitro and tumor growth in vivo. In addition, YAP modulated the protein level, subcellular location and transcription activity of β-catenin via regulating the activity of GSK3β. Lastly, β-catenin partially mediated the effect of YAP on glioma cell proliferation. Conclusion Our findings identify that YAP promotes human glioma growth through enhancing Wnt/β-catenin signaling. In addition, this study provides a new crosstalk mechanism between Hippo/YAP and Wnt/β-catenin pathways, which suggests a new strategy for human glioma treatment. Electronic supplementary material The online version of this article (10.1186/s13046-017-0606-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, Jiangsu, 221002, People's Republic of China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng Pan
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Present address: Department of Neurosurgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu, China
| | - Zhaohao Wang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Peng Xie
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Decheng Geng
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Jiang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, Jiangsu, 221002, People's Republic of China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiuping Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-hai Road, Xuzhou, Jiangsu, 221002, People's Republic of China. .,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
49
|
Keck M, Fournier A, Gualtieri F, Walker A, von Rüden EL, Russmann V, Deeg CA, Hauck SM, Krause R, Potschka H. A systems level analysis of epileptogenesis-associated proteome alterations. Neurobiol Dis 2017; 105:164-178. [PMID: 28576708 DOI: 10.1016/j.nbd.2017.05.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 05/22/2017] [Accepted: 05/29/2017] [Indexed: 12/18/2022] Open
Abstract
Despite intense research efforts, the knowledge about the mechanisms of epileptogenesis and epilepsy is still considered incomplete and limited. However, an in-depth understanding of molecular pathophysiological processes is crucial for the rational selection of innovative biomarkers and target candidates. Here, we subjected proteomic data from different phases of a chronic rat epileptogenesis model to a comprehensive systems level analysis. Weighted Gene Co-expression Network analysis identified several modules of interconnected protein groups reflecting distinct molecular aspects of epileptogenesis in the hippocampus and the parahippocampal cortex. Characterization of these modules did not only further validate the data but also revealed regulation of molecular processes not described previously in the context of epilepsy development. The data sets also provide valuable information about temporal patterns, which should be taken into account for development of preventive strategies in particular when it comes to multi-targeting network pharmacology approaches. In addition, principal component analysis suggests candidate biomarkers, which might inform the design of novel molecular imaging approaches aiming to predict epileptogenesis during different phases or confirm epilepsy manifestation. Further studies are necessary to distinguish between molecular alterations, which correlate with epileptogenesis versus those reflecting a mere consequence of the status epilepticus.
Collapse
Affiliation(s)
- Michael Keck
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Anna Fournier
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4367 Belvaux, Luxembourg
| | - Fabio Gualtieri
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Andreas Walker
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Eva-Lotta von Rüden
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Vera Russmann
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany
| | - Cornelia A Deeg
- Institute of Animal Physiology, Department of Veterinary Sciences, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany; Experimental Ophthalmology, Philipps University of Marburg, 35037 Marburg, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Roland Krause
- Bioinformatics Core, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4367 Belvaux, Luxembourg.
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University (LMU), 80539 Munich, Germany.
| |
Collapse
|
50
|
Rybarczyk A, Klacz J, Wronska A, Matuszewski M, Kmiec Z, Wierzbicki PM. Overexpression of the YAP1 oncogene in clear cell renal cell carcinoma is associated with poor outcome. Oncol Rep 2017; 38:427-439. [DOI: 10.3892/or.2017.5642] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/27/2017] [Indexed: 11/05/2022] Open
|