1
|
Yakubov R, Kaloti R, Persaud P, McCracken A, Zadeh G, Bunda S. It's all downstream from here: RTK/Raf/MEK/ERK pathway resistance mechanisms in glioblastoma. J Neurooncol 2025; 172:327-345. [PMID: 39821893 PMCID: PMC11937199 DOI: 10.1007/s11060-024-04930-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND The receptor tyrosine kinase (RTK)/Ras/Raf/MEK/ERK signaling pathway is one of the most tumorigenic pathways in cancer, with its hyperactivation strongly linked to the aggressive nature of glioblastoma (GBM). Although extensive research has focused on developing therapeutics targeting this pathway, clinical success remains elusive due to the emergence of resistance mechanisms. OBJECTIVE This review investigates how inhibition of the RTK/Ras/Raf/MEK/ERK pathway alters transcription factors, contributing to acquired resistance mechanisms in GBM. It also highlights the critical role of transcription factor dysregulation in therapeutic resistance. METHODS & RESULTS Findings from key studies on the RTK/Ras/Raf/MEK/ERK pathway in GBM were synthesized to explore the role of transcription factor dysregulation in resistance to targeted therapies, radiation, and chemotherapy. The review highlights that transcription factors undergo significant dysregulation following RTK/Ras/Raf/MEK/ERK pathway inhibition, contributing to therapeutic resistance. CONCLUSION Transcription factors are promising targets for overcoming treatment resistance in GBM, with cotreatment strategies combining RTK/Ras/Raf/MEK/ERK pathway inhibitors and transcription factor-targeted therapies presenting a novel approach. Despite the challenges of targeting complex structures and interactions, advancements in drug development and precision technologies hold great potential. Continued research is essential to refine these strategies and improve outcomes for GBM and other aggressive cancers.
Collapse
Affiliation(s)
- Rebeca Yakubov
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ramneet Kaloti
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Phooja Persaud
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Anna McCracken
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Gelareh Zadeh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada.
| | - Severa Bunda
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
2
|
Mair MJ, Leibetseder A, Heller G, Tomasich E, Müller L, Busse I, Wöhrer A, Kiesel B, Widhalm G, Eckert F, Weis S, Pichler J, Preusser M, Berghoff AS. Clinical characteristics, molecular reclassification trajectories and DNA methylation patterns of long- and short-term survivors of WHO grade II and III glioma. J Neurol 2025; 272:210. [PMID: 39954095 PMCID: PMC11829921 DOI: 10.1007/s00415-025-12923-6] [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: 12/02/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE The prognosis of diffuse gliomas previously classified as "lower-grade" is heterogeneous and complicates clinical decisions. We aimed to investigate the molecular profile of clinical outliers to gain insight into biological drivers of long and short-term survivors. METHODS Here, patients aged ≥ 18 years and diagnosed with diffuse glioma, WHO grade II/2 or III/3 were included. Short-term survivors (STS) were defined as overall survival (OS) < 1 years, and long-term survivors (LTS) as OS > 10 years. DNA methylation profiling was performed using the Illumina EPIC 850k platform. RESULTS In total, 385 patients (294 LTS, 91 STS) were included. Median overall survival was 234 months (95%CI: 207-248) in LTS and 7.3 months (95%CI: 6.4-8.1) in STS. Compared to STS, LTS were younger, had higher Karnofsky Performance Status, more extensive resections, and lower symptomatic burden (p < 0.001, respectively). Molecular reclassification showed IDH-mutant gliomas in 240/246 (95.5%) LTS and 10/79 (12.7%) STS. Initial diagnosis (tumor type and/or grading) changed in 69/325 (21.2%) patients based on reclassification according to WHO 2016 and in 45/258 (17.4%) as per WHO 2021. DNA methylation analysis indicated two clusters, one with mainly STS (39/41, 95.1%) and heterogeneous IDH-wildtype tumors (cluster A) and one with mainly LTS (82/106, 77.4%) and IDH-mutant tumors (cluster B). Functional enrichment analysis of rare subtypes indicated altered Hippo/Notch and synaptic/neurotransmitter signaling pathway members. CONCLUSION LTS and STS show distinct clinical and molecular features, underscoring the importance of extended molecular workup for diagnosis. Further characterization of rare subtypes is needed to optimize treatment strategies and clinical trial planning.
Collapse
Affiliation(s)
- Maximilian J Mair
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Annette Leibetseder
- Department of Neurology 1, Neuromed Campus, Kepler University Hospital, Johannes Kepler University Linz, and Clinical Research Institute for Neuroscience, Linz, Austria
| | - Gerwin Heller
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Erwin Tomasich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Lisa Müller
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Ilka Busse
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Adelheid Wöhrer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Barbara Kiesel
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Franziska Eckert
- Department of Radiation Oncology, Medical University of Vienna, Vienna, Austria
| | - Serge Weis
- Division of Neuropathology, Department of Pathology and Molecular Pathology, Neuromed Campus, Kepler University Hospital, and Clinical Research Institute for Neuroscience, Johannes Kepler University Linz, Linz, Austria
| | - Josef Pichler
- Department of Internal Medicine and Neurooncology, Neuromed Campus, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, Austria.
| |
Collapse
|
3
|
Turpo-Peqqueña AG, Luna-Prado S, Valencia-Arce RJ, Del-Carpio-Carrazco FL, Gómez B. A Theoretical Study on the Efficacy and Mechanism of Combined YAP-1 and PARP-1 Inhibitors in the Treatment of Glioblastoma Multiforme Using Peruvian Maca Lepidium meyenii. Curr Issues Mol Biol 2025; 47:40. [PMID: 39852155 PMCID: PMC11763394 DOI: 10.3390/cimb47010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/25/2024] [Accepted: 01/05/2025] [Indexed: 01/26/2025] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive and treatment-resistant forms of brain cancer. Current therapeutic strategies, including surgery, chemotherapy, and radiotherapy, often fail due to the tumor's ability to develop resistance. The proteins YAP-1 (Yes-associated protein 1) and PARP-1 (Poly-(ADP-ribose)-polymerase-1) have been implicated in this resistance, playing crucial roles in cell proliferation and DNA repair mechanisms, respectively. This study explored the inhibitory potential of natural compounds from Lepidium meyenii (Peruvian Maca) on the YAP-1 and PARP-1 protein systems to develop novel therapeutic strategies for GBM. By molecular dynamics simulations, we identified N-(3-Methoxybenzyl)-(9Z,12Z,15Z)- octadecatrienamide (DK5) as the most promising natural inhibitor for PARP-1 and stearic acid (GK4) for YAP-1. Although synthetic inhibitors, such as Olaparib (ODK) for PARP-1 and Verteporfin (VER) for YAP-1, only VER was superior to the naturally occurring molecule and proved a promising alternative. In conclusion, natural compounds from Lepidium meyenii (Peruvian Maca) offer a potentially innovative approach to improve GBM treatment, complementing existing therapies with their inhibitory action on PARP-1 and YAP-1.
Collapse
Affiliation(s)
- Albert Gabriel Turpo-Peqqueña
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Medicina Humana, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
- Facultad de Biología, Universidad Nacional de San Agustín, Av. Alcides Carrión s/n, Arequipa 04001, Peru
| | - Sebastian Luna-Prado
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| | - Renato Javier Valencia-Arce
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| | - Fabio Leonardo Del-Carpio-Carrazco
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| | - Badhin Gómez
- Centro de Investigación en Ingeniería Molecular–CIIM, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru; (A.G.T.-P.); (S.L.-P.); (R.J.V.-A.); (F.L.D.-C.-C.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Urb. San José s/n, Umacollo, Arequipa 04013, Peru
| |
Collapse
|
4
|
Wang X, Guo Y, Lin P, Yu M, Song S, Xu W, Kong D, Wang Y, Zhang Y, Lu F, Xie Q, Ma X. Nuclear receptor E75/NR1D2 promotes tumor malignant transformation by integrating Hippo and Notch pathways. EMBO J 2024; 43:6336-6363. [PMID: 39516282 PMCID: PMC11649922 DOI: 10.1038/s44318-024-00290-3] [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: 02/21/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Hormone therapy resistance and the ensuing aggressive tumor progression present a significant clinical challenge. However, the mechanisms underlying the induction of tumor malignancy upon inhibition of steroid hormone signaling remain poorly understood. Here, we demonstrate that Drosophila malignant epithelial tumors show a similar reduction in ecdysone signaling, the main steroid hormone pathway. Our analysis of ecdysone-induced downstream targets reveals that overexpression of the nuclear receptor E75, particularly facilitates the malignant transformation of benign tumors. Genome-wide DNA binding profiles and biochemistry data reveal that E75 not only binds to the transcription factors of both Hippo and Notch pathways, but also exhibits widespread co-binding to their target genes, thus contributing to tumor malignancy. We further validated these findings by demonstrating that depletion of NR1D2, the mammalian homolog of E75, inhibits the activation of Hippo and Notch target genes, impeding glioblastoma progression. Together, our study unveils a novel mechanism by which hormone inhibition promotes tumor malignancy, and describes an evolutionarily conserved role of the oncogene E75/NR1D2 in integration of Hippo and Notch pathway activity during tumor progression.
Collapse
Affiliation(s)
- Xianping Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Yifan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Peng Lin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sha Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Du Kong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Yin Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Yanxiao Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Fei Lu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Qi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
5
|
Kim D, Olson JM, Cooper JA. N-cadherin dynamically regulates pediatric glioma cell migration in complex environments. J Cell Biol 2024; 223:e202401057. [PMID: 38477830 PMCID: PMC10937189 DOI: 10.1083/jcb.202401057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that intercellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.
Collapse
Affiliation(s)
- Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - James M. Olson
- Clinical Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jonathan A. Cooper
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
6
|
Kim D, Olson JM, Cooper JA. N-cadherin dynamically regulates pediatric glioma cell migration in complex environments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.04.535599. [PMID: 38260559 PMCID: PMC10802396 DOI: 10.1101/2023.04.04.535599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Pediatric high-grade gliomas are highly invasive and essentially incurable. Glioma cells migrate between neurons and glia, along axon tracts, and through extracellular matrix surrounding blood vessels and underlying the pia. Mechanisms that allow adaptation to such complex environments are poorly understood. N-cadherin is highly expressed in pediatric gliomas and associated with shorter survival. We found that inter-cellular homotypic N-cadherin interactions differentially regulate glioma migration according to the microenvironment, stimulating migration on cultured neurons or astrocytes but inhibiting invasion into reconstituted or astrocyte-deposited extracellular matrix. N-cadherin localizes to filamentous connections between migrating leader cells but to epithelial-like junctions between followers. Leader cells have more surface and recycling N-cadherin, increased YAP1/TAZ signaling, and increased proliferation relative to followers. YAP1/TAZ signaling is dynamically regulated as leaders and followers change position, leading to altered N-cadherin levels and organization. Together, the results suggest that pediatric glioma cells adapt to different microenvironments by regulating N-cadherin dynamics and cell-cell contacts.
Collapse
Affiliation(s)
- Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James M Olson
- Clinical Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, 98101, USA
| | - Jonathan A Cooper
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| |
Collapse
|
7
|
Wang C, Zhang Y, Jiang Q, Chen S, Zhang L, Qiu H. Oridonin suppresses the growth of glioblastoma cells via inhibiting Hippo/YAP axis. Arch Biochem Biophys 2024; 751:109845. [PMID: 38043888 DOI: 10.1016/j.abb.2023.109845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/13/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Glioma is a brain tumor that originates from brain or spine glial cells. Despite alternative treatments, the overall survival rate remains low. Oridonin (ORI) is purified from the Chinese herb Rabdosia rubescens, which has exhibited positive effects on tumors. This study aimed to investigate the effect of ORI on U87MG glioblastoma cells and whether the Hippo/YAP-related signaling pathway was involved. Malignant glioblastoma U87MG cells and male athymic nude mice (BALB/cnu/nu) were used as the experimental models. The YAP inhibitor Verteporfin (VP) and the overexpression of YAP were used to investigate its potential relation with glioma. Here, we found that ORI inhibited cell proliferation and promoted cell apoptosis in a dose-dependent manner in U87MG cells. Moreover, ORI inhibited Bcl-2, YAP, and c-Myc protein expression but increased Bax, caspase-3, and p-YAP protein expression. Furthermore, the effect of ORI was also confirmed in a mouse model bearing glioma. ORI reversed the effect of overexpression of YAP. Collectively, oridonin suppressed glioblastoma oncogenesis via the Hippo/YAP signaling pathway and could be a potential therapeutic target in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Chen Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China; Department of Clinical Pharmacy, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, 400016, China
| | - Yonghong Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
| | - Qingsong Jiang
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
| | - Shuang Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
| | - Liang Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China
| | - Hongmei Qiu
- Department of Pharmacology, College of Pharmacy, Chongqing Key Lab of Biochemistry and Molecular Pharmacology, Chongqing Key Lab Drug Metabolism, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
8
|
Osama M, Essibayi MA, Osama M, Ibrahim IA, Nasr Mostafa M, Şakir Ekşi M. The impact of interaction between verteporfin and yes-associated protein 1/transcriptional coactivator with PDZ-binding motif-TEA domain pathway on the progression of isocitrate dehydrogenase wild-type glioblastoma. J Cent Nerv Syst Dis 2023; 15:11795735231195760. [PMID: 37600236 PMCID: PMC10439684 DOI: 10.1177/11795735231195760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Verteporfin and 5-ALA are used for visualizing malignant tissue components in different body tumors and as photodynamic therapy in treating isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM). Additionally, verteporfin interferes with Yes-associated protein 1 (YAP)/Transcriptional coactivator with PDZ-binding motif - TEA domain (TAZ-TEAD) pathway, thus inhibiting the downstream effect of these oncogenes and reducing the malignant properties of GBM. Animal studies have shown verteporfin to be successful in increasing survival rates, which have led to the conduction of phase 1 and 2 clinical trials to further investigate its efficacy in treating GBM. In this article, we aimed to review the novel mechanism of verteporfin's action, the impact of its interaction with YAP/TAZ-TEAD, its effect on glioblastoma stem cells, and its role in inducing ferroptosis.
Collapse
Affiliation(s)
- Mahmoud Osama
- Department of Neurosurgery, Nasser Institute for Research and Treatment, Cairo, Egypt
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Muhammed Amir Essibayi
- Department of Neurosurgery, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Mona Osama
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ismail A. Ibrahim
- Department of Physical Therapy and Rehabilitation, Fenerbahce University, Istanbul, Turkey
| | | | - Murat Şakir Ekşi
- Neurosurgery Clinic, FSM Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
9
|
Miao N, Wang ZQ, Zhang N, Ma ZP, Su LP, Zhai YY, Hu YR, Sang W, Zhang W. Overexpression of ZEB1 and YAP1 is related to poor prognosis in patients with gliomas with different IDH1 status. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2023; 16:138-149. [PMID: 37559682 PMCID: PMC10408435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 06/02/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE Whether there is a correlation between zinc-finger E-box-binding homolog 1 (ZEB1) and Yes-associated protein 1 (YAP1) with clinical outcome in gliomas remains unclear. Hence, this study aimed to investigate the effects of ZEB1 and YAP1 on the prognosis of human gliomas and its relationship with the isocitrate dehydrogenase 1 (IDH1) gene state. METHODS Immunohistochemical staining was used to record the expression levels of ZEB1, YAP1, and p-YAP1 in 122 cases of low-grade glioma (LGGs) and 69 cases of glioblastoma (GBMs). The correlations of ZEB1 and YAP1 with pathological data were determined by Pearson's Chi-square test. Spearman correlation analysis was then used for analyzing the relationship among YAP1, ZEB1, and IDH1 gene status. The effects of ZEB1 and YAP1 on prognosis were investigated through survival analysis. RESULTS We detected high ZEB1 expression levels in 29 LGGs (23.8%) and 39 GBMs (56.5%), and high YAP1 expression levels in 22 LGGs (18.0%) and 44 of GBM (63.8%). These results revealed that the protein expression levels of ZEB1 and YAP1 were higher in GBM (P < 0.001). There was a significantly positive correlation between ZEB1 and YAP1 (P < 0.001; r = 0.533). High ZEB1 expression was related to tumor grade (P < 0.001) and Ki-67 (P = 0.0037). YAP1 overexpression was correlated with Ki-67 (P < 0.001), P53 (P = 0.009), tumor grade (P < 0.001), and tumor location (P = 0.018). Patients with ZEB1 and YAP1 high expression had worse overall survival (OS) (P < 0.001). The multivariate analysis showed that YAP1 was an independent prognostic factor for OS. In the LGG group, worse OS were observed in glioma patients with elevated YAP1 expression level. Spearman correlation analysis revealed no association between ZEB1 expression and IDH1 state (P = 0.360; r = -0.084), and YAP1 expression had a negative correlation with IDH1 mutation (P < 0.001, r = -0.364). CONCLUSIONS Our study showed that ZEB1 and YAP1 were significantly activated in GBM, and patients with high ZEB1 and YAP1 expression had worse OS. ZEB1 expression was significantly correlated with YAP1 in glioma. ZEB1 and YAP1 coexpression may serve as a useful prognostic biomarker for glioma, and aberrant YAP1 expression may be associated with IDH1 gene state.
Collapse
Affiliation(s)
- Na Miao
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| | - Zhi-Qiang Wang
- The Second Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
| | - Ning Zhang
- Surgery Department of Urology, The Third People’s Hospital of Xinjiang Uygur Autonomous RegionUrumqi 830054, Xinjiang, P. R. China
| | - Zhi-Ping Ma
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| | - Li-Ping Su
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| | - Yang-Yang Zhai
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| | - Yan-Ran Hu
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| | - Wei Sang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| | - Wei Zhang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi 830054, Xinjiang, P. R. China
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central AsiaUrumqi 830054, Xinjiang, P. R. China
| |
Collapse
|
10
|
Lin WH, Feathers RW, Cooper LM, Lewis-Tuffin LJ, Chen J, Sarkaria JN, Anastasiadis PZ. A Syx-RhoA-Dia1 signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in glioblastoma. JCI Insight 2023; 8:e157491. [PMID: 37427593 PMCID: PMC10371349 DOI: 10.1172/jci.insight.157491] [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: 12/13/2021] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Glioblastomas (GBM) are aggressive tumors that lack effective treatments. Here, we show that the Rho family guanine nucleotide exchange factor Syx promotes GBM cell growth both in vitro and in orthotopic xenografts derived from patients with GBM. Growth defects upon Syx depletion are attributed to prolonged mitosis, increased DNA damage, G2/M cell cycle arrest, and cell apoptosis, mediated by altered mRNA and protein expression of various cell cycle regulators. These effects are phenocopied by depletion of the Rho downstream effector Dia1 and are due, at least in part, to increased phosphorylation, cytoplasmic retention, and reduced activity of the YAP/TAZ transcriptional coactivators. Furthermore, targeting Syx signaling cooperates with radiation treatment and temozolomide (TMZ) to decrease viability in GBM cells, irrespective of their inherent response to TMZ. The data indicate that a Syx-RhoA-Dia1-YAP/TAZ signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in GBM and argue for its targeting for cancer treatment.
Collapse
Affiliation(s)
- Wan-Hsin Lin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ryan W. Feathers
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Lisa M. Cooper
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
11
|
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: 6] [Impact Index Per Article: 3.0] [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
|
12
|
Pontes B, Mendes FA. Mechanical Properties of Glioblastoma: Perspectives for YAP/TAZ Signaling Pathway and Beyond. Diseases 2023; 11:86. [PMID: 37366874 DOI: 10.3390/diseases11020086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Glioblastoma is a highly aggressive brain tumor with a poor prognosis. Recent studies have suggested that mechanobiology, the study of how physical forces influence cellular behavior, plays an important role in glioblastoma progression. Several signaling pathways, molecules, and effectors, such as focal adhesions, stretch-activated ion channels, or membrane tension variations, have been studied in this regard. Also investigated are YAP/TAZ, downstream effectors of the Hippo pathway, which is a key regulator of cell proliferation and differentiation. In glioblastoma, YAP/TAZ have been shown to promote tumor growth and invasion by regulating genes involved in cell adhesion, migration, and extracellular matrix remodeling. YAP/TAZ can be activated by mechanical cues such as cell stiffness, matrix rigidity, and cell shape changes, which are all altered in the tumor microenvironment. Furthermore, YAP/TAZ have been shown to crosstalk with other signaling pathways, such as AKT, mTOR, and WNT, which are dysregulated in glioblastoma. Thus, understanding the role of mechanobiology and YAP/TAZ in glioblastoma progression could provide new insights into the development of novel therapeutic strategies. Targeting YAP/TAZ and mechanotransduction pathways in glioblastoma may offer a promising approach to treating this deadly disease.
Collapse
Affiliation(s)
- Bruno Pontes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Fabio A Mendes
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
13
|
Patrick S, Lathoria K, Suri V, Sen E. Reduced YAP1 and FOLR1 in gliomas predict better response to chemotherapeutics. Cell Signal 2023:110738. [PMID: 37269960 DOI: 10.1016/j.cellsig.2023.110738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/21/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Gliomas harbouring mutations in IDH1 (isocitrate dehydrogenase 1) are characterized by greater sensitivity to chemotherapeutics. These mutants also exhibit diminished levels of transcriptional coactivator YAP1 (yes-associated protein 1). Enhanced DNA damage in IDH1 mutant cells, as evidenced by γH2AX formation (phosphorylation of histone variant H2A.X) and ATM (serine/threonine kinase; ataxia telangiectasia mutated) phosphorylation, was accompanied by reduced FOLR1 (folate receptor 1) expression. Diminished FOLR1, concomitant with heightened γH2AX levels, was also observed in patient-derived IDH1 mutant glioma tissues. Chromatin immunoprecipitation, overexpression of mutant YAP1, and treatment with YAP1-TEAD (TEA domain transcription factors) complex inhibitor verteporfin demonstrated regulation of FOLR1 expression by YAP1 and its partner transcription factor TEAD2. TCGA (The Cancer Genome Atlas) data analysis demonstrated better patient survival with reduced FOLR1 expression. Depletion of FOLR1 rendered IDH1 wild-type gliomas more susceptible to temozolomide-mediated death. Despite heightened DNA damage, IDH1 mutants exhibited reduced levels of IL6 (interleukin 6) and IL8 (interleukin 8) - pro-inflammatory cytokines known to be associated with persistent DNA damage. While both FOLR1 and YAP1 influenced DNA damage, only YAP1 was involved in regulating IL6 and IL8. ESTIMATE and CIBERSORTx analyses revealed the association between YAP1 expression and immune cell infiltration in gliomas. By identifying the influence of YAP1-FOLR1 link in DNA damage, our findings suggest that simultaneous depletion of both could amplify the potency of DNA damaging agents, while concomitantly reducing the release of inflammatory mediators and potentially affecting immune modulation. This study also highlights the novel role of FOLR1 as a probable prognostic marker in gliomas, predicting responsiveness to temozolomide and other DNA damaging agents.
Collapse
Affiliation(s)
| | | | - Vaishali Suri
- All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ellora Sen
- National Brain Research Centre, Manesar 122052, India.
| |
Collapse
|
14
|
Capuz A, Osien S, Karnoub MA, Aboulouard S, Laurent E, Coyaud E, Raffo-Romero A, Duhamel M, Bonnefond A, Derhourhi M, Trerotola M, El Yazidi-Belkoura I, Devos D, Zilkova M, Kobeissy F, Vanden Abeele F, Fournier I, Cizkova D, Rodet F, Salzet M. Astrocytes express aberrant immunoglobulins as putative gatekeeper of astrocytes to neuronal progenitor conversion. Cell Death Dis 2023; 14:237. [PMID: 37015912 PMCID: PMC10073301 DOI: 10.1038/s41419-023-05737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 04/06/2023]
Abstract
Using multi-omics analyses including RNAseq, RT-PCR, RACE-PCR, and shotgun proteomic with enrichment strategies, we demonstrated that newborn rat astrocytes produce neural immunoglobulin constant and variable heavy chains as well as light chains. However, their edification is different from the ones found in B cells and they resemble aberrant immunoglobulins observed in several cancers. Moreover, the complete enzymatic V(D)J recombination complex has also been identified in astrocytes. In addition, the constant heavy chain is also present in adult rat astrocytes, whereas in primary astrocytes from human fetus we identified constant and variable kappa chains as well as the substitution lambda chains known to be involved in pre-B cells. To gather insights into the function of these neural IgGs, CRISPR-Cas9 of IgG2B constant heavy chain encoding gene (Igh6), IgG2B overexpression, proximal labeling of rat astrocytes IgG2B and targets identification through 2D gels were performed. In Igh6 KO astrocytes, overrepresentation of factors involved in hematopoietic cells, neural stem cells, and the regulation of neuritogenesis have been identified. Moreover, overexpression of IgG2B in astrocytes induces the CRTC1-CREB-BDNF signaling pathway known to be involved in gliogenesis, whereas Igh6 KO triggers the BMP/YAP1/TEAD3 pathway activated in astrocytes dedifferentiation into neural progenitors. Proximal labeling experiments revealed that IgG2B is N-glycosylated by the OST complex, addressed to vesicle membranes containing the ATPase complex, and behaves partially like CD98hc through its association with LAT1. These experiments also suggest that proximal IgG2B-LAT1 interaction occurs concomitantly with MACO-1 and C2CD2L, at the heart of a potentially novel cell signaling platform. Finally, we demonstrated that these chains are synthesized individually and associated to recognize specific targets. Indeed, intermediate filaments Eif4a2 and Pdia6 involved in astrocyte fate constitute targets for these neural IgGs. Taken together, we hypothese that neural aberrant IgG chains may act as gatekeepers of astrocytes' fate.
Collapse
Affiliation(s)
- Alice Capuz
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Sylvain Osien
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Mélodie Anne Karnoub
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Soulaimane Aboulouard
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Estelle Laurent
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Etienne Coyaud
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Antonella Raffo-Romero
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Marie Duhamel
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
| | - Amélie Bonnefond
- Univ. Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, CHU de Lille, 1 place de Verdun, 59000, Lille, France
| | - Mehdi Derhourhi
- Univ. Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, CHU de Lille, 1 place de Verdun, 59000, Lille, France
| | - Marco Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), University 'G. D'Annunzio', Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, University 'G. D'Annunzio', Chieti, Italy
| | - Ikram El Yazidi-Belkoura
- Université de Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59655, Villeneuve d'Ascq, France
| | - David Devos
- Université de Lille, INSERM, U1172, CHU-Lille, Lille Neuroscience Cognition Research Centre, 1 place de Verdun, 59000, Lille, France
| | - Monika Zilkova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 84510, Bratislava, Slovakia
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fabien Vanden Abeele
- Université de Lille, INSERM U1003, Laboratory of Cell Physiology, 59655, Villeneuve d'Ascq, France
| | - Isabelle Fournier
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
- Institut Universitaire de France, 75005, Paris, France
| | - Dasa Cizkova
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 84510, Bratislava, Slovakia
- Centre for Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy in Kosice, Kosice, Slovakia
| | - Franck Rodet
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France.
| | - Michel Salzet
- Univ. Lille, Inserm, U-1192 - Laboratoire Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, 59655, Villeneuve d'Ascq, France.
- Institut Universitaire de France, 75005, Paris, France.
| |
Collapse
|
15
|
Nejabat M, Samie A, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. An Overview on Gold Nanorods as Versatile Nanoparticles in Cancer Therapy. J Control Release 2023; 354:221-242. [PMID: 36621644 DOI: 10.1016/j.jconrel.2023.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/10/2023]
Abstract
Gold nanorods (GNRs/AuNRs) are a group of gold nanoparticles which their simple surface chemistry allows for various surface modifications, providing the possibility of using them in the fabrication of biocompatible and functional nano-agents for cancer therapy. AuNRs, moreover, exhibit a maximum absorption of longitudinal localized surface plasmon resonance (LSPR) in the near-infrared (NIR) region which overlaps with NIR bio-tissue 'window' suggesting that they are proper tools for thermal ablation of cancer cells. AuNRs can be used for induction of mono or combination therapies by administering various therapeutic approaches such as photothermal therapy (PTT), photodynamic therapy (PDT), chemotherapy (CT), radiotherapy (RT), and gene therapy (GT). In this review, anticancer therapeutic capacities of AuNRs along with different surface modifications are summarized comprehensively. The roles of AuNRs in fabrication of various nano-constructs are also discussed.
Collapse
Affiliation(s)
- Masoud Nejabat
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Samie
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Masliantsev K, Mordrel M, Banor T, Desette A, Godet J, Milin S, Wager M, Karayan-Tapon L, Guichet PO. Yes-Associated Protein Nuclear Translocation Is Regulated by Epidermal Growth Factor Receptor Activation Through Phosphatase and Tensin Homolog/AKT Axis in Glioblastomas. J Transl Med 2023; 103:100053. [PMID: 36801645 DOI: 10.1016/j.labinv.2022.100053] [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: 09/21/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Gliomas are the most common and lethal primary brain tumors in adults. Glioblastomas, the most frequent and aggressive form of gliomas, represent a therapeutic challenge as no curative treatment exists to date, and the prognosis remains extremely poor. Recently, the transcriptional cofactors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) belonging to the Hippo pathway have emerged as a major determinant of malignancy in solid tumors, including gliomas. However, the mechanisms involved in its regulation, particularly in brain tumors, remain ill-defined. In glioblastomas, EGFR represents one of the most altered oncogenes affected by chromosomal rearrangements, mutations, amplifications, and overexpression. In this study, we investigated the potential link between epidermal growth factor receptor (EGFR) and the transcriptional cofactors YAP and TAZ by in situ and in vitro approaches. We first studied their activation on tissue microarray, including 137 patients from different glioma molecular subtypes. We observed that YAP and TAZ nuclear location was highly associated with isocitrate dehydrogenase 1/2 (IDH1/2) wild-type glioblastomas and poor patient outcomes. Interestingly, we found an association between EGFR activation and YAP nuclear location in glioblastoma clinical samples, suggesting a link between these 2 markers contrary to its ortholog TAZ. We tested this hypothesis in patient-derived glioblastoma cultures by pharmacologic inhibition of EGFR using gefinitib. We showed an increase of S397-YAP phosphorylation associated with decreased AKT phosphorylation after EGFR inhibition in phosphatase and tensin homolog (PTEN) wild-type cultures, unlike PTEN-mutated cell lines. Finally, we used bpV(HOpic), a potent PTEN inhibitor, to mimic the effect of PTEN mutations. We found that the inhibition of PTEN was sufficient to revert back the effect induced by Gefitinib in PTEN-wild-type cultures. Altogether, to our knowledge, these results show for the first time the regulation of pS397-YAP by the EGFR-AKT axis in a PTEN-dependent manner.
Collapse
Affiliation(s)
- Konstantin Masliantsev
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Laboratoire de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - Margaux Mordrel
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Service d'Oncologie Médicale CHU de Poitiers, Poitiers, France
| | - Tania Banor
- Service de Neurochirurgie, CHU de Poitiers, Poitiers, France
| | - Amandine Desette
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Laboratoire de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - Julie Godet
- Service d'Anatomo-Cytopathologie, CHU de Poitiers, Poitiers, France
| | - Serge Milin
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Service d'Anatomo-Cytopathologie, CHU de Poitiers, Poitiers, France
| | - Michel Wager
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Service de Neurochirurgie, CHU de Poitiers, Poitiers, France
| | - Lucie Karayan-Tapon
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Laboratoire de Cancérologie Biologique, CHU de Poitiers, Poitiers, France
| | - Pierre-Olivier Guichet
- Université de Poitiers, CHU de Poitiers, ProDiCeT, Poitiers, France; Laboratoire de Cancérologie Biologique, CHU de Poitiers, Poitiers, France.
| |
Collapse
|
17
|
Hosseinalizadeh H, Ebrahimi A, Tavakoli A, Monavari SH. Glioblastoma as a Novel Drug Repositioning Target: Updated State. Anticancer Agents Med Chem 2023; 23:1253-1264. [PMID: 36733195 DOI: 10.2174/1871520623666230202163112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 02/04/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive form of adult brain tumor that can arise from a low-grade astrocytoma. In recent decades, several new conventional therapies have been developed that have significantly improved the prognosis of patients with GBM. Nevertheless, most patients have a limited long-term response to these treatments and survive < 1 year. Therefore, innovative anti-cancer drugs that can be rapidly approved for patient use are urgently needed. One way to achieve accelerated approval is drug repositioning, extending the use of existing drugs for new therapeutic purposes, as it takes less time to validate their biological activity as well as their safety in preclinical models. In this review, a comprehensive analysis of the literature search was performed to list drugs with antiviral, antiparasitic, and antidepressant properties that may be effective in GBM and their putative anti-tumor mechanisms in GBM cells.
Collapse
Affiliation(s)
- Hamed Hosseinalizadeh
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ammar Ebrahimi
- Department of Biomedical Sciences, University of Lausanne, Rue Du Bugnon, Lausanne, Switzerland
| | - Ahmad Tavakoli
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
18
|
Jeising S, Geerling G, Guthoff R, Hänggi D, Sabel M, Rapp M, Nickel AC. In-Vitro Use of Verteporfin for Photodynamic Therapy in Glioblastoma. Photodiagnosis Photodyn Ther 2022; 40:103049. [PMID: 35932958 DOI: 10.1016/j.pdpdt.2022.103049] [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: 06/06/2022] [Revised: 07/12/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Stummer et al. established fluorescence-guided surgery (FGS) for glioblastoma (GBM) using 5-aminolevulinic acid (5-ALA). Its metabolite, protoporphyrin IX (PPIX), is also a photosensitizer and can be used for photodynamic therapy (PDT) using a laser beam of 635 nm. The porphyrin derivate verteporfin (VP) was discovered to have properties to penetrate the brain, pharmacologically target glioma cells, and is approved for PDT of choroidal neovascularization in wet age-related macular degeneration at 689 nm. OBJECTIVE To elucidate whether GBM cell lines are susceptible to PDT with second-generation photosensitizer VP. METHODS Human glioma cell lines LN229, HSR-GBM1, and a low-passage patient-derived GBM cell line P1 were treated with variable concentrations of VP for 24 h, followed by PDT at 689 nm using a diode laser light. Cell viability was measured using the MTT assay and VP uptake was measured using a desktop cytometer. RESULTS Significantly higher cell death following PDT with VP compared to VP treatment alone or no treatment was detected in all cell models (LN229, HSR-GBM1, P1). Flowcytometric measurements revealed a concentration-dependent cellular uptake of VP after 24 h incubation up to 99% at 10 µM (HSR-GBM1). CONCLUSION This study demonstrates that PDT with VP causes cell death in GBM cells at marginal concentrations. Additionally, red spectrum fluorescence was detected at therapeutic concentrations in all cell lines, validating the cellular uptake of VP in GBM cells. VP, therefore, is not only a potential drug for targeting GBM pharmacologically but can be used as an optical imaging dye in surgery and photosensitizer to make GBM susceptible to PDT.
Collapse
Affiliation(s)
- Sebastian Jeising
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Gerd Geerling
- Department of Ophthalmology, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Rainer Guthoff
- Department of Ophthalmology, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Daniel Hänggi
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Michael Sabel
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Marion Rapp
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany.
| | - Ann-Christin Nickel
- Department of Neurosurgery, University Hospital Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
19
|
Xu J, Sun S, Zhang W, Dong J, Huang C, Wang X, Jia M, Yang H, Wang Y, Jiang Y, Cao L, Huang Z. Irigenin inhibits glioblastoma progression through suppressing YAP/β-catenin signaling. Front Pharmacol 2022; 13:1027577. [PMID: 36532767 PMCID: PMC9748621 DOI: 10.3389/fphar.2022.1027577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/16/2022] [Indexed: 09/14/2024] Open
Abstract
Glioblastoma (GBM) is the most malignant glioma in brain tumors with low survival and high recurrence rate. Irigenin, as an isoflavone compound extracted from Shegan, has shown many pharmacological functions such as antioxidant, anti-inflammatory and anti-tumor. However, the effects of irigenin on GBM cells and the related molecular mechanisms remain unexplored. In this study, we found that irigenin inhibited the proliferation of GBM cells in a dose-dependent manner by several assays in vitro. Subsequently, we found that irigenin arrested cell cycle at G2/M phase and induced apoptosis of GBM cells in vitro. In addition, irigenin inhibited the migration of GBM cells. Mechanically, we found that irigenin treatment decreased the expression of YAP (yes-associated protein), suppressed β-catenin signaling. Furthermore, overexpression of YAP partially restored the anti-tumor effects of irigenin on GBM cells in vitro. Finally, we found that irigenin inhibited the growth of tumor in GBM xenograft mice model through inactivation of YAP. Taken together, these results suggest that irigenin exerts its anticancer effects on GBM via inhibiting YAP/β-catenin signaling, which may provide a new strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Jiayun Xu
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Shanshan Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Wei Zhang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jianhong Dong
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Changgang Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xin Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengxian Jia
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Hao Yang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yongjie Wang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yuanyuan Jiang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Liying Cao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Laboratory of Aging and Cancer Biology of Zhejiang Province, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zhihui Huang
- College of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Yang J, Wu X, Wang J, Guo X, Chen J, Yang X, Zhong J, Li X, Deng Z. Feedforward loop between IMP1 and YAP/TAZ promotes tumorigenesis and malignant progression in glioblastoma. Cancer Sci 2022; 114:2053-2062. [PMID: 36308276 PMCID: PMC10154852 DOI: 10.1111/cas.15636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/16/2022] [Accepted: 10/06/2022] [Indexed: 11/30/2022] Open
Abstract
YAP/TAZ have been identified as master regulators in malignant phenotypes of glioblastoma (GBM); however, YAP/TAZ transcriptional disruptor in GBM treatment remains ineffective. Whether post-transcriptional dysregulation of YAP/TAZ improves GBM outcome is currently unknown. Here, we report that insulin-like growth factor 2 (IGF2) mRNA-binding protein 1 (IGF2BP1 or IMP1) is upregulated in mesenchymal GBM compared with proneural GBM and correlates with worse patient outcome. Overexpression of IMP1 in proneural glioma stem-like cells (GSCs) promotes while IMP1 knockdown in mesenchymal GSCs attenuates tumorigenesis and mesenchymal signatures. IMP1 binds to and stabilizes m6A-YAP mRNA, leading to activation of YAP/TAZ signaling, depending on its m6A recognition and binding domain. On the other hand, TAZ functions as enhancer for IMP1 expression. Collectively, our data reveal a feedforward loop between IMP1 and YAP/TAZ maintaining GBM/GSC tumorigenesis and malignant progression and a promising molecular target in GBM.
Collapse
Affiliation(s)
- Jia Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Xujia Wu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Jia Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xing Guo
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong, China
| | - Junju Chen
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Xuesong Yang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Jian Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Xixi Li
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Zhong Deng
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| |
Collapse
|
21
|
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 2022; 42:2055-2074. [PMID: 33893939 PMCID: PMC11421619 DOI: 10.1007/s10571-021-01092-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
22
|
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
|
23
|
Current Opportunities for Targeting Dysregulated Neurodevelopmental Signaling Pathways in Glioblastoma. Cells 2022; 11:cells11162530. [PMID: 36010607 PMCID: PMC9406959 DOI: 10.3390/cells11162530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most common and highly lethal type of brain tumor, with poor survival despite advances in understanding its complexity. After current standard therapeutic treatment, including tumor resection, radiotherapy and concomitant chemotherapy with temozolomide, the median overall survival of patients with this type of tumor is less than 15 months. Thus, there is an urgent need for new insights into GBM molecular characteristics and progress in targeted therapy in order to improve clinical outcomes. The literature data revealed that a number of different signaling pathways are dysregulated in GBM. In this review, we intended to summarize and discuss current literature data and therapeutic modalities focused on targeting dysregulated signaling pathways in GBM. A better understanding of opportunities for targeting signaling pathways that influences malignant behavior of GBM cells might open the way for the development of novel GBM-targeted therapies.
Collapse
|
24
|
Terry BK, Kim S. The Role of Hippo-YAP/TAZ Signaling in Brain Development. Dev Dyn 2022; 251:1644-1665. [PMID: 35651313 DOI: 10.1002/dvdy.504] [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: 12/17/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/08/2022] Open
Abstract
In order for our complex nervous system to develop normally, both precise spatial and temporal regulation of a number of different signaling pathways is critical. During both early embryogenesis and in organ development, one pathway that has been repeatedly implicated is the Hippo-YAP/TAZ signaling pathway. The paralogs YAP and TAZ are transcriptional co-activators that play an important role in cell proliferation, cell differentiation, and organ growth. Regulation of these proteins by the Hippo kinase cascade is therefore important for normal development. In this article, we review the growing field of research surrounding the role of Hippo-YAP/TAZ signaling in normal and atypical brain development. Starting from the development of the neural tube to the development and refinement of the cerebral cortex, cerebellum, and ventricular system, we address the typical role of these transcriptional co-activators, the functional consequences that manipulation of YAP/TAZ and their upstream regulators have on brain development, and where further research may be of benefit. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bethany K Terry
- Shriners Hospitals Pediatrics Research Center, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA.,Biomedical Sciences Graduate Program, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Seonhee Kim
- Shriners Hospitals Pediatrics Research Center, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA
| |
Collapse
|
25
|
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: 0.3] [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
|
26
|
Misek SA, Newbury PA, Chekalin E, Paithankar S, Doseff AI, Chen B, Gallo KA, Neubig RR. Ibrutinib Blocks YAP1 Activation and Reverses BRAF Inhibitor Resistance in Melanoma Cells. Mol Pharmacol 2022; 101:1-12. [PMID: 34732527 PMCID: PMC11037454 DOI: 10.1124/molpharm.121.000331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/01/2021] [Indexed: 11/22/2022] Open
Abstract
Most B-Raf proto-oncogene (BRAF)-mutant melanoma tumors respond initially to BRAF inhibitor (BRAFi)/mitogen-activated protein kinase kinase 1 inhibitor (MEKi) therapy, although few patients have durable long-term responses to these agents. The goal of this study was to use an unbiased computational approach to identify inhibitors that reverse an experimentally derived BRAFi resistance gene expression signature. Using this approach, we found that ibrutinib effectively reverses this signature, and we demonstrate experimentally that ibrutinib resensitizes a subset of BRAFi-resistant melanoma cells to vemurafenib. Ibrutinib is used clinically as an inhibitor of the Src family kinase Bruton tyrosine kinase (BTK); however, neither BTK deletion nor treatment with acalabrutinib, another BTK inhibitor with reduced off-target activity, resensitized cells to vemurafenib. These data suggest that ibrutinib acts through a BTK-independent mechanism in vemurafenib resensitization. To better understand this mechanism, we analyzed the transcriptional profile of ibrutinib-treated BRAFi-resistant melanoma cells and found that the transcriptional profile of ibrutinib was highly similar to that of multiple Src proto-oncogene kinase inhibitors. Since ibrutinib, but not acalabrutinib, has appreciable off-target activity against multiple Src family kinases, it suggests that ibrutinib may be acting through this mechanism. Furthermore, genes that are differentially expressed in ibrutinib-treated cells are enriched in Yes1-associated transcriptional regulator (YAP1) target genes, and we showed that ibrutinib, but not acalabrutinib, reduces YAP1 activity in BRAFi-resistant melanoma cells. Taken together, these data suggest that ibrutinib, or other Src family kinase inhibitors, may be useful for treating some BRAFi/MEKi-refractory melanoma tumors. SIGNIFICANCE STATEMENT: MAPK-targeted therapies provide dramatic initial responses, but resistance develops rapidly; a subset of these tumors may be rendered sensitive again by treatment with an approved Src family kinase inhibitor-ibrutinub-potentially providing improved clinical outcomes.
Collapse
Affiliation(s)
- Sean A Misek
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Patrick A Newbury
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Evgenii Chekalin
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Shreya Paithankar
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Andrea I Doseff
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Bin Chen
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Kathleen A Gallo
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| | - Richard R Neubig
- Departments of Physiology (S.A.M., A.I.D., K.A.G.), Pediatrics and Human Development (P.A.N., E.C., S.P., B.C.), and Pharmacology (A.I.D., B.C., R.R.N.), Michigan State University, East Lansing, Michigan
| |
Collapse
|
27
|
Noorbakhsh N, Hayatmoghadam B, Jamali M, Golmohammadi M, Kavianpour M. The Hippo signaling pathway in leukemia: function, interaction, and carcinogenesis. Cancer Cell Int 2021; 21:705. [PMID: 34953494 PMCID: PMC8710012 DOI: 10.1186/s12935-021-02408-7] [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: 08/24/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer can be considered as a communication disease between and within cells; nevertheless, there is no effective therapy for the condition, and this disease is typically identified at its late stage. Chemotherapy, radiation, and molecular-targeted treatment are typically ineffective against cancer cells. A better grasp of the processes of carcinogenesis, aggressiveness, metastasis, treatment resistance, detection of the illness at an earlier stage, and obtaining a better therapeutic response will be made possible. Researchers have discovered that cancerous mutations mainly affect signaling pathways. The Hippo pathway, as one of the main signaling pathways of a cell, has a unique ability to cause cancer. In order to treat cancer, a complete understanding of the Hippo signaling system will be required. On the other hand, interaction with other pathways like Wnt, TGF-β, AMPK, Notch, JNK, mTOR, and Ras/MAP kinase pathways can contribute to carcinogenesis. Phosphorylation of oncogene YAP and TAZ could lead to leukemogenesis, which this process could be regulated via other signaling pathways. This review article aimed to shed light on how the Hippo pathway interacts with other cellular signaling networks and its functions in leukemia.
Collapse
Affiliation(s)
| | - Bentolhoda Hayatmoghadam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Jamali
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Golmohammadi
- Applied Cell Sciences and Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maria Kavianpour
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Patrick S, Gowda P, Lathoria K, Suri V, Sen E. YAP1-mediated regulation of mitochondrial dynamics in IDH1 mutant gliomas. J Cell Sci 2021; 134:273515. [PMID: 34651186 DOI: 10.1242/jcs.259188] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/07/2021] [Indexed: 11/20/2022] Open
Abstract
Mutation of the isocitrate dehydrogenase 1 (IDH1) gene leads to the production of oncometabolite D-2-hydroxyglutarate (2-HG) from α-ketoglutarate and is associated with better prognosis in glioma. As Yes-associated protein 1 (YAP1) is an important regulator of tumor progression, its role in glioma expressing IDH1 with an R132H mutation was investigated. Diminished nuclear levels of YAP1 in IDH1 mutant glioma tissues and cell lines were accompanied by decreased levels of mitochondrial transcription factor A (TFAM). Luciferase reporter assays and chromatin immunoprecipitation were used to investigate the functionality of the TEAD2-binding site on the TFAM promoter in mediating its YAP1-dependent expression. YAP1-dependent mitochondrial fragmentation and ROS generation were accompanied by decreased telomerase reverse transcriptase (TERT) levels and increased mitochondrial TERT localization in IDH1 R132H cells. Treatment with the Src kinase inhibitor bosutinib, which prevents extranuclear shuttling of TERT, further elevated ROS in IDH1 R132H cells and triggered apoptosis. Importantly, bosutinib treatment also increased ROS levels and induced apoptosis in IDH1 wild-type cells when YAP1 was concurrently depleted. These findings highlight the involvement of YAP1 in coupling mitochondrial dysfunction with mitochondrial shuttling of TERT to constitute an essential non-canonical function of YAP1 in the regulation of redox homeostasis. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Shruti Patrick
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, India
| | - Pruthvi Gowda
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, India
| | - Kirti Lathoria
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Ellora Sen
- Division of Cellular and Molecular Neuroscience, National Brain Research Centre, Manesar 122052, India
| |
Collapse
|
29
|
The Expression and Prognostic Value of ILK and YAP1 in Glioma. Appl Immunohistochem Mol Morphol 2021; 30:e21-e29. [DOI: 10.1097/pai.0000000000000984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/13/2021] [Indexed: 11/26/2022]
|
30
|
Kim HR, Seo CW, Yoo K, Han SJ, Kim J. Yes-associated protein 1 as a prognostic biomarker and its correlation with telomerase in various cancers. Osong Public Health Res Perspect 2021; 12:324-332. [PMID: 34719224 PMCID: PMC8561022 DOI: 10.24171/j.phrp.2021.0207] [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: 07/26/2021] [Accepted: 09/02/2021] [Indexed: 11/06/2022] Open
Abstract
Objectives The aims of this study were to investigate the expression of Yes-associated protein 1 (YAP1), its prognostic significance, and the correlation between YAP1 and telomerase in various cancers. Methods The Gene Expression Profiling Interactive Analysis database was used to analyze RNA sequencing data and the survival rate of patients with various cancers in The Cancer Genome Atlas (TCGA) database. PrognoScan was used to analyze the prognostic value of YAP1 expression in various cancers. Tumor Immune Estimation Resource was used to determine the correlation between YAP1 expression and telomerase in various cancer types based on TCGA data. Results The analysis suggested that YAP1 was differentially expressed between tissues of various cancers and non-tumor tissues. High YAP1 expression was also related to a poor prognosis in adrenocortical carcinoma, bladder urothelial carcinoma, and pancreatic adenocarcinoma. Moreover, YAP1 expression was correlated with the expression of telomerase reverse transcriptase and telomerase RNA component in various cancer types. Conclusion These results suggest that YAP1 is a potential biomarker with prognostic significance and relevance for oncogene research in various cancer types. The correlation between the expression of YAP1 and telomere-associated genes will help to understand their cancer-promoting mechanisms and interactions.
Collapse
Affiliation(s)
- Hye-Ran Kim
- Department of Biomedical Laboratory Science, Dong-Eui Institute of Technology, Busan, Korea
| | - Choong-Won Seo
- Department of Biomedical Laboratory Science, Dong-Eui Institute of Technology, Busan, Korea
| | - Keunje Yoo
- Department of Environmental Engineering, Korea Maritime and Ocean University, Busan, Korea
| | - Sang Jun Han
- Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, Busan, Korea
| | - Jongwan Kim
- Department of Biomedical Laboratory Science, Dong-Eui Institute of Technology, Busan, Korea
| |
Collapse
|
31
|
Wang X, Lu J, Li J, Liu Y, Guo G, Huang Q. CYT387, a potent IKBKE inhibitor, suppresses human glioblastoma progression by activating the Hippo pathway. J Transl Med 2021; 19:396. [PMID: 34544426 PMCID: PMC8454155 DOI: 10.1186/s12967-021-03070-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/02/2021] [Indexed: 01/18/2023] Open
Abstract
Recent studies have showed that IKBKE is overexpressed in several kinds of cancers and that IKBKE-knockdown inhibits tumor progression. In this article, we first verified that two glioblastoma cell lines, U87-MG and LN-229, were sensitive to CYT387 by measuring the half maximal inhibitory concentration (IC50) with a CCK-8 assay and then demonstrated that CYT387, as a potent IKBKE inhibitor, suppressed glioblastoma cell proliferation, migration and invasion. Additionally, CYT387 induced cell apoptosis and arrested the cell cycle at the G2/M checkpoint in vitro. Furthermore, we showed that CYT387 did not simply inhibit IKBKE activity but also decreased IKBKE expression at the protein level rather than at the mRNA level. We discovered that CYT387 restrained malignant tumor progression by activating the Hippo pathway in vitro. By coimmunoprecipitation (co-IP), we showed that IKBKE interacted with TEAD2 and YAP1, thus accelerating TEAD2 and YAP1 transport into the nucleus. In subsequent in vivo experiments, we found that CYT387 inhibited subcutaneous nude mouse tumor growth but had little impact on intracranial orthotopic xenografts, probably due to a limited ability to penetrate the blood–brain barrier (BBB). These results suggest that CYT387 has potential as a new antiglioblastoma drug, but an approach to allow passage through the blood–brain barrier (BBB) is needed.
Collapse
Affiliation(s)
- Xin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jie Lu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan, Shandong, China
| | - Jing Li
- Department of Nursing, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yang Liu
- Department of Neurosurgery, Renmin Hospital of Henan Province, Zhengzhou, Henan, China
| | - Gaochao Guo
- Department of Neurosurgery, Renmin Hospital of Henan Province, Zhengzhou, Henan, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China. .,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, People's Republic of China.
| |
Collapse
|
32
|
Wang Y, Wang X, Wang X, Wu D, Qi J, Zhang Y, Wang K, Zhou D, Meng QM, Nie E, Wang Q, Yu RT, Zhou XP. Imipramine impedes glioma progression by inhibiting YAP as a Hippo pathway independent manner and synergizes with temozolomide. J Cell Mol Med 2021; 25:9350-9363. [PMID: 34469035 PMCID: PMC8500960 DOI: 10.1111/jcmm.16874] [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: 06/11/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with malignant glioma often suffered from depression, which leads to an increased risk of detrimental outcomes. Imipramine, an FDA‐approved tricyclic antidepressant, has been commonly used to relieve depressive symptoms in the clinic. Recently, imipramine has been reported to participate in the suppression of tumour progression in several human cancers, including prostate cancer, colon cancer and lymphomas. However, the effect of imipramine on malignant glioma is largely unclear. Here, we show that imipramine significantly retarded proliferation of immortalized and primary glioma cells. Mechanistically, imipramine suppressed tumour proliferation by inhibiting yes‐associated protein (YAP), a recognized oncogene in glioma, independent of Hippo pathway. In addition to inhibiting YAP transcription, imipramine also promoted the subcellular translocation of YAP from nucleus into cytoplasm. Consistently, imipramine administration significantly reduced orthotopic tumour progression and prolonged survival of tumour‐bearing mice. Moreover, exogenous overexpression of YAP partially restored the inhibitory effect of imipramine on glioma progression. Most importantly, compared with imipramine or temozolomide (TMZ) monotherapy, combination therapy with imipramine and TMZ exhibited enhanced inhibitory effect on glioma growth both in vitro and in vivo, suggesting the synergism of both agents. In conclusion, we found that tricyclic antidepressant imipramine impedes glioma progression by inhibiting YAP. In addition, combination therapy with imipramine and TMZ may potentially serve as promising anti‐glioma regimens, thus predicting a broad prospect of clinical application.
Collapse
Affiliation(s)
- Yan Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiang Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Xu Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Di Wu
- Pathological Diagnosis Center, Xuzhou Central Hospital, Xuzhou, China
| | - Ji Qi
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Kai Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Ding Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Qing-Ming Meng
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Er Nie
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qiang Wang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ru-Tong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiu-Ping Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
33
|
Holmes B, Benavides-Serrato A, Saunders JT, Kumar S, Nishimura RN, Gera J. mTORC2-mediated direct phosphorylation regulates YAP activity promoting glioblastoma growth and invasive characteristics. Neoplasia 2021; 23:951-965. [PMID: 34343821 PMCID: PMC8347669 DOI: 10.1016/j.neo.2021.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/24/2022]
Abstract
The Hippo and mTOR signaling cascades are major regulators of cell growth and division. Aberrant regulation of these pathways has been demonstrated to contribute to gliomagenesis and result in enhanced glioblastoma proliferation and invasive characteristics. Several crosstalk mechanisms have been described between these two pathways, although a complete picture of these signaling interactions is lacking and is required for effective therapeutic targeting. Here we report the ability of mTORC2 to directly phosphorylate YAP at serine 436 (Ser436) positively regulating YAP activity. We show that mTORC2 activity enhances YAP transcriptional activity and the induction of YAP-dependent target gene expression while its ablation via genetic or pharmacological means has the opposite affects on YAP function. mTORC2 interacts with YAP via Sin1 and mutational analysis of serine 436 demonstrates that this phosphorylation event affects several properties of YAP leading to enhanced transactivation potential. Moreover, YAP serine 436 mutants display altered glioblastoma growth, migratory capacity and invasiveness both in vitro and in xenograft experiments. We further demonstrate that mTORC2 is able to regulate a Hippo pathway resistant allele of YAP suggesting that mTORC2 can regulate YAP independent of Hippo signaling. Correlative associations between the expression of these components in GBM patient samples also supported the presence of this signaling relationship. These results advance a direct mTORC2/YAP signaling axis driving GBM growth, motility and invasiveness.
Collapse
Affiliation(s)
- Brent Holmes
- Departments of Medicine; Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CL
| | - Angelica Benavides-Serrato
- Departments of Medicine; Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CL
| | - Jacquelyn T Saunders
- Departments of Medicine; Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CL
| | - Sunil Kumar
- Department of Pharmaceutical and Biomedical Sciences, California Health Sciences University, Clovis, CL; Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CL
| | - Robert N Nishimura
- Neurology, David Geffen School of Medicine at UCLA; Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CL
| | - Joseph Gera
- Departments of Medicine; Jonnson Comprehensive Cancer Center; Molecular Biology Institute, University of California-Los Angeles, CL; Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CL.
| |
Collapse
|
34
|
Trejo-Solis C, Escamilla-Ramirez A, Jimenez-Farfan D, Castillo-Rodriguez RA, Flores-Najera A, Cruz-Salgado A. Crosstalk of the Wnt/β-Catenin Signaling Pathway in the Induction of Apoptosis on Cancer Cells. Pharmaceuticals (Basel) 2021; 14:ph14090871. [PMID: 34577571 PMCID: PMC8465904 DOI: 10.3390/ph14090871] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays a major role in cell survival and proliferation, as well as in angiogenesis, migration, invasion, metastasis, and stem cell renewal in various cancer types. However, the modulation (either up- or downregulation) of this pathway can inhibit cell proliferation and apoptosis both through β-catenin-dependent and independent mechanisms, and by crosstalk with other signaling pathways in a wide range of malignant tumors. Existing studies have reported conflicting results, indicating that the Wnt signaling can have both oncogenic and tumor-suppressing roles, depending on the cellular context. This review summarizes the available information on the role of the Wnt/β-catenin pathway and its crosstalk with other signaling pathways in apoptosis induction in cancer cells and presents a modified dual-signal model for the function of β-catenin. Understanding the proapoptotic mechanisms induced by the Wnt/β-catenin pathway could open new therapeutic opportunities.
Collapse
Affiliation(s)
- Cristina Trejo-Solis
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (A.E.-R.); (A.C.-S.)
- Correspondence:
| | - Angel Escamilla-Ramirez
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (A.E.-R.); (A.C.-S.)
| | - Dolores Jimenez-Farfan
- Laboratorio de Inmunología, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | | | - Athenea Flores-Najera
- Centro Médico Nacional 20 de Noviembre, Departamento de Cirugía General, Ciudad de Mexico 03229, Mexico;
| | - Arturo Cruz-Salgado
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico 14269, Mexico; (A.E.-R.); (A.C.-S.)
| |
Collapse
|
35
|
Feng Y, Ci H, Wu Q. Expression of mammalian sterile 20-like kinase 1 and 2 and Yes-associated protein 1 proteins in triple-negative breast cancer and the clinicopathological significance. Medicine (Baltimore) 2021; 100:e27032. [PMID: 34449481 PMCID: PMC8389968 DOI: 10.1097/md.0000000000027032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/07/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND AIM Mammalian sterile 20-like kinase 1 and 2 (MST1/2) and Yes-associated protein 1 (YAP1) are the core molecules of the Hippo signaling pathway, which have been found to be unbalanced in the occurrence of tumors and promote the development of the lesions. The present study aimed to investigate the expression of MST1/2 and YAP1 proteins in triple-negative breast cancer (TNBC) and their clinicopathological significance. METHODS Immunohistochemistry was used to detect the expression level of protein in tissues. According to the percentage of positive cells and staining intensity, the expression intensity of MST1/2 and YAP1 proteins in the tissue samples was scored, and the correlation between MST1/2 and the clinicopathological features of TNBC were discussed. RESULTS The expression of MST1/2 and YAP1 was associated with histological grade, metastasis, lymph node metastasis stage, and tumor node metastasis stage. The overexpression of YAP1 predicted a poor prognosis in terms of overall survival and disease-free survival time. The MST1/2 expression was associated with improved overall survival and disease free survival of the patients. CONCLUSION MST1/2 and YAP1 may be used as prognostic indicators to evaluate the recurrence of TNBC and might become one of the new targets for breast cancer treatment.
Collapse
Affiliation(s)
- Yang Feng
- Department of Pathology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, Anhui, China
- Department of Pathology, Bengbu Medical College, Bengbu, Anhui, China
| | - Hongfei Ci
- Department of Pathology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, Anhui, China
- Department of Pathology, Bengbu Medical College, Bengbu, Anhui, China
| | - Qiong Wu
- Department of Pathology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), Suzhou, Anhui, China
- Department of Pathology, Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
36
|
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: 1.5] [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
|
37
|
Nuclear Dbf2-related (NDR1) functions as tumor suppressor in glioblastoma by phosphorylation of Yes-associated protein (YAP). Chin Med J (Engl) 2021; 134:2054-2065. [PMID: 34343153 PMCID: PMC8440018 DOI: 10.1097/cm9.0000000000001653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: The Nuclear Dbf2-related (NDR1) kinase is a member of the NDR/LATS family, which was a supplementary of Hippo pathway. However, whether NDR1 could inhibit glioblastoma (GBM) growth by phosphorylating Yes-associated protein (YAP) remains unknown. Meanwhile, the role of NDR1 in GBM was not clear. This study aimed to investigate the role of NDR1-YAP pathway in GBM. Methods: Bioinformation analysis and immunohistochemistry (IHC) were performed to identify the expression of NDR1 in GBM. The effect of NDR1 on cell proliferation and cell cycle was analyzed utilizing CCK-8, clone formation, immunofluorescence and flow cytometry, respectively. In addition, the xenograft tumor model was established as well. Protein interaction was examined by Co-immunoprecipitation and immunofluorescence to observe co-localization. Results: Bioinformation analysis and IHC of our patients’ tumor tissues showed that expression of NDR1 in tumor tissue was relatively lower than that in normal tissues and was positively related to a lower survival rate. NDR1 could markedly reduce the proliferation and colony formation of U87 and U251. Furthermore, the results of flow cytometry showed that NDR1 led to cell cycle arrest at the G1 phase. Tumor growth was also inhibited in xenograft nude mouse models in NDR1-overexpression group. Western blotting and immunofluorescence showed that NDR1 could integrate with and phosphorylate YAP at S127 site. Meanwhile, NDR1 could mediate apoptosis process. Conclusion: In summary, our findings point out that NDR1 functions as a tumor suppressor in GBM. NDR1 is identified as a novel regulator of YAP, which gives us an in-depth comprehension of the Hippo signaling pathway.
Collapse
|
38
|
Wu B, Tang X, Ke H, Zhou Q, Zhou Z, Tang S, Ke R. Gene Regulation Network of Prognostic Biomarker YAP1 in Human Cancers: An Integrated Bioinformatics Study. Pathol Oncol Res 2021; 27:1609768. [PMID: 34257617 PMCID: PMC8262238 DOI: 10.3389/pore.2021.1609768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/24/2021] [Indexed: 12/20/2022]
Abstract
Background: Yes-associated protein 1 (YAP1) is the main downstream effector of the Hippo signaling pathway, which is involved in tumorigenesis. This study aimed to comprehensively understand the prognostic performances of YAP1 expression and its potential mechanism in pan-cancers by mining databases. Methods: The YAP1 expression was evaluated by the Oncomine database and GEPIA tool. The clinical significance of YAP1 expression was analyzed by the UALCAN, GEPIA, and DriverDBv3 database. Then, the co-expressed genes with YAP1 were screened by the LinkedOmics, and annotated by the Metascape and DAVID database. Additionally, by the MitoMiner 4.0 v tool, the YAP1 co-expressed genes were screened to obtain the YAP1-associated mitochondrial genes that were further enriched by DAVID and analyzed by MCODE for the hub genes. Results: YAP1 was differentially expressed in human cancers. Higher YAP1 expression was significantly associated with poorer overall survival and disease-free survival in adrenocortical carcinoma (ACC), brain Lower Grade Glioma (LGG), and pancreatic adenocarcinoma (PAAD). The LinkedOmics analysis revealed 923 co-expressed genes with YAP1 in adrenocortical carcinoma, LGG and PAAD. The 923 genes mainly participated in mitochondrial functions including mitochondrial gene expression and mitochondrial respiratory chain complex I assembly. Of the 923 genes, 112 mitochondrial genes were identified by MitoMiner 4.0 v and significantly enriched in oxidative phosphorylation. The MCODE analysis identified three hub genes including CHCHD1, IDH3G and NDUFAF5. Conclusion: Our findings showed that the YAP1 overexpression could be a biomarker for poor prognosis in ACC, LGG and PAAD. Specifically, the YAP1 co-expression genes were mainly involved in the regulation of mitochondrial function especially in oxidative phosphorylation. Thus, our findings provided evidence of the carcinogenesis of YAP1 in human cancers and new insights into the mechanisms underlying the role of YAP1 in mitochondrial dysregulation.
Collapse
Affiliation(s)
- Baojin Wu
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinjie Tang
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Honglin Ke
- Department of Emergency, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Qiong Zhou
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Zhaoping Zhou
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Shao Tang
- Department of Statistics, Florida State University, Tallahassee, FL, United States
| | - Ronghu Ke
- Department of Plastic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
39
|
Ma D, Zhan D, Fu Y, Wei S, Lal B, Wang J, Li Y, Lopez-Bertoni H, Yalcin F, Dzaye O, Eberhart CG, Laterra J, Wilson MA, Ying M, Xia S. Mutant IDH1 promotes phagocytic function of microglia/macrophages in gliomas by downregulating ICAM1. Cancer Lett 2021; 517:35-45. [PMID: 34098063 DOI: 10.1016/j.canlet.2021.05.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/15/2022]
Abstract
Tumor-associated microglia/macrophages (TAMs) are the main innate immune effector cells in malignant gliomas and have both pro- and anti-tumor functions. The plasticity of TAMs is partially dictated by oncogenic mutations in tumor cells. Heterozygous IDH1 mutation is a cancer driver gene prevalent in grade II/III gliomas, and IDH1 mutant gliomas have relatively favorable clinical outcomes. It is largely unknown how IDH mutation alters TAM phenotypes to influence glioma growth. Here we established clinically relevant isogenic glioma models carrying monoallelic IDH1 R132H mutation (IDH1R132H/WT) and found that IDH1R132H/WT significantly downregulated immune response-related pathways in glioma cells, indicating an immunomodulation role of mutant IDH1. Co-culturing IDH1R132H/WT glioma cells with human macrophages promoted anti-tumor phenotypes of macrophages and increased macrophage migration and phagocytic capacity. In orthotopic xenografts, IDH1R132H/WT decreased tumor growth and prolonged animal survival, accompanied by increased TAM recruitment and upregulated phagocytosis markers, suggesting the induction of anti-tumor TAM functions. Using human cytokine arrays that query 36 proteins, we identified significant downregulation of ICAM-1/CD54 in IDH1R132H/WT gliomas, which was further confirmed by ELISA and immunoblotting analyses. ICAM1 gain-of-function studies revealed that ICAM1 downregulation in IDH1R132H/WT cells played a mechanistic role to mediate the immunomodulation function of IDH1R132H/WT. ICAM-1 silencing in IDH1 wild-type glioma cells decreased tumor growth and increased the anti-tumor function of TAMs. Together, our studies support a new TAM-mediated phagocytic function within IDH1 mutant gliomas, and improved understanding of this process may uncover novel approaches to targeting IDH1 wild type gliomas.
Collapse
Affiliation(s)
- Ding Ma
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Blood and Cell Therapy Institute, University of Science and Technology of China, Anhui Provincial Hospital, Hefei, Anhui, China.
| | - Daqian Zhan
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Fu
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jie Wang
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yunqing Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hernando Lopez-Bertoni
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fatih Yalcin
- Department of Radiology and Neuroradiology, Charité, Berlin, Germany; University Hospital Center Schleswig Holstein, Department of Neurosurgery, Kiel, Schleswig-Holstein, Germany; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Omar Dzaye
- Department of Radiology and Neuroradiology, Charité, Berlin, Germany; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles G Eberhart
- Departments of Pathology, Oncology, Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Oncology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mary Ann Wilson
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingyao Ying
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Shuli Xia
- Hugo W. Moser Research Institute at Kennedy Krieger, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
40
|
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: 4.0] [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
|
41
|
Lee SY, Yen IC, Lin JC, Chung MC, Liu WH. 4-Acetylantrocamol LT3 Inhibits Glioblastoma Cell Growth and Downregulates DNA Repair Enzyme O 6-Methylguanine-DNA Methyltransferase. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:983-999. [PMID: 33827387 DOI: 10.1142/s0192415x21500476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Glioblastoma multiforme (GBM) is a deadly malignant brain tumor that is resistant to most clinical treatments. Novel therapeutic agents that are effective against GBM are required. Antrodia cinnamomea has shown antiproliferative effects in GBM cells. However, the exact mechanisms and bioactive components remain unclear. Thus, the present study aimed to investigate the effect and mechanism of 4-acetylantrocamol LT3 (4AALT3), a new ubiquinone from Antrodia cinnamomeamycelium, in vitro. U87 and U251 cell lines were treated with the indicated concentration of 4AALT3. Cell viability, cell colony-forming ability, migration, and the expression of proteins in well-known signaling pathways involved in the malignant properties of glioblastoma were then analyzed by CCK-8, colony formation, wound healing, and western blotting assays, respectively. We found that 4AALT3 significantly decreased cell viability, colony formation, and cell migration in both in vitro models. The epidermal growth factor receptor (EGFR), phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), Hippo/yes-associated protein (YAP), and cAMP-response element binding protein (CREB) pathways were suppressed by 4AALT3. Moreover, 4AALT3 decreased the level of DNA repair enzyme O6-methylguanine-DNA methyltransferase and showed a synergistic effect with temozolomide. Our findings provide the basis for exploring the beneficial effect of 4AALT3 on GBM in vivo.
Collapse
Affiliation(s)
- Shih-Yu Lee
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - I-Chuan Yen
- School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
| | - Jang-Chun Lin
- Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Min-Chieh Chung
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Hsiu Liu
- Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan.,Department of Neurological Surgery Tri-Service General Hospital and National Defense Medical Center, No. 325, Sec. 2 Cheng-Kung Road Taipei 11490, Taiwan
| |
Collapse
|
42
|
Sungura R, Onyambu C, Mpolya E, Sauli E, Vianney JM. The extended scope of neuroimaging and prospects in brain atrophy mitigation: A systematic review. INTERDISCIPLINARY NEUROSURGERY 2021. [DOI: 10.1016/j.inat.2020.100875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
43
|
Castellan M, Guarnieri A, Fujimura A, Zanconato F, Battilana G, Panciera T, Sladitschek HL, Contessotto P, Citron A, Grilli A, Romano O, Bicciato S, Fassan M, Porcù E, Rosato A, Cordenonsi M, Piccolo S. Single-cell analyses reveal YAP/TAZ as regulators of stemness and cell plasticity in Glioblastoma. NATURE CANCER 2021; 2:174-188. [PMID: 33644767 PMCID: PMC7116831 DOI: 10.1038/s43018-020-00150-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma (GBM) is a devastating human malignancy. GBM stem-like cells (GSCs) drive tumor initiation and progression. Yet, the molecular determinants defining GSCs in their native state in patients remain poorly understood. Here we used single cell datasets and identified GSCs at the apex of the differentiation hierarchy of GBM. By reconstructing the GSCs' regulatory network, we identified the YAP/TAZ coactivators as master regulators of this cell state, irrespectively of GBM subtypes. YAP/TAZ are required to install GSC properties in primary cells downstream of multiple oncogenic lesions, and required for tumor initiation and maintenance in vivo in different mouse and human GBM models. YAP/TAZ act as main roadblock of GSC differentiation and their inhibition irreversibly lock differentiated GBM cells into a non-tumorigenic state, preventing plasticity and regeneration of GSC-like cells. Thus, GSC identity is linked to a key molecular hub integrating genetics and microenvironmental inputs within the multifaceted biology of GBM.
Collapse
Affiliation(s)
| | | | - Atsushi Fujimura
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Giusy Battilana
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Tito Panciera
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | | | - Anna Citron
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Fassan
- Department of Medicine - Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy
| | - Elena Porcù
- Department of Woman and Children Health, University of Padua, Padua, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
44
|
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: 2.0] [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
|
45
|
Koehler A, Karve A, Desai P, Arbiser J, Plas DR, Qi X, Read RD, Sasaki AT, Gawali VS, Toukam DK, Bhattacharya D, Kallay L, Pomeranz Krummel DA, Sengupta S. Reuse of Molecules for Glioblastoma Therapy. Pharmaceuticals (Basel) 2021; 14:99. [PMID: 33525329 PMCID: PMC7912673 DOI: 10.3390/ph14020099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly malignant primary brain tumor. The current standard of care for GBM is the Stupp protocol which includes surgical resection, followed by radiotherapy concomitant with the DNA alkylator temozolomide; however, survival under this treatment regimen is an abysmal 12-18 months. New and emerging treatments include the application of a physical device, non-invasive 'tumor treating fields' (TTFs), including its concomitant use with standard of care; and varied vaccines and immunotherapeutics being trialed. Some of these approaches have extended life by a few months over standard of care, but in some cases are only available for a minority of GBM patients. Extensive activity is also underway to repurpose and reposition therapeutics for GBM, either alone or in combination with the standard of care. In this review, we present select molecules that target different pathways and are at various stages of clinical translation as case studies to illustrate the rationale for their repurposing-repositioning and potential clinical use.
Collapse
Affiliation(s)
- Abigail Koehler
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| | - Aniruddha Karve
- Division of Pharmaceutical Sciences, University of Cincinnati James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (A.K.); (P.D.)
| | - Pankaj Desai
- Division of Pharmaceutical Sciences, University of Cincinnati James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (A.K.); (P.D.)
| | - Jack Arbiser
- Department of Dermatology, Emory School of Medicine, Atlanta, GA 30322, USA;
- Atlanta Veterans Administration Medical Center, Decatur, GA 30033, USA
| | - David R. Plas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Xiaoyang Qi
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (X.Q.); (A.T.S.)
| | - Renee D. Read
- Department of Pharmacology and Chemical Biology, Emory School of Medicine, Atlanta, GA 30322, USA;
| | - Atsuo T. Sasaki
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (X.Q.); (A.T.S.)
| | - Vaibhavkumar S. Gawali
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| | - Donatien K. Toukam
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| | - Debanjan Bhattacharya
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| | - Laura Kallay
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| | - Daniel A. Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (A.K.); (V.S.G.); (D.K.T.); (D.B.); (L.K.); (D.A.P.K.)
| |
Collapse
|
46
|
Masliantsev K, Karayan-Tapon L, Guichet PO. Hippo Signaling Pathway in Gliomas. Cells 2021; 10:184. [PMID: 33477668 PMCID: PMC7831924 DOI: 10.3390/cells10010184] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
The Hippo signaling pathway is a highly conserved pathway involved in tissue development and regeneration that controls organ size through the regulation of cell proliferation and apoptosis. The core Hippo pathway is composed of a block of kinases, MST1/2 (Mammalian STE20-like protein kinase 1/2) and LATS1/2 (Large tumor suppressor 1/2), which inhibits nuclear translocation of YAP/TAZ (Yes-Associated Protein 1/Transcriptional co-activator with PDZ-binding motif) and its downstream association with the TEAD (TEA domain) family of transcription factors. This pathway was recently shown to be involved in tumorigenesis and metastasis in several cancers such as lung, breast, or colorectal cancers but is still poorly investigated in brain tumors. Gliomas are the most common and the most lethal primary brain tumors representing about 80% of malignant central nervous system neoplasms. Despite intensive clinical protocol, the prognosis for patients remains very poor due to systematic relapse and treatment failure. Growing evidence demonstrating the role of Hippo signaling in cancer biology and the lack of efficient treatments for malignant gliomas support the idea that this pathway could represent a potential target paving the way for alternative therapeutics. Based on recent advances in the Hippo pathway deciphering, the main goal of this review is to highlight the role of this pathway in gliomas by a state-of-the-art synthesis.
Collapse
Affiliation(s)
- Konstantin Masliantsev
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| | - Lucie Karayan-Tapon
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| | - Pierre-Olivier Guichet
- Inserm U1084, Laboratoire de Neurosciences Expérimentales et Cliniques, F-86073 Poitiers, France; (K.M.); (L.K.-T.)
- Université de Poitiers, F-86073 Poitiers, France
- CHU de Poitiers, Laboratoire de Cancérologie Biologique, F-86022 Poitiers, France
| |
Collapse
|
47
|
Mao W, Mai J, Peng H, Wan J, Sun T. YAP in pancreatic cancer: oncogenic role and therapeutic strategy. Theranostics 2021; 11:1753-1762. [PMID: 33408779 PMCID: PMC7778590 DOI: 10.7150/thno.53438] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer, especially pancreatic ductal adenocarcinoma (PDAC), remains a fatal disease with few efficacious treatments. The Hippo signaling pathway, an evolutionarily conserved signaling module, plays critical roles in tissue homeostasis, organ size control and tumorigenesis. The transcriptional coactivator yes-associated protein (YAP), a major downstream effector of the Hippo pathway, is associated with various human cancers including PDAC. Considering its importance in cancer, YAP is emerging as a promising therapeutic target. In this review, we summarize the current understanding of the oncogenic role and regulatory mechanism of YAP in PDAC, and the potential therapeutic strategies targeting YAP.
Collapse
|
48
|
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: 74] [Impact Index Per Article: 14.8] [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
|
49
|
Wang W, Li Y, Zhi S, Li J, Miao J, Ding Z, Peng Y, Huang Y, Zheng R, Yu H, Qi P, Wang J, Fu X, Hu M, Chen S. LncRNA-ROR/microRNA-185-3p/YAP1 axis exerts function in biological characteristics of osteosarcoma cells. Genomics 2020; 113:450-461. [PMID: 32898639 DOI: 10.1016/j.ygeno.2020.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/21/2020] [Accepted: 09/03/2020] [Indexed: 11/19/2022]
Abstract
AIM The co-expression network of long non-coding RNA ROR (lncRNA-ROR) and microRNA-185-3p (miR-185-3p) has not been focused on osteosarcoma. Therein, this work was initiated to uncover lncRNA-ROR and miR-185-3p functions in osteosarcoma. METHODS LncRNA-ROR, miR-185-3p and Yes-associated protein 1 (YAP1) expression in osteosarcoma tissues and cells were detected. The screened cells (MG63 and U2OS) were transfected with decreased and/or increased lncRNA-ROR and miR-185-3p to explore osteosarcoma progression. Tumor growth was detected by tumor xenografts in mice. RESULTS Up-regulated lncRNA-ROR and YAP1 and down-regulated miR-185-3p were found in osteosarcoma. LncRNA ROR knockdown or miR-185-3p overexpression inhibited osteosarcoma cell progression while lncRNA ROR elevation or miR-185-3p inhibition presented the opposite effects. Function of lncRNA ROR was rescued by miR-185-3p and regulated the growth and metastasis of osteosarcoma cells via modulating YAP1, the target gene of miR-185-3p. CONCLUSION This work illustrates that lncRNA-ROR down-regulation or miR-185-3p up-regulation inhibits osteosarcoma progression via YAP1 repression.
Collapse
Affiliation(s)
- Weiguo Wang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yuezhan Li
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Shuang Zhi
- Four Gynecological Wards, Ningbo Women & Children's Hospital, Ningbo 315000, Zhejiang, China
| | - Jinsong Li
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Jinglei Miao
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhiyu Ding
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yi Peng
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Yan Huang
- The Second Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Ruping Zheng
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China
| | - Haiyang Yu
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan, China
| | - Pei Qi
- Department of pharmacy, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan, China
| | - Jianlong Wang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xingchang Fu
- Department of Orthopedics, Hunan Aerospace hospital, Changsha 410205, Hunan, China
| | - Minghua Hu
- Department of Anatomy, Histology and Embryology, Changsha Medical University, Changsha 410219, China
| | - Shijie Chen
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
50
|
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: 0.8] [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
|