1
|
Shi Q, Xue C, Zeng Y, Yuan X, Chu Q, Jiang S, Wang J, Zhang Y, Zhu D, Li L. Notch signaling pathway in cancer: from mechanistic insights to targeted therapies. Signal Transduct Target Ther 2024; 9:128. [PMID: 38797752 PMCID: PMC11128457 DOI: 10.1038/s41392-024-01828-x] [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: 01/18/2024] [Revised: 03/31/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Notch signaling, renowned for its role in regulating cell fate, organ development, and tissue homeostasis across metazoans, is highly conserved throughout evolution. The Notch receptor and its ligands are transmembrane proteins containing epidermal growth factor-like repeat sequences, typically necessitating receptor-ligand interaction to initiate classical Notch signaling transduction. Accumulating evidence indicates that the Notch signaling pathway serves as both an oncogenic factor and a tumor suppressor in various cancer types. Dysregulation of this pathway promotes epithelial-mesenchymal transition and angiogenesis in malignancies, closely linked to cancer proliferation, invasion, and metastasis. Furthermore, the Notch signaling pathway contributes to maintaining stem-like properties in cancer cells, thereby enhancing cancer invasiveness. The regulatory role of the Notch signaling pathway in cancer metabolic reprogramming and the tumor microenvironment suggests its pivotal involvement in balancing oncogenic and tumor suppressive effects. Moreover, the Notch signaling pathway is implicated in conferring chemoresistance to tumor cells. Therefore, a comprehensive understanding of these biological processes is crucial for developing innovative therapeutic strategies targeting Notch signaling. This review focuses on the research progress of the Notch signaling pathway in cancers, providing in-depth insights into the potential mechanisms of Notch signaling regulation in the occurrence and progression of cancer. Additionally, the review summarizes pharmaceutical clinical trials targeting Notch signaling for cancer therapy, aiming to offer new insights into therapeutic strategies for human malignancies.
Collapse
Affiliation(s)
- Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shuwen Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jinzhi Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yaqi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
2
|
Ji L, She Q, Zhou P, Qin Y. Butorphanol inhibits ferroptosis to attenuate PC12 cell injury by blocking JNK/p38 signaling. Exp Ther Med 2024; 27:8. [PMID: 38223326 PMCID: PMC10785043 DOI: 10.3892/etm.2023.12295] [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: 04/06/2023] [Accepted: 08/16/2023] [Indexed: 01/16/2024] Open
Abstract
Butorphanol is a synthetic selective opioid receptor antagonist that exhibits substantial analgesic effects. The present study aimed to explore the effects of butorphanol on a neurodegenerative disease cell model and to investigate its specific regulatory mechanism. Cell viability of PC12 cells was assessed using the Cell Counting Kit-8 assay. Oxidative stress levels were measured by the corresponding kits and western blotting of specific protein markers. Apoptosis was determined using the terminal-deoxynucleoitidyl transferase mediated nick end labeling assay and by western blotting. Western blotting was used to analyze the expression levels of c-Jun NH2-terminal kinase (JNK)/p38 signaling pathway-related proteins. Thiobarbituric acid-reactive substances and Fe+2 content were detected using the corresponding assay kits and the expression levels of ferroptosis-associated proteins were assessed by western blotting following the addition of the JNK activator anisomycin (ANI). Oxidative stress and apoptosis were examined with the aforementioned assays following the supplementation of ANI or the ferroptosis inducer erastin (ERA). It was revealed that butorphanol dose-dependently enhanced the viability and suppressed the oxidative stress and apoptosis of H2O2-treated PC12 cells. In addition, butorphanol blocked JNK/p38 signaling and hampered ferroptosis, while this effect was reversed by ANI. ANI or ERA reversed the effects of butorphanol on oxidative stress and apoptosis of H2O2-treated PC12 cells. In summary, butorphanol suppressed ferroptosis by blocking JNK/p38 signaling to impart inhibitory effects on oxidative stress and apoptosis in a neurodegenerative disease cell model.
Collapse
Affiliation(s)
- Lulu Ji
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qing She
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ping Zhou
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yibin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
3
|
Danielpour D, Corum S, Leahy P, Bangalore A. Jagged-1 is induced by mTOR inhibitors in renal cancer cells through an Akt/ALK5/Smad4-dependent mechanism. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100117. [PMID: 35992379 PMCID: PMC9389240 DOI: 10.1016/j.crphar.2022.100117] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) plays an important role in the aggressiveness and therapeutic resistance of many cancers. Targeting mTOR continues to be under clinical investigation for cancer therapy. Despite the notable clinical success of mTOR inhibitors in extending the overall survival of patients with certain malignancies including metastatic renal cell carcinomas (RCCs), the overall impact of mTOR inhibitors on cancers has been generally disappointing and attributed to various compensatory responses. Here we provide the first report that expression of the Notch ligand Jagged-1 (JAG1), which is associated with aggressiveness of RCCs, is induced by several inhibitors of mTOR (rapamycin (Rap), BEZ235, KU-0063794) in human clear cell RCC (ccRCC) cells. Using both molecular and chemical inhibitors of PI3K, Akt, and TGF-β signaling, we provide evidence that the induction of JAG1 expression by mTOR inhibitors in ccRCC cells depends on the activation of Akt and occurs through an ALK5 kinase/Smad4-dependent mechanism. Furthermore, we show that mTOR inhibitors activate Notch1 and induce the expression of drivers of epithelial-mesenchymal transition, notably Hic-5 and Slug. Silencing JAG1 with selective shRNAs blocked the ability of KU-0063794 and Rap to induce Hic-5 in ccRCC cells. Moreover, Rap enhanced TGF-β-induced expression of Hic-5 and Slug, both of which were repressed in JAG1-silenced ccRCC cells. Silencing JAG1 selectively decreased the motility of ccRCC cells treated with Rap or TGF-β1. Moreover, inhibition of Notch signaling with γ-secretase inhibitors enhanced or permitted mTOR inhibitors to suppress the motility of ccRCC cells. We suggest targeting JAG1 may enhance therapeutic responses to mTOR inhibitors in ccRCCs.
Collapse
Key Words
- ALK5, Activin-like kinase 5 (TGF-β type I receptor)
- ANOVA, Analysis of variance
- Akt
- BEZ235
- BSA, Bovine serum albumin
- EDTA, Ethylenediaminetetraacetic acid
- EMT
- FBS, Fetal bovine serum
- Hic-5
- Hic-5, Hydrogen peroxide-inducible clone 5, also known as transforming growth factor beta induced transcript
- IRS-1, Insulin receptor substrate-1
- JAG1, Jagged-1
- KU-0063794
- MAML-1, Mastermind-like protein-1
- Myr, Myristoylated
- PI3K
- PI3K, Phosphatidylinositol 3-kinase
- RCC, RCC
- Rap, Rapamycin
- Rapamycin
- Renal cancer
- Rheb, Ras homologue enriched in brain
- SE, Standard error
- Slug
- Slug, Snail family of transcription factors encoded by the SNAI2 gene
- Smad, Mothers against decapentaplegic homolog
- Smad4
- TGF-beta
- TGF-β, Transforming growth factor-beta
- TSC, Tuberous Sclerosis Complex
- TβRI, Transforming growth factor β receptor type 1
- TβRII, Transforming growth factor β receptor type 2
- ccRCC, Clear cell renal cell carcinoma
- mRCC, Metastatic renal cell carcinoma
- mTOR
- mTORC1, Mammalian target of rapamycin complex 1
- mTORC2, Mammalian target of rapamycin complex 2
Collapse
Affiliation(s)
- David Danielpour
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Pharmacology Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Urology University Hospitals of Cleveland, Cleveland, OH, 44106, USA
| | - Sarah Corum
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Patrick Leahy
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Anusha Bangalore
- Case Comprehensive Cancer Center Research Laboratories, The Division of General Medical Sciences-Oncology Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
4
|
Wang S, Hao X, He S, Liu C, Wang Q. Suppressive effects of RASAL2 on renal cell carcinoma via SOX2/ERK/p38 MAPK pathway. Exp Ther Med 2020; 20:151. [PMID: 33093889 PMCID: PMC7571372 DOI: 10.3892/etm.2020.9280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
Metastatic renal cell carcinoma (RCC) is associated with poor prognosis. Ras protein activator like 2 (RASAL2) protein has been previously demonstrated to serves as a tumor suppressor in a variety of malignancies. Therefore, the aim of the present study was to investigate the role of RASAL2 in RCC. Reverse transcription-quantitative PCR, western blot analysis and immunohistochemistry were performed to measure mRNA and protein expression in RCC tissues, whilst immunofluorescence and western blotting were performed to evaluate protein expression in RCC cells. A Cell Counting Kit-8 and 5-bromo-2'-deoxyuridine staining were applied to determine cell viability, and Transwell assays were conducted to measure RCC cell invasion and migration. RASAL2 expression was identified to be downregulated in RCC tissues, which associate negatively with RCC pathological grade. Sox2 expression, in addition to ERK1/2 and p38 MAPK phosphorylation, were demonstrated to be increased in RCC tissues. In RCC cells, RASAL2 overexpression decreased the expression of Sox2 and the activation of ERK1/2 and p38 MAPK. Physiologically, RASAL2 overexpression decreased RCC cell viability, invasion and migration. The expression of metalloproteinase-2/9 and tissue inhibitor of metalloproteinase 1 were also identified to be decreased and increased by RASAL2 overexpression, respectively. By contrast, RASAL2 knockdown exerted opposite effects on RCC cells compared with those observed following RASAL2 overexpression. RASAL2 expression decreased RCC cell viability, migration and invasion, which was demonstrated to be associated with the inactivation of SOX2/ERK1/2/p38 MAPK signaling. These results suggest that RASAL2 may potentially serve as a potential target for the development of novel therapeutic intervention strategies against RCC.
Collapse
Affiliation(s)
- Sen Wang
- Department of Urinary Surgery, Shaanxi Friendship Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xiaomin Hao
- Department of Internal Medicine, Shaanxi Province Tuberculosis Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Sai He
- Department of Breast Cancer, Shaanxi Province Oncology Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Changli Liu
- Department of Internal Medicine, Shaanxi Province Tuberculosis Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Qilong Wang
- Department of General Surgery, Shaanxi Friendship Hospital, Xi'an, Shaanxi 710068, P.R. China
| |
Collapse
|
5
|
Qiu PC, Lu YY, Zhang S, Li H, Bao H, Ji YQ, Fang F, Tang HF, Cheng G. Reduction of SCUBE3 by a new marine-derived asterosaponin leads to arrest of glioma cells in G1/S. Oncogenesis 2020; 9:71. [PMID: 32764572 PMCID: PMC7411020 DOI: 10.1038/s41389-020-00252-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/18/2020] [Accepted: 06/30/2020] [Indexed: 12/31/2022] Open
Abstract
Many saponins are characterized as exhibiting a wide spectrum of antitumor activities at low concentrations. Most of the previous studies that aimed to understand the mechanisms underlying anticancer saponins have focused on numerous classical signaling pathways. However, at the oncogene level, little is known about the action of saponins, especially asterosaponin. In this study, CN-3, a new asterosaponin isolated from the starfish Culcita novaeguineae, decreased the proliferation of U87 and U251 cells at low doses in a dose- and time-dependent manner. Microarray analysis revealed CN-3 significantly induced the differential expression of 661 genes that are related to its antiglioma effect in U251. Nine downregulated genes (SCUBE3, PSD4, PGM2L1, ACSL3, PRICKLE1, ABI3BP, STON1, EDIL3, and KCTD12) were selected, for further verification of their low expression. Then, shRNA transfection and high-content screening were performed and significantly decreased U251 cell proliferation rate was only observed for the SCUBE3 knockdown. qPCR confirmed SCUBE3 was highly expressed in U251 and U87 cells, and had medium expression levels in U373 cells. Real-time cellular analysis using iCELLigence demonstrated that SCUBE3 is an oncogene in U251 and U87 cells, with knockdown of SCUBE3 inhibiting U251 and U87 cell proliferation while, conversely, SCUBE3 overexpression promoted their proliferation. Afterward, SCUBE3 protein was found to have high expression in primary glioma specimens from patients examined by immunohistochemistry but low expression in normal brain. PathScan ELISA analysis in conjunction with TEM observation demonstrated that the effect of SCUBE3 knockdown in U251 does not appear to be related to the induction of apoptosis. Employing CCK-8, iCELLigence, flow cytometry, western blotting, and shRNA transfection (knockdown and overexpression) experiments, we reveal that the reduction of SCUBE3 expression, induced by CN-3, mediated both inhibition and G1/S arrest of U251 via the Akt/p-Akt/p53/p21/p27/E2F1 pathway.
Collapse
Affiliation(s)
- Peng-Cheng Qiu
- Institute of Materia Medica, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, 710032, Xi'an, People's Republic of China
| | - Yun-Yang Lu
- Institute of Materia Medica, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, 710032, Xi'an, People's Republic of China
| | - Shan Zhang
- Institute of Materia Medica, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, 710032, Xi'an, People's Republic of China.,School of Pharmacy, Shaanxi University of Chinese Medicine, 712046, Xianyang, People's Republic of China
| | - Hua Li
- Institute of Materia Medica, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, 710032, Xi'an, People's Republic of China
| | - Han Bao
- Institute of Materia Medica, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, 710032, Xi'an, People's Republic of China
| | - Yu-Qiang Ji
- Central Laboratory of Xi'an No.1 Hospital, 710002, Xi'an, People's Republic of China
| | - Fei Fang
- Central Laboratory of Xi'an No.1 Hospital, 710002, Xi'an, People's Republic of China
| | - Hai-Feng Tang
- Institute of Materia Medica, Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, School of Pharmacy, Air Force Medical University, 710032, Xi'an, People's Republic of China.
| | - Guang Cheng
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Air Force Medical University, 710032, Xi'an, People's Republic of China.
| |
Collapse
|
6
|
Notch1 promotes mouse spinal neural stem and progenitor cells proliferation via p-p38-pax6 induced cyclin D1 activation. Exp Cell Res 2018; 373:80-90. [DOI: 10.1016/j.yexcr.2018.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/09/2018] [Accepted: 09/29/2018] [Indexed: 02/06/2023]
|
7
|
Cai Z, Zhao B, Deng Y, Shangguan S, Zhou F, Zhou W, Li X, Li Y, Chen G. Notch signaling in cerebrovascular diseases (Review). Mol Med Rep 2016; 14:2883-98. [PMID: 27574001 PMCID: PMC5042775 DOI: 10.3892/mmr.2016.5641] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 07/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Notch signaling pathway is a crucial regulator of numerous fundamental cellular processes. Increasing evidence suggests that Notch signaling is involved in inflammation and oxidative stress, and thus in the progress of cerebrovascular diseases. In addition, Notch signaling in cerebrovascular diseases is associated with apoptosis, angiogenesis and the function of blood-brain barrier. Despite the contradictory results obtained to date as to whether Notch signaling is harmful or beneficial, the regulation of Notch signaling may provide a novel strategy for the treatment of cerebrovascular diseases.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Bin Zhao
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanqing Deng
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Shouqin Shangguan
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Faming Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Wenqing Zhou
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiaoli Li
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Yanfeng Li
- Department of Neurology, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Guanghui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| |
Collapse
|