1
|
Li C, Xiao N, Song W, Lam AHC, Liu F, Cui X, Ye Z, Chen Y, Ren P, Cai J, Lee ACY, Chen H, Ou Z, Chan JFW, Yuen KY, Chu H, Zhang AJX. Chronic lung inflammation and CK14+ basal cell proliferation induce persistent alveolar-bronchiolization in SARS-CoV-2-infected hamsters. EBioMedicine 2024; 108:105363. [PMID: 39326207 PMCID: PMC11470415 DOI: 10.1016/j.ebiom.2024.105363] [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: 05/21/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Post-acute sequalae of COVID-19 defines a wide range of ongoing symptoms and conditions long after SARS-CoV-2 infection including respiratory diseases. The histopathological changes in the lung and underlying mechanism remain elusive. METHODS We investigated lung histopathological and transcriptional changes in SARS-CoV-2-infected male hamsters at 7, 14, 42, 84 and 120dpi, and compared with A (H1N1)pdm09 infection. FINDINGS We demonstrated viral residue, inflammatory and fibrotic changes in lung after SARS-CoV-2 but not H1N1 infection. The most prominent histopathological lesion was multifocal alveolar-bronchiolization observed in every SARS-CoV-2 infected hamster (31/31), from 42dpi to 120dpi. Proliferating (Ki67+) CK14+ basal cells accumulated in alveoli adjacent to bronchioles at 7dpi, where they proliferated and differentiated into SCGB1A+ club cell or Tubulin+ ciliated cells forming alveolar-bronchiolization foci. Molecularly, Notch pathway significantly upregulated with intensive Notch3 and Hes1 protein expression in alveolar-bronchiolization foci at 42 and 120dpi, suggesting Notch signaling involving the persistence of alveolar-bronchiolization. This is further demonstrated by spatial transcriptomic analysis. Intriguingly, significant upregulation of some cell-growth promoting pathways and genes such as Tubb4b, Stxbp4, Grb14 and Mlf1 were spatially overlapping with bronchiolization lesion. INTERPRETATION Incomplete resolution of SARS-CoV-2 infection in lung with viral residue, chronic inflammatory and fibrotic damage and alveolar-bronchiolization impaired respiratory function. Aberrant activation of CK14+ basal cells during tissue regeneration led to persistent alveolar-bronchiolization due to sustained Notch signaling. This study advances our understanding of respiratory PASC, sheds light on disease management and highlights the necessity for monitoring disease progression in people with respiratory PASC. FUNDING Funding is listed in the Acknowledgements section.
Collapse
Affiliation(s)
- Can Li
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Na Xiao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wenchen Song
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Alvin Hiu-Chung Lam
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Feifei Liu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | - Zhanhong Ye
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Yanxia Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | | | - Jianpiao Cai
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Andrew Chak-Yiu Lee
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Honglin Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | | | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China.
| | - Anna Jin-Xia Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong SAR, China.
| |
Collapse
|
2
|
Paredes de la Fuente R, Sucre S, Ponce C, Rattani AAA, Peters MLB. Somatic Mutation Profile as a Predictor of Treatment Response and Survival in Unresectable Pancreatic Ductal Adenocarcinoma Treated with FOLFIRINOX and Gemcitabine Nab-Paclitaxel. Cancers (Basel) 2024; 16:2734. [PMID: 39123462 PMCID: PMC11312283 DOI: 10.3390/cancers16152734] [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: 07/11/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
(1) Background: Pancreatic ductal adenocarcinoma (PDAC) has low survival rates despite treatment advancements. Aim: This study aims to show how molecular profiling could possibly guide personalized treatment strategies, which may help improve survival outcomes in patients with PDAC. (2) Materials and Methods: A retrospective analysis of 142 PDAC patients from a single academic center was conducted. Patients underwent chemotherapy and next-generation sequencing for molecular profiling. Key oncogenic pathways were identified using the Reactome pathway database. Survival analysis was performed using Kaplan-Meier curves and Cox Proportional Hazards Regression. (3) Results: Patients mainly received FOLFIRINOX (n = 62) or gemcitabine nab-paclitaxel (n = 62) as initial chemotherapy. The median OS was 13.6 months. Longer median OS was noted in patients with NOTCH (15 vs. 12.3 months, p = 0.007) and KIT pathway mutations (21.3 vs. 12.12 months, p = 0.04). Combinatorial pathway analysis indicated potential synergistic effects on survival. In the PFS, PI3K pathway (6.6 vs. 5.7 months, p = 0.03) and KIT pathway (10.3 vs. 6.2 months, p = 0.03) mutations correlated with improved PFS within the gemcitabine nab-paclitaxel subgroup. (4) Conclusions: Molecular profiling could play a role in PDAC for predicting outcomes and responses to therapies like FOLFIRINOX and gemcitabine nab-paclitaxel. Integrating genomic data into clinical decision-making can benefit PDAC treatment, though further validation is needed to fully utilize precision oncology in PDAC management.
Collapse
Affiliation(s)
| | - Santiago Sucre
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| | - Cristina Ponce
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| | - Ahmed Anwer Ali Rattani
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| | - Mary Linton B. Peters
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA (A.A.A.R.)
| |
Collapse
|
3
|
Liu H, Wang L, Shi X, Yin L, Zhai W, Gao S, Chen Y, Zhang T. Calcium saccharate/DUSP6 suppresses renal cell carcinoma glycolytic metabolism and boosts sunitinib efficacy via the ERK-AKT pathway. Biochem Pharmacol 2024; 224:116247. [PMID: 38697311 DOI: 10.1016/j.bcp.2024.116247] [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: 12/12/2023] [Revised: 04/13/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Current therapeutic options for renal cell carcinoma (RCC) are very limited, which is largely due to inadequate comprehension of molecular pathological mechanisms as well as RCC's resistance to chemotherapy. Dual-specificity phosphatase 6 (DUSP6) has been associated with numerous human diseases. However, its role in RCC is not well understood. Here, we show that diminished DUSP6 expression is linked to RCC progression and unfavorable prognosis. Mechanistically, DUSP6 serves as a tumor suppressor in RCC by intervening the TAF10 and BSCL2 via the ERK-AKT pathway. Further, DUSP6 is also transcriptionally regulated by HNF-4a. Moreover, docking experiments have indicated that DUSP6 expression is enhanced when bound by Calcium saccharate, which also inhibits RCC cell proliferation, metabolic rewiring, and sunitinib resistance. In conclusion, our study identifies Calcium saccharate as a prospective pharmacological therapeutic approach for RCC.
Collapse
Affiliation(s)
- Huan Liu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Longsheng Wang
- Department of Urology, Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan, China; Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaokai Shi
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Lei Yin
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenglin Gao
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China; Department of Urology, Gonghe County Hospital of Traditional Chinese Medicine, Qinghai, China; Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| | - Yonghui Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Tao Zhang
- Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China..
| |
Collapse
|
4
|
Shen T, Shi J, Zhao X, Fu L, Wang N, Zheng X, Chen Y, Li M, Ma C, Liu P, Zhu D. Presenilin 1 Is a Therapeutic Target in Pulmonary Hypertension and Promotes Vascular Remodeling. Am J Respir Cell Mol Biol 2024; 70:468-481. [PMID: 38381098 DOI: 10.1165/rcmb.2022-0426oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/20/2024] [Indexed: 02/22/2024] Open
Abstract
Small muscular pulmonary artery remodeling is a dominant feature of pulmonary arterial hypertension (PAH). PSEN1 affects angiogenesis, cancer, and Alzheimer's disease. We aimed to determine the role of PSEN1 in the pathogenesis of vascular remodeling in pulmonary hypertension (PH). Hemodynamics and vascular remodeling in the Psen1-knockin and smooth muscle-specific Psen1-knockout mice were assessed. The functional partners of PSEN1 were predicted by bioinformatics analysis and biochemical experiments. The therapeutic effect of PH was evaluated by administration of the PSEN1-specific inhibitor ELN318463. We discovered that both the mRNA and protein levels of PSEN1 were increased over time in hypoxic rats, monocrotaline rats, and Su5416/hypoxia mice. Psen1 transgenic mice were highly susceptible to PH, whereas smooth muscle-specific Psen1-knockout mice were resistant to hypoxic PH. STRING analysis showed that Notch1/2/3, β-catenin, Cadherin-1, DNER (delta/notch-like epidermal growth factor-related receptor), TMP10, and ERBB4 appeared to be highly correlated with PSEN1. Immunoprecipitation confirmed that PSEN1 interacts with β-catenin and DNER, and these interactions were suppressed by the catalytic PSEN1 mutations D257A, D385A, and C410Y. PSEN1 was found to mediate the nuclear translocation of the Notch1 intracellular domains and activated RBP-Jκ. Octaarginine-coated liposome-mediated pharmacological inhibition of PSEN1 significantly prevented and reversed the pathological process in hypoxic and monocrotaline-induced PH. PSEN1 essentially drives the pathogenesis of PAH and interacted with the noncanonical Notch ligand DNER. PSEN1 can be used as a promising molecular target for treating PAH. PSEN1 inhibitor ELN318463 can prevent and reverse the progression of PH and can be developed as a potential anti-PAH drug.
Collapse
Affiliation(s)
- TingTing Shen
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - JiuCheng Shi
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - XiJuan Zhao
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - Li Fu
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - Na Wang
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - XiaoDong Zheng
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - YingLi Chen
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - MingHui Li
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - Cui Ma
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - PiXu Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - DaLing Zhu
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
- College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Ning X, Fu Z, Zhang J, Gao S, Cui Z, Cong M, Guo Q, Sun X, Li J, Zhang M, Wang S. The role of alternative splicing in lung cancer. Cancer Chemother Pharmacol 2023; 92:83-95. [PMID: 37335335 DOI: 10.1007/s00280-023-04553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 06/12/2023] [Indexed: 06/21/2023]
Abstract
Aberrant alternative splicing (AS) events are frequently observed in lung cancer, which can be attributed to aberrant gene AS, alterations in splicing regulatory factors, or changes in splicing regulatory mechanisms. Consequently, the dysregulation of alternative RNA splicing is the fundamental cause of lung cancer. In this review, we have summarized the pivotal role of AS in the development, progression, invasion, metastasis, angiogenesis, and drug resistance of lung cancer. Ultimately, this review emphasizes the potential of AS as biomarkers in lung cancer prognosis and diagnosis, and introduces some applications of AS isoform in the treatment of lung cancer. The comprehension of the AS may provide a glimmer of hope for the eradication of lung cancer.
Collapse
Affiliation(s)
- Xuelian Ning
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Zitong Fu
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Jing Zhang
- Department of Oncology, Chifeng Municipal Hospital, No.1 Zhaowuda Road, Chifeng, 024000, China
| | - Shuangshu Gao
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Zihan Cui
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Mingqi Cong
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Qingyu Guo
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Xixi Sun
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Jing Li
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China
| | - Minghui Zhang
- Department of Oncology, Chifeng Municipal Hospital, No.1 Zhaowuda Road, Chifeng, 024000, China.
| | - Shuoshuo Wang
- Department of Pathology, Harbin Medical University, No.157 Baojian Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
6
|
Zhang C, Qin S, Xie H, Qiu Q, Wang H, Zhang J, Luo D, Zhang J. RO4929097, a Selective γ-Secretase Inhibitor, Inhibits Subretinal Fibrosis Via Suppressing Notch and ERK1/2 Signaling in Laser-Induced Mouse Model. Invest Ophthalmol Vis Sci 2022; 63:14. [PMID: 36155746 PMCID: PMC9526367 DOI: 10.1167/iovs.63.10.14] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study aimed to explore whether RO4929097 (RO), a specific γ-secretase inhibitor, could inhibit the subretinal fibrosis in laser-induced mouse model and the relevant molecular mechanisms. Methods Male C57BL/6J mice were used to produce choroidal neovascularization (CNV) and subretinal fibrosis by laser photocoagulation, and RO was administered intravitreally 1 day after laser induction. The sizes of CNV and subretinal fibrosis were measured and quantified in both 2D and 3D constructions. The ARPE-19 cell line and primary human RPE (phRPE) cells were treated with TGFβ1, in combination with or without RO, to examine Notch related molecules, epithelial mesenchymal transition (EMT), cell viability, migration, and contractile function, as well as the crosstalk between Notch and other EMT relevant signaling pathways. Results Intravitreal injection of RO reduced the sizes of both CNV and subretinal fibrosis in laser-induced young and old mice at day 7 and day 14 after laser induction. Moreover, EMT and Notch activation in RPE-choroid complexes from laser-induced mice were significantly attenuated by RO. In vitro, TGFβ1 activated Notch signaling and induced EMT in ARPE-19 cells, accompanied by enhanced EMT-related function, which were inhibited by RO. The inhibition of RO on EMT was further confirmed in TGFβ1-treated phRPE cells. Blockage of Notch signaling by RO could inhibit ERK1/2 signaling; whereas ERK1/2 inhibition had no effect on Notch. The action of RO was independent of Smad2/3 or p38, and co-inhibition of Notch and Smad2/3 showed synergistic effect on EMT inhibition. Conclusions RO exerts its antifibrotic effect by directly inhibiting Notch signaling and indirectly suppressing ERK1/2 signaling. Targeting Notch signaling might provide a therapeutic strategy in prevention and treatment of subretinal fibrosis in neovascular age-related macular degeneration (nAMD).
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Shiyue Qin
- Department of Ophthalmology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai Xie
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Qinghua Qiu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.,Department of Ophthalmology, Shigatse People's Hospital, Xizang, China
| | - Haiyan Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
7
|
Pongjantarasatian S, Nowwarote N, Rotchanakitamnuai V, Srirodjanakul W, Saehun R, Janebodin K, Manokawinchoke J, Fournier BPJ, Osathanon T. A γ-Secretase Inhibitor Attenuates Cell Cycle Progression and Invasion in Human Oral Squamous Cell Carcinoma: An In Vitro Study. Int J Mol Sci 2022; 23:8869. [PMID: 36012128 PMCID: PMC9408752 DOI: 10.3390/ijms23168869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 12/13/2022] Open
Abstract
Notch signaling is associated with many human malignancies, including oral squamous cell carcinoma (OSCC). However, the exact function of Notch signaling in OSCC remains unclear. Here, we investigated the effect of Notch signaling inhibition using a γ-secretase inhibitor (DAPT) on OSCC behaviours in vitro. Bioinformatic analysis of public-available gene expression profiles revealed the dysregulation of the Notch signaling pathway in OSCC compared with normal tissues, indicating the role of Notch signaling in OSCC regulation. RNA sequencing analysis of DAPT-treated human OSCC cells revealed the dysregulation of genes related to cell cycle-related pathways. Blocking Notch signaling significantly inhibited cell proliferation. DAPT-induced G0/G1 cell cycle arrest induced cell apoptosis. Furthermore, cell migration and invasion were also reduced in DAPT-treated cells. These findings indicate that Notch signaling activation participates in OSCC regulation by promoting cell growth, cell cycle progression, cell migration, and invasion. These mechanisms could facilitate OSCC progression. These results imply the potential use of Notch signaling inhibitors as a candidate adjuvant treatment in OSCC patients.
Collapse
Affiliation(s)
- Sarai Pongjantarasatian
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nunthawan Nowwarote
- Department of Oral Biology, Faculty of Dentistry, Universite Paris Cite, 75006 Paris, France
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Universite Paris Cite, Sorbonne Universite, 75006 Paris, France
| | - Varumporn Rotchanakitamnuai
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watcharee Srirodjanakul
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ritmongkol Saehun
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kajohnkiart Janebodin
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Jeeranan Manokawinchoke
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Benjamin P. J. Fournier
- Department of Oral Biology, Faculty of Dentistry, Universite Paris Cite, 75006 Paris, France
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Universite Paris Cite, Sorbonne Universite, 75006 Paris, France
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
8
|
Sanders BE, Yamamoto TM, McMellen A, Woodruff ER, Berning A, Post MD, Bitler BG. Targeting DUSP Activity as a Treatment for High-Grade Serous Ovarian Carcinoma. Mol Cancer Ther 2022; 21:1285-1295. [PMID: 35587258 PMCID: PMC9357222 DOI: 10.1158/1535-7163.mct-21-0682] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/21/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023]
Abstract
Identifying novel, durable treatments for high-grade serous ovarian cancer (HGSOC) is paramount to extend both progression-free survival (PFS) and overall survival (OS) in patients afflicted with this disease. Dual-specificity phosphatase 1 (DUSP1) was identified as one of seven genes that may significantly affect prognosis in patients with HGSOC; however, the role of DUSP inhibition (DUSPi) in the treatment of HGSOC remains largely unknown. In this study, we show that DUSP1 is highly expressed in HGSOC and confers worse PFS and OS. Further, we corroborate data that show DUSP1 expression is directly associated with therapy resistance. Using a tissue microarray of 137 different serous ovarian carcinomas, we demonstrate the high expression of DUSP1 in primary and recurrent serous ovarian cancer. In both acquired and de novo therapy HGSOC-resistant models, DUSPi both inhibited cellular proliferation and promoted cell death. RPPA analysis of HGSOC cells revealed DUSPi led to the differential regulation of several pathways, including AMPK and mTORC. Further, in a patient-derived xenograft HGSOC model, DUSPi significantly inhibited tumor progression.
Collapse
Affiliation(s)
- Brooke E. Sanders
- Department of Obstetrics & Gynecology, Division of
Gynecologic Oncology, The University of Colorado Anschutz Medical Campus, Aurora, CO
80045, USA
- University of Colorado Comprehensive Cancer Center, Aurora,
CO 80045, USA
| | - Tomomi M. Yamamoto
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| | - Alexandra McMellen
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| | - Elizabeth R. Woodruff
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| | - Amber Berning
- Department of Pathology, The University of Colorado
Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Miriam D. Post
- Department of Pathology, The University of Colorado
Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin G. Bitler
- University of Colorado Comprehensive Cancer Center, Aurora,
CO 80045, USA
- Department of Obstetrics & Gynecology, Division of
Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora,
CO 80045, USA
| |
Collapse
|
9
|
Rohatgi A, Govindan R. Targeting KRAS G12C mutation in lung adenocarcinoma. Lung Cancer 2022; 165:28-33. [PMID: 35066360 DOI: 10.1016/j.lungcan.2021.12.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/24/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023]
Abstract
Lung cancer continues to be a major cause of cancer related death globally. Therapies targeting driver mutations have significantly extended the survival of patients whose lung cancer cells harbor these mutations. Patients with KRAS mutations, however, lacked specific targeted therapy until the recent FDA approval of sotorasib, a specific inhibitor of KRAS G12C mutant protein. We will discuss the efficacy and toxicities of the novel KRAS G12C inhibitors as well as other indirect strategies for targeting oncogenic KRAS mutations. We will review the limited literature on acquired resistance to these inhibitors and the novel combinatorial treatment strategies that are being tested currently in clinical trials.
Collapse
Affiliation(s)
- Anjali Rohatgi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, United States; Alvin J. Siteman Cancer Center, St Louis, MO, United States
| | - Ramaswamy Govindan
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, United States; Alvin J. Siteman Cancer Center, St Louis, MO, United States.
| |
Collapse
|
10
|
Logeay R, Géminard C, Lassus P, Rodríguez-Vázquez M, Kantar D, Heron-Milhavet L, Fischer B, Bray SJ, Colinge J, Djiane A. Mechanisms underlying the cooperation between loss of epithelial polarity and Notch signaling during neoplastic growth in Drosophila. Development 2022; 149:274230. [PMID: 35005772 DOI: 10.1242/dev.200110] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/17/2021] [Indexed: 12/16/2022]
Abstract
Aggressive neoplastic growth can be initiated by a limited number of genetic alterations, such as the well-established cooperation between loss of cell architecture and hyperactive signaling pathways. However, our understanding of how these different alterations interact and influence each other remains very incomplete. Using Drosophila paradigms of imaginal wing disc epithelial growth, we have monitored the changes in Notch pathway activity according to the polarity status of cells (scrib mutant). We show that the scrib mutation impacts the direct transcriptional output of the Notch pathway, without altering the global distribution of Su(H), the Notch-dedicated transcription factor. The Notch-dependent neoplasms require, however, the action of a group of transcription factors, similar to those previously identified for Ras/scrib neoplasm (namely AP-1, Stat92E, Ftz-F1 and basic leucine zipper factors), further suggesting the importance of this transcription factor network during neoplastic growth. Finally, our work highlights some Notch/scrib specificities, in particular the role of the PAR domain-containing basic leucine zipper transcription factor and Notch direct target Pdp1 for neoplastic growth.
Collapse
Affiliation(s)
- Rémi Logeay
- IRCM, Inserm, University of Montpellier, ICM, Montpellier, France
| | - Charles Géminard
- IRCM, Inserm, University of Montpellier, ICM, Montpellier, France
| | - Patrice Lassus
- IRCM, Inserm, University of Montpellier, ICM, CNRS, Montpellier, France
| | | | - Diala Kantar
- IRCM, Inserm, University of Montpellier, ICM, Montpellier, France
| | | | - Bettina Fischer
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Sarah J Bray
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Jacques Colinge
- IRCM, Inserm, University of Montpellier, ICM, Montpellier, France
| | - Alexandre Djiane
- IRCM, Inserm, University of Montpellier, ICM, Montpellier, France
| |
Collapse
|
11
|
Bai H, Song M, Jiao R, Li W, Zhao J, Xiao M, Jin M, Zhang Z, Deng H. DUSP7 inhibits cervical cancer progression by inactivating the RAS pathway. J Cell Mol Med 2021; 25:9306-9318. [PMID: 34435746 PMCID: PMC8500958 DOI: 10.1111/jcmm.16865] [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: 03/13/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 12/13/2022] Open
Abstract
To determine the differentially expressed proteins (DEPs) between paired samples of cervical cancer (CC) and paracancerous tissue by quantitative proteomics and to examine the effects of DUSP7 expression on the tumorigenesis and progression of CC. Proteomic profiles of three paired samples of CC and paracancerous tissue were quantitatively analysed to identify DEPs. The relationship between DEP expression and patient clinicopathological characteristics and prognosis was evaluated. The effects of the selected DEPs on CC progression were examined in SIHA cells. A total of 129 DEPs were found. Western blot and immunohistochemistry (IHC) staining analyses confirmed the results from quantitative proteomic analysis showing that the selected DEP, HRAS, P-ERK1/2, and PLD1 levels were increased, whereas the DUSP7 level was decreased in CC tissue compared with the paired normal paracancerous tissues. The IHC results from the CC TMA analysis showed that the decreased expression of DUSP7 (p = 0.045 and 0.044) was significantly associated with a tumour size >2 cm and parametrial infiltration. In addition, the decreased expression of DUSP7 and increased expression of p-ERK1/2 were adversely related to patient relapse (p = 0.003 and 0.001) and survival (p = 0.034 and 0.006). The expression of HRAS and p-ERK1/2 was decreased in DUSP7-SIHA cells compared with NC-SIHA cells (p = 0.0003 and 0.0026). Biological functions in vitro, including invasion, migration and proliferation and tumour formation in vivo were decreased in DUSP7-SIHA cells (all p < 0.05) but increased in shDUSP7-SIHA cells (all p < 0.05). DUSP7 inhibits cervical cancer progression by inactivating the RAS pathway.
Collapse
Affiliation(s)
- Huimin Bai
- Department of Obstetrics and Gynecology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Meiying Song
- Department of Obstetrics and Gynecology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China.,Department of Obstetrics and Gynecology, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Ruili Jiao
- Department of Obstetrics and Gynecology, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing, China
| | - Weihua Li
- Department of Obstetrics and Gynecology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Jing Zhao
- Department of Obstetrics and Gynecology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Meizhu Xiao
- Department of Obstetrics and Gynecology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Mulan Jin
- Department of Pathology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Zhengyu Zhang
- Department of Obstetrics and Gynecology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Zhu Y, Wang H, Fei M, Tang T, Niu W, Zhang L. Smarcd1 Inhibits the Malignant Phenotypes of Human Glioblastoma Cells via Crosstalk with Notch1. Mol Neurobiol 2021; 58:1438-1452. [PMID: 33190170 PMCID: PMC7932991 DOI: 10.1007/s12035-020-02190-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 10/29/2020] [Indexed: 11/29/2022]
Abstract
Smarcd1 is a component of an evolutionary conserved chromatin remodeling complex-SWI/SNF, which is involved in transcription factor recruitment, DNA replication, recombination, and repair. Suppression of the SWI/SNF complex required for cellular differentiation and gene regulation may be inducible for cell proliferation and tumorigenicity. However, the inhibitory role of Smarcd1 in human glioblastoma cells has not been well illustrated. Both U87 and U251 human glioblastoma cell lines were employed in the present study. The lentivirus-mediated gene knockdown and overexpression approach was conducted to determine the function of Smarcd1. The protein levels were tested by western blot, and the relative mRNA contents were detected by quantitative real-time PCR. Cell viability was tested by CCK-8 and colony-forming assay. Transwell assays were utilized to evaluate the motility and invasive ability. Flow cytometry was employed to analyze cell cycle and apoptosis. SPSS software was used for statistical analysis. Low expression of Smarcd1 was observed in glioblastoma cell lines and in patients with high-grade glioma. Importantly, the depletion of Smarcd1 promoted cell proliferation, invasion, and chemoresistance, whereas enhanced expression of Smarcd1 inhibited tumor-malignant phenotypes. Mechanistic research demonstrated that overexpression of Smarcd1 decreased the expression of Notch1, while knockdown of Notch1 increased the expression of Smarcd1 through Hes1 suppression. Hence, the crosstalk between Smarcd1 and Notch1, which formed a feedback loop, was crucial in regulation of glioblastoma malignant phenotypes. Furthermore, targeting Smarcd1 could be a potential strategy for human glioblastoma treatment.
Collapse
Affiliation(s)
- Yihao Zhu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
- Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China.
| | - Maoxing Fei
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Ting Tang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Wenhao Niu
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, People's Republic of China
| |
Collapse
|
13
|
Tong X, Chen Y, Zhu X, Ye Y, Xue Y, Wang R, Gao Y, Zhang W, Gao W, Xiao L, Chen H, Zhang P, Ji H. Nanog maintains stemness of Lkb1-deficient lung adenocarcinoma and prevents gastric differentiation. EMBO Mol Med 2021; 13:e12627. [PMID: 33439550 PMCID: PMC7933951 DOI: 10.15252/emmm.202012627] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Growing evidence supports that LKB1-deficient KRAS-driven lung tumors represent a unique therapeutic challenge, displaying strong cancer plasticity that promotes lineage conversion and drug resistance. Here we find that murine lung tumors from the KrasLSL-G12D/+ ; Lkb1flox/flox (KL) model show strong plasticity, which associates with up-regulation of stem cell pluripotency genes such as Nanog. Deletion of Nanog in KL model initiates a gastric differentiation program and promotes mucinous lung tumor growth. We find that NANOG is not expressed at a meaningful level in human lung adenocarcinoma (ADC), as well as in human lung invasive mucinous adenocarcinoma (IMA). Gastric differentiation involves activation of Notch signaling, and perturbation of Notch pathway by the γ-secretase inhibitor LY-411575 remarkably impairs mucinous tumor formation. In contrast to non-mucinous tumors, mucinous tumors are resistant to phenformin treatment. Such therapeutic resistance could be overcome through combined treatments with LY-411575 and phenformin. Overall, we uncover a previously unappreciated plasticity of LKB1-deficient tumors and identify the Nanog-Notch axis in regulating gastric differentiation, which holds important therapeutic implication for the treatment of mucinous lung cancer.
Collapse
Affiliation(s)
- Xinyuan Tong
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Yueqing Chen
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xinsheng Zhu
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yi Ye
- School of Life Science and TechnologyShanghai Tech UniversityShanghaiChina
| | - Yun Xue
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Rui Wang
- Department of Thoracic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yijun Gao
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Wenjing Zhang
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Weiqiang Gao
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Lei Xiao
- College of Animal Science and Zhejiang University School of MedicineZhejiang UniversityHangzhouChina
| | - Haiquan Chen
- Department of Thoracic SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Peng Zhang
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Hongbin Ji
- State Key Laboratory of Cell BiologyShanghai Institute of Biochemistry and Cell BiologyCenter for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- School of Life Science and TechnologyShanghai Tech UniversityShanghaiChina
| |
Collapse
|
14
|
Soylu H, Kırca M, Avcı S, Ozpolat B, Ustunel I. Antiandrogen abiraterone and docetaxel treatments affect Notch1, Jagged1 and Hes1 expressions in metastatic prostate cancer cells. Exp Mol Pathol 2021; 119:104607. [PMID: 33482170 DOI: 10.1016/j.yexmp.2021.104607] [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: 10/21/2020] [Revised: 12/10/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Prostate cancer is the most common cancer in men. A Notch signaling pathway is an important pathway in cell proliferation, differentiation, and fate. However, currently, the effects of abiraterone based-anti-androgene therapy and docetaxel, the most commonly used standard chemotherapy in prostate cancer treatment, on Notch signaling pathway are unknown. This study aimed to investigate the effects of abiraterone acetate and docetaxel on the expression of Notch1, Jagged1 and Hes1 in prostate cancer cell lines. METHODS In vitro effects of abiraterone acetate and docetaxel were examined on Notch1, Jagged1, and Hes1 expression in LNCaP and PC3 PCa cell lines by immunofluorescence, Western blot, and qRT-PCR. MTT proliferation assay was used to evaluate cell proliferation and survival. RESULTS We found that in the treatment of PC3 cells with abiraterone acetate, docetaxel, and their combination, only mRNA expressions of Notch1, Jagged1 and Hes1 were affected compared to control, but these expression differences were not observed in protein expression. In LNCaP cells, abiraterone acetate and the combination groups reduced Notch1 protein expression. All treatment groups did not alter Jagged1 expression compared to control, but significantly increased the Hes1 gene and protein expression. CONCLUSION Our findings suggest that abiraterone and docetaxel treatments affects the expression of Notch signal pathway proteins. But these drugs especially cause significant upregulation in Hes1 expression in PCa cells. Therefore, co-application of Notch signaling inhibitors together with docetaxel and abiraterone chemotherapy, it was thought that decreased Hes1 expression could be stopped the deterioration of the prognosis of the patient.
Collapse
Affiliation(s)
- Hakan Soylu
- Department of Histology and Embryology, Faculty of Medicine, Duzce University, 81620 Duzce, Turkey
| | - Mustafa Kırca
- Department of Biochemistry, Faculty of Medicine, Kutahya Health Sciences University, 43100 Kutahya, Turkey
| | - Sema Avcı
- Department of Histology and Embryology, Faculty of Medicine, Alanya Alaaddin Keykubat University, 07070 Antalya, Turkey
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Ismail Ustunel
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, 07070 Antalya, TURKEY.
| |
Collapse
|
15
|
Abstract
Head and neck cancer is a group of neoplastic diseases affecting the facial, oral, and neck region. It is one of the most common cancers worldwide with an aggressive, invasive evolution. Due to the heterogeneity of the tissues affected, it is particularly challenging to study the molecular mechanisms at the basis of these tumors, and to date we are still lacking accurate targets for prevention and therapy. The Notch signaling is involved in a variety of tumorigenic mechanisms, such as regulation of the tumor microenvironment, aberrant intercellular communication, and altered metabolism. Here, we provide an up-to-date review of the role of Notch in head and neck cancer and draw parallels with other types of solid tumors where the Notch pathway plays a crucial role in emergence, maintenance, and progression of the disease. We therefore give a perspective view on the importance of the pathway in neoplastic development in order to define future lines of research and novel therapeutic approaches.
Collapse
|
16
|
Bousquet Mur E, Bernardo S, Papon L, Mancini M, Fabbrizio E, Goussard M, Ferrer I, Giry A, Quantin X, Pujol JL, Calvayrac O, Moll HP, Glasson Y, Pirot N, Turtoi A, Cañamero M, Wong KK, Yarden Y, Casanova E, Soria JC, Colinge J, Siebel CW, Mazieres J, Favre G, Paz-Ares L, Maraver A. Notch inhibition overcomes resistance to tyrosine kinase inhibitors in EGFR-driven lung adenocarcinoma. J Clin Invest 2020; 130:612-624. [PMID: 31671073 DOI: 10.1172/jci126896] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 10/15/2019] [Indexed: 12/29/2022] Open
Abstract
EGFR-mutated lung adenocarcinoma patients treated with gefitinib and osimertinib show a therapeutic benefit limited by the appearance of secondary mutations, such as EGFRT790M and EGFRC797S. It is generally assumed that these secondary mutations render EGFR completely unresponsive to the inhibitors, but contrary to this, we uncovered here that gefitinib and osimertinib increased STAT3 phosphorylation (p-STAT3) in EGFRT790M and EGFRC797S tumoral cells. Interestingly, we also found that concomitant Notch inhibition with gefitinib or osimertinib treatment induced a p-STAT3-dependent strong reduction in the levels of the transcriptional repressor HES1. Importantly, we showed that tyrosine kinase inhibitor-resistant tumors, with EGFRT790M and EGFRC797S mutations, were highly responsive to the combined treatment of Notch inhibitors with gefitinib or osimertinib, respectively. Finally, in patients with EGFR mutations treated with tyrosine kinase inhibitors, HES1 protein levels increased during relapse and correlated with shorter progression-free survival. Therefore, our results offer a proof of concept for an alternative treatment to chemotherapy in lung adenocarcinoma osimertinib-treated patients after disease progression.
Collapse
Affiliation(s)
- Emilie Bousquet Mur
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Sara Bernardo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Laura Papon
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Maicol Mancini
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Eric Fabbrizio
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Marion Goussard
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Irene Ferrer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France.,Unidad de Investigación Clínica de Cáncer de Pulmón, Instituto de Investigación Hospital 12 de Octubre-CNIO, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Anais Giry
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Xavier Quantin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Jean-Louis Pujol
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France.,Montpellier Academic Hospital, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Olivier Calvayrac
- INSERM, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France; Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France; University of Toulouse III (Paul Sabatier), Toulouse, France
| | - Herwig P Moll
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Yaël Glasson
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Nelly Pirot
- Réseau d'Histologie Expérimentale de Montpellier, BioCampus, UMS3426 CNRS-US009 INSERM-UM, Montpellier, France
| | - Andrei Turtoi
- IRCM, Université de Montpellier, ICM, Montpellier, France
| | - Marta Cañamero
- Roche Pharmaceutical Research and Early Development, Translational Medicine, Roche Innovation Center, Munich, Germany
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York, USA
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Emilio Casanova
- Department of Physiology, Center of Physiology and Pharmacology and Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
| | - Jean-Charles Soria
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Paris-Sud University, Villejuif, France
| | | | - Christian W Siebel
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California, USA
| | - Julien Mazieres
- INSERM, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France; Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France; University of Toulouse III (Paul Sabatier), Toulouse, France.,Thoracic Oncology Department, Larrey Hospital, University Hospital of Toulouse, France; INSERM, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France; University of Toulouse III (Paul Sabatier), Toulouse, France
| | - Gilles Favre
- INSERM, Centre de Recherche en Cancérologie de Toulouse, CRCT UMR-1037, Toulouse, France; Institut Claudius Regaud, IUCT-Oncopole, Laboratoire de Biologie Médicale Oncologique, Toulouse, France; University of Toulouse III (Paul Sabatier), Toulouse, France
| | - Luis Paz-Ares
- Unidad de Investigación Clínica de Cáncer de Pulmón, Instituto de Investigación Hospital 12 de Octubre-CNIO, Madrid, Spain.,Montpellier Academic Hospital, Hôpital Arnaud de Villeneuve, Montpellier, France.,Medical School, Universidad Complutense, Madrid, Spain
| | - Antonio Maraver
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
17
|
Notch Transduction in Non-Small Cell Lung Cancer. Int J Mol Sci 2020; 21:ijms21165691. [PMID: 32784481 PMCID: PMC7461113 DOI: 10.3390/ijms21165691] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily-conserved Notch signaling pathway plays critical roles in cell communication, function and homeostasis equilibrium. The pathway serves as a cell-to-cell juxtaposed molecular transducer and is crucial in a number of cell processes including cell fate specification, asymmetric cell division and lateral inhibition. Notch also plays critical roles in organismal development, homeostasis, and regeneration, including somitogenesis, left-right asymmetry, neurogenesis, tissue repair, self-renewal and stemness, and its dysregulation has causative roles in a number of congenital and acquired pathologies, including cancer. In the lung, Notch activity is necessary for cell fate specification and expansion, and its aberrant activity is markedly linked to various defects in club cell formation, alveologenesis, and non-small cell lung cancer (NSCLC) development. In this review, we focus on the role this intercellular signaling device plays during lung development and on its functional relevance in proximo-distal cell fate specification, branching morphogenesis, and alveolar cell determination and maturation, then revise its involvement in NSCLC formation, progression and treatment refractoriness, particularly in the context of various mutational statuses associated with NSCLC, and, lastly, conclude by providing a succinct outlook of the therapeutic perspectives of Notch targeting in NSCLC therapy, including an overview on prospective synthetic lethality approaches.
Collapse
|
18
|
Saad MI, Alhayyani S, McLeod L, Yu L, Alanazi M, Deswaerte V, Tang K, Jarde T, Smith JA, Prodanovic Z, Tate MD, Balic JJ, Watkins DN, Cain JE, Bozinovski S, Algar E, Kohmoto T, Ebi H, Ferlin W, Garbers C, Ruwanpura S, Sagi I, Rose-John S, Jenkins BJ. ADAM17 selectively activates the IL-6 trans-signaling/ERK MAPK axis in KRAS-addicted lung cancer. EMBO Mol Med 2020; 11:emmm.201809976. [PMID: 30833304 PMCID: PMC6460353 DOI: 10.15252/emmm.201809976] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oncogenic KRAS mutations are major drivers of lung adenocarcinoma (LAC), yet the direct therapeutic targeting of KRAS has been problematic. Here, we reveal an obligate requirement by oncogenic KRAS for the ADAM17 protease in LAC In genetically engineered and xenograft (human cell line and patient-derived) Kras G12D-driven LAC models, the specific blockade of ADAM17, including with a non-toxic prodomain inhibitor, suppressed tumor burden by reducing cellular proliferation. The pro-tumorigenic activity of ADAM17 was dependent upon its threonine phosphorylation by p38 MAPK, along with the preferential shedding of the ADAM17 substrate, IL-6R, to release soluble IL-6R that drives IL-6 trans-signaling via the ERK1/2 MAPK pathway. The requirement for ADAM17 in Kras G12D-driven LAC was independent of bone marrow-derived immune cells. Furthermore, in KRAS mutant human LAC, there was a significant positive correlation between augmented phospho-ADAM17 levels, observed primarily in epithelial rather than immune cells, and activation of ERK and p38 MAPK pathways. Collectively, these findings identify ADAM17 as a druggable target for oncogenic KRAS-driven LAC and provide the rationale to employ ADAM17-based therapeutic strategies for targeting KRAS mutant cancers.
Collapse
Affiliation(s)
- Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Sultan Alhayyani
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Liang Yu
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Mohammad Alanazi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Virginie Deswaerte
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Ke Tang
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Thierry Jarde
- Cancer Program, Monash Biomedicine Discovery Institute, Clayton, Vic., Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic., Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia
| | - Julian A Smith
- Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Clayton, Vic., Australia.,Department of Cardiothoracic Surgery, Monash Health, Clayton, Vic., Australia
| | | | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Jesse J Balic
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - D Neil Watkins
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - Jason E Cain
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia.,Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Vic., Australia
| | - Elizabeth Algar
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Genetics and Molecular Pathology Laboratory, Monash Health, Clayton, Vic., Australia
| | - Tomohiro Kohmoto
- Department of Human Genetics, Tokushima University Graduate School of Medicine, Tokushima, Japan.,Division of Molecular Genetics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan.,Division of Advanced Cancer Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Christoph Garbers
- Department of Pathology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Saleela Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia.,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia .,Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| |
Collapse
|
19
|
Katoh M, Katoh M. Precision medicine for human cancers with Notch signaling dysregulation (Review). Int J Mol Med 2020; 45:279-297. [PMID: 31894255 PMCID: PMC6984804 DOI: 10.3892/ijmm.2019.4418] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
NOTCH1, NOTCH2, NOTCH3 and NOTCH4 are transmembrane receptors that transduce juxtacrine signals of the delta‑like canonical Notch ligand (DLL)1, DLL3, DLL4, jagged canonical Notch ligand (JAG)1 and JAG2. Canonical Notch signaling activates the transcription of BMI1 proto‑oncogene polycomb ring finger, cyclin D1, CD44, cyclin dependent kinase inhibitor 1A, hes family bHLH transcription factor 1, hes related family bHLH transcription factor with YRPW motif 1, MYC, NOTCH3, RE1 silencing transcription factor and transcription factor 7 in a cellular context‑dependent manner, while non‑canonical Notch signaling activates NF‑κB and Rac family small GTPase 1. Notch signaling is aberrantly activated in breast cancer, non‑small‑cell lung cancer and hematological malignancies, such as T‑cell acute lymphoblastic leukemia and diffuse large B‑cell lymphoma. However, Notch signaling is inactivated in small‑cell lung cancer and squamous cell carcinomas. Loss‑of‑function NOTCH1 mutations are early events during esophageal tumorigenesis, whereas gain‑of‑function NOTCH1 mutations are late events during T‑cell leukemogenesis and B‑cell lymphomagenesis. Notch signaling cascades crosstalk with fibroblast growth factor and WNT signaling cascades in the tumor microenvironment to maintain cancer stem cells and remodel the tumor microenvironment. The Notch signaling network exerts oncogenic and tumor‑suppressive effects in a cancer stage‑ or (sub)type‑dependent manner. Small‑molecule γ‑secretase inhibitors (AL101, MRK‑560, nirogacestat and others) and antibody‑based biologics targeting Notch ligands or receptors [ABT‑165, AMG 119, rovalpituzumab tesirine (Rova‑T) and others] have been developed as investigational drugs. The DLL3‑targeting antibody‑drug conjugate (ADC) Rova‑T, and DLL3‑targeting chimeric antigen receptor‑modified T cells (CAR‑Ts), AMG 119, are promising anti‑cancer therapeutics, as are other ADCs or CAR‑Ts targeting tumor necrosis factor receptor superfamily member 17, CD19, CD22, CD30, CD79B, CD205, Claudin 18.2, fibroblast growth factor receptor (FGFR)2, FGFR3, receptor‑type tyrosine‑protein kinase FLT3, HER2, hepatocyte growth factor receptor, NECTIN4, inactive tyrosine‑protein kinase 7, inactive tyrosine‑protein kinase transmembrane receptor ROR1 and tumor‑associated calcium signal transducer 2. ADCs and CAR‑Ts could alter the therapeutic framework for refractory cancers, especially diffuse‑type gastric cancer, ovarian cancer and pancreatic cancer with peritoneal dissemination. Phase III clinical trials of Rova‑T for patients with small‑cell lung cancer and a phase III clinical trial of nirogacestat for patients with desmoid tumors are ongoing. Integration of human intelligence, cognitive computing and explainable artificial intelligence is necessary to construct a Notch‑related knowledge‑base and optimize Notch‑targeted therapy for patients with cancer.
Collapse
Affiliation(s)
| | - Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
20
|
Tomatidine Represses Invasion and Migration of Human Osteosarcoma U2OS and HOS Cells by Suppression of Presenilin 1 and c-Raf-MEK-ERK Pathway. Molecules 2020; 25:molecules25020326. [PMID: 31941156 PMCID: PMC7024336 DOI: 10.3390/molecules25020326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma, which is the most prevalent malignant bone tumor, is responsible for the great majority of bone cancer-associated deaths because of its highly metastatic potential. Although tomatidine is suggested to serve as a chemosensitizer in multidrug-resistant tumors, the anti-metastatic effect of tomatidine in osteosarcoma is still unknown. Here, we tested the hypothesis that tomatidine suppresses migration and invasion, features that are associated with metastatic process in human osteosarcoma cells and also investigate its underlying pathway. Tomatidine, up to 100 μM, without cytotoxicity, inhibited the invasion and migration capabilities of human osteosarcoma U2OS and HOS cells and repressed presenilin 1 (PS-1) expression of U2OS cells. After the knockdown of PS-1, U2OS and HOS cells’ biological behaviors of cellular invasion and migratory potential were significantly reduced. While tomatidine significantly decreased the phosphorylation of c-Raf, mitogen/extracellular signal-regulated kinase (MEK), and extracellular signal-regulated protein kinase (ERK)1/2 in U2OS cells, no obvious influences on p-Jun N-terminal kinase, p38, and Akt, including their phosphorylation, were observed. In ERK 1 silencing U2 OS cells, tomatidine further enhanced the decrease of their migratory potential and invasive activities. We conclude that both PS-1 derived from U2OS and HOS cells and the c-Raf–MEK–ERK pathway contribute to cellular invasion and migration and tomatidine could inhibit the phenomenons. These findings indicate that tomatidine might be a potential candidate for anti-metastasis treatment of human osteosarcoma.
Collapse
|
21
|
Elko EA, Cunniff B, Seward DJ, Chia SB, Aboushousha R, van de Wetering C, van der Velden J, Manuel A, Shukla A, Heintz NH, Anathy V, van der Vliet A, Janssen-Heininger YMW. Peroxiredoxins and Beyond; Redox Systems Regulating Lung Physiology and Disease. Antioxid Redox Signal 2019; 31:1070-1091. [PMID: 30799628 PMCID: PMC6767868 DOI: 10.1089/ars.2019.7752] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Significance: The lung is a unique organ, as it is constantly exposed to air, and thus it requires a robust antioxidant defense system to prevent the potential damage from exposure to an array of environmental insults, including oxidants. The peroxiredoxin (PRDX) family plays an important role in scavenging peroxides and is critical to the cellular antioxidant defense system. Recent Advances: Exciting discoveries have been made to highlight the key features of PRDXs that regulate the redox tone. PRDXs do not act in isolation as they require the thioredoxin/thioredoxin reductase/NADPH, sulfiredoxin (SRXN1) redox system, and in some cases glutaredoxin/glutathione, for their reduction. Furthermore, the chaperone function of PRDXs, controlled by the oxidation state, demonstrates the versatility in redox regulation and control of cellular biology exerted by this class of proteins. Critical Issues: Despite the long-known observations that redox perturbations accompany a number of pulmonary diseases, surprisingly little is known about the role of PRDXs in the etiology of these diseases. In this perspective, we review the studies that have been conducted thus far to address the roles of PRDXs in lung disease, or experimental models used to study these diseases. Intriguing findings, such as the secretion of PRDXs and the formation of autoantibodies, raise a number of questions about the pathways that regulate secretion, redox status, and immune response to PRDXs. Future Directions: Further understanding of the mechanisms by which individual PRDXs control lung inflammation, injury, repair, chronic remodeling, and cancer, and the importance of PRDX oxidation state, configuration, and client proteins that govern these processes is needed.
Collapse
Affiliation(s)
- Evan A Elko
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - David J Seward
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Shi Biao Chia
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Allison Manuel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Arti Shukla
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Nicholas H Heintz
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
22
|
Moghbeli M, Mosannen Mozaffari H, Memar B, Forghanifard MM, Gholamin M, Abbaszadegan MR. Role of MAML1 in targeted therapy against the esophageal cancer stem cells. J Transl Med 2019; 17:126. [PMID: 30992079 PMCID: PMC6469193 DOI: 10.1186/s12967-019-1876-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Esophageal cancer is the sixth-leading cause of cancer-related deaths worldwide. Cancer stem cells (CSCs) are the main reason for tumor relapse in esophageal squamous cell carcinoma (ESCC). The NOTCH pathway is important in preservation of CSCs, therefore it is possible to target such cells by targeting MAML1 as the main component of the NOTCH transcription machinery. METHODS In present study we isolated the CD44+ ESCC CSCs and designed a MAML1-targeted therapy to inhibit the NOTCH signaling pathway. CSCs were isolated using magnetic cell sorting utilizing the CD44 cell surface marker. Several stem cell markers were analyzed in the levels of protein and mRNA expression. The isolated CSCs were characterized in vivo in NUDE mice. Biological role of MAML1 was assessed in isolated CD44+ CSCs. A drug resistance assay was also performed to assess the role of MAML1 in CD44+ CSCs with 5FU resistance. RESULTS The CD44+ CSCs had ability to form tumors in NUDE mice. MAML1 silencing caused a significant decrease (p = 0.019) and ectopic expression caused a significant increase in migration of CD44+ CSCs (p = 0.012). Moreover, MAML1 silencing and ectopic expression significantly increased and decreased 5FU resistance, respectively (p < 0.05). MAML1 silencing significantly increased the number of cells in G1 phase (p = 0.008), and its ectopic expression significantly increased the number of CD44+ CSCS in S phase (p = 0.037). CONCLUSIONS MAML1 may be utilized for targeted therapy with a low side effect to eliminate the CD44+ CSCs through inhibition of canonical NOTCH pathway in ESCC patients.
Collapse
MESH Headings
- Aged, 80 and over
- Animals
- Biomarkers, Tumor/metabolism
- Cell Cycle
- Cell Movement
- DNA-Binding Proteins/metabolism
- Drug Resistance, Neoplasm
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/pathology
- Gene Expression Regulation, Neoplastic
- Humans
- Hyaluronan Receptors/metabolism
- Male
- Mice, Nude
- Molecular Targeted Therapy
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Notch/metabolism
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hooman Mosannen Mozaffari
- Gastroenterology and Hepatology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahram Memar
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mehran Gholamin
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
23
|
Cui Q, Xing J, Gu Y, Nan X, Ma W, Chen Y, Zhao H. GXYLT2 accelerates cell growth and migration by regulating the Notch pathway in human cancer cells. Exp Cell Res 2019; 376:1-10. [PMID: 30716301 DOI: 10.1016/j.yexcr.2019.01.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/27/2019] [Accepted: 01/31/2019] [Indexed: 12/14/2022]
Abstract
Glucoside xylosyltransferase2 (GXYLT2), a member of the human α-1,3-D-xylosyltransferases, functions to modify the first xylose to the O-Glucose residue on epidermal growth factor (EGF) repeats of Notch receptors. It is well-established that the Notch signaling pathway plays a critical role in proper development and homeostasis. However, the regulatory role of EGF xylosylation in Notch signaling and different cell activities in human cells remains unknown. In this study, we showed that knockdown of GXYLT2 suppressed human cell proliferation and induced G1/S phase cell cycle arrest. GXYLT2 downregulation also inhibited cell migration and invasion, whereas the overexpression of GXYLT2 had the opposite effects. Additionally, GXYLT2 activated Notch signaling and promoted the phosphorylation of MAPKs but not PI3K and Akt. Taken together, our findings indicated that GXYLT2 plays an important role in cell activities via regulation of the Notch signaling.
Collapse
Affiliation(s)
- Qi Cui
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Jinhao Xing
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Yajuan Gu
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Xu Nan
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Wenping Ma
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Yingyu Chen
- Peking University Center for Human Disease Genomics, Beijing 100191, China; Department of Immunology, Peking University School of Basic Medical Science, Key Laboratory of Medical Immunology, Beijing 100191, China
| | - Hongshan Zhao
- Department of Medical Genetics, Peking University School of Basic Medical Sciences, Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China.
| |
Collapse
|
24
|
Hong M, He J, Li S. SNW1 regulates Notch signaling in neuroblastoma through interacting with RBPJ. Biochem Biophys Res Commun 2019; 509:869-876. [PMID: 30642633 DOI: 10.1016/j.bbrc.2019.01.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 01/20/2023]
Abstract
The Notch signaling pathway is highly conserved, involved in cell-cell communication, and essential for multicellular organism development. The recombination signal binding protein for immunoglobulin kappa J region (RBPJ) transcriptional regulator plays a vital role in Notch signaling. When not complexed with Notch proteins, RBPJ acts as a transcriptional repressor. However, when bound to the Notch intracellular domain (NOTCH ICD) in the nucleus, RBPJ activates the transcription of Notch target genes. However, the role of RBPJ in the Notch signaling pathway in neuroblastoma (NB) remains unclear. In this study, we show that SNW domain containing 1 (SNW1) acts as a novel RBPJ partner and regulates the expression of Notch target genes. Additionally, we show that silencing RBPJ expression can prevent SNW1-mediated Notch gene expression. Public datasets and clinical NB tissues indicate that RBPJ and SNW1 are upregulated in NB, and are associated with unfavorable patient outcomes. Taken together, our results highlight that SNW1 interacts with RBPJ to regulate the Notch signaling pathway and presents a potential therapeutic target for NB.
Collapse
Affiliation(s)
- Mei Hong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing He
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwang Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
25
|
Sosa Iglesias V, Theys J, Groot AJ, Barbeau LMO, Lemmens A, Yaromina A, Losen M, Houben R, Dubois L, Vooijs M. Synergistic Effects of NOTCH/γ-Secretase Inhibition and Standard of Care Treatment Modalities in Non-small Cell Lung Cancer Cells. Front Oncol 2018; 8:460. [PMID: 30464927 PMCID: PMC6234899 DOI: 10.3389/fonc.2018.00460] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022] Open
Abstract
Background: Lung cancer is the leading cause of cancer death worldwide. More effective treatments are needed to increase durable responses and prolong patient survival. Standard of care treatment for patients with non-operable stage III-IV NSCLC is concurrent chemotherapy and radiation. An activated NOTCH signaling pathway is associated with poor outcome and treatment resistance in non-small cell lung cancer (NSCLC). NOTCH/γ-secretase inhibitors have been effective in controlling tumor growth in preclinical models but the therapeutic benefit of these inhibitors as monotherapy in patients has been limited so far. Because NOTCH signaling has been implicated in treatment resistance, we hypothesized that by combining NOTCH inhibitors with chemotherapy and radiotherapy this could result in an increased therapeutic effect. A direct comparison of the effects of NOTCH inhibition when combined with current treatment combinations for NSCLC is lacking. Methods: Using monolayer growth assays, we screened 101 FDA-approved drugs from the Cancer Therapy Evaluation Program alone, or combined with radiation, in the H1299 and H460 NSCLC cell lines to identify potent treatment interactions. Subsequently, using multicellular three-dimensional tumor spheroid assays, we tested a selection of drugs used in clinical practice for NSCLC patients, and combined these with a small molecule inhibitor, currently being tested in clinical trials, of the NOTCH pathway (BMS-906024) alone, or in combination with radiation, and measured specific spheroid growth delay (SSGD). Statistical significance was determined by one-way ANOVA with post-hoc Bonferroni correction, and synergism was assessed using two-way ANOVA. Results: Monolayer assays in H1299 and H460 suggest that 21 vs. 5% were synergistic, and 17 vs. 11% were additive chemoradiation interactions, respectively. In H1299 tumor spheroids, significant SSGD was obtained for cisplatin, etoposide, and crizotinib, which increased significantly after the addition of the NOTCH inhibitor BMS-906024 (but not for paclitaxel and pemetrexed), and especially in triple combination with radiation. Synergistic interactions were observed when BMS-906024 was combined with chemoradiation (cisplatin, paclitaxel, docetaxel, and crizotinib). Similar results were observed for H460 spheroids using paclitaxel or crizotinib in dual combination treatment with NOTCH inhibition and triple with radiation. Conclusions: Our findings point to novel synergistic combinations of NOTCH inhibition and chemoradiation that should be tested in NSCLC in vivo models for their ability to achieve an improved therapeutic ratio.
Collapse
Affiliation(s)
- Venus Sosa Iglesias
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Jan Theys
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Arjan J Groot
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Lydie M O Barbeau
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Alyssa Lemmens
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Ala Yaromina
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Mario Losen
- Department of Psychology and Neuropsychology, MHeNS, Maastricht University Medical Center, Maastricht, Netherlands
| | - Ruud Houben
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands.,MAASTRO Clinic, Maastricht, Netherlands
| | - Ludwig Dubois
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Marc Vooijs
- Department of Radiotherapy (MAASTRO), GROW-School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands.,MAASTRO Clinic, Maastricht, Netherlands
| |
Collapse
|
26
|
Dai G, Deng S, Guo W, Yu L, Yang J, Zhou S, Gao T. Notch pathway inhibition using DAPT, a γ-secretase inhibitor (GSI), enhances the antitumor effect of cisplatin in resistant osteosarcoma. Mol Carcinog 2018; 58:3-18. [PMID: 29964327 DOI: 10.1002/mc.22873] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/29/2018] [Indexed: 12/28/2022]
Abstract
Overcoming platinum drug resistance represents a major clinical challenge in osteosarcoma (OS) treatment. The high rates and patterns of therapeutic failure seen in patients are consistent with a steady accumulation of drug-resistant cancer stem cells (CSCs). Notch signaling is implicated in regulating CSCs and tumor resistance to platinum. Thus, we attempt to investigate whether inhibiting of Notch pathway could sensitize cisplatin (CDDP) to CDDP-resistant OS cells and the underlying molecular mechanisms. OS cell lines resistant to CDDP were treated with DAPT, CDDP or combination, we present evidences that DAPT enhances the cytotoxic effect of CDDP in resistant OS by inhibiting proliferation, resulting in G0/G1 cell-cycle arrest, inducing apoptosis, and reducing motility. In addition, DAPT targeting depletes OS stem cells (OSCs), thus increasing tumor sensitivity to platinum, which indicating that a dual combination targeting both OSCs and the bulk of tumor cells are needed for tumor eradication. We also found that the combination of CDDP and DAPT exhibit additive suppression on phosphorylated AKT and ERK, contributing to the anti-cancer effects. In animal model, this combination therapy inhibits the growth and metastasis of CDDP resistant tumor xenografts in nude mice to a greater extent than treatment with either reagent alone. Based on these results, we conclude that CDDP plus DAPT was able to sensitize CDDP-resistant human OS cells to CDDP by downregulation of Notch signaling. CDDP and DAPT combination treatment may be effective and promising for advanced OS.
Collapse
Affiliation(s)
- Guo Dai
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China.,Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China.,Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Shuang Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China.,Laboratory of Clinical Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Ling Yu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Jian Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Sheng Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China.,Department of Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, P.R. China
| | - Tian Gao
- Department of Orthopedic Oncology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| |
Collapse
|
27
|
Kong G, You X, Wen Z, Chang YI, Qian S, Ranheim EA, Letson C, Zhang X, Zhou Y, Liu Y, Rajagopalan A, Zhang J, Stieglitz E, Loh M, Hofmann I, Yang D, Zhong X, Padron E, Zhou L, Pear WS, Zhang J. Downregulating Notch counteracts Kras G12D-induced ERK activation and oxidative phosphorylation in myeloproliferative neoplasm. Leukemia 2018; 33:671-685. [PMID: 30206308 PMCID: PMC6405304 DOI: 10.1038/s41375-018-0248-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 07/16/2018] [Accepted: 08/07/2018] [Indexed: 12/21/2022]
Abstract
The Notch signaling pathway contributes to the pathogenesis of a wide spectrum of human cancers, including hematopoietic malignancies. Its functions are highly dependent on the specific cellular context. Gain-of-function NOTCH1 mutations are prevalent in human T cell leukemia, while loss of Notch signaling is reported in myeloid leukemias. Here, we report a novel oncogenic function of Notch signaling in oncogenic Kras-induced myeloproliferative neoplasm (MPN). We find that downregulation of Notch signaling in hematopoietic cells via DNMAML expression or Pofut1 deletion significantly blocks MPN development in KrasG12D mice in a cell-autonomous manner. Further mechanistic studies indicate that inhibition of Notch signaling significantly upregulates Dusp1, a dual phosphatase that inactivates p-ERK, and downregulates cytokine-evoked ERK activation in KrasG12D cells. Moreover, mitochondrial metabolism is greatly enhanced in KrasG12D cells but significantly reprogrammed by DNMAML close to that in control cells. Consequently, cell proliferation and expanded myeloid compartment in KrasG12D mice are significantly reduced. Consistent with these findings, combined inhibition of the MEK/ERK pathway and mitochondrial oxidative phosphorylation effectively inhibited the growth of human and mouse leukemia cells in vitro. Our study provides a strong rational to target both ERK signaling and aberrant metabolism in oncogenic Ras-driven myeloid leukemia.
Collapse
Affiliation(s)
- Guangyao Kong
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xian Jiaotong University, Xian, China. .,McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA.
| | - Xiaona You
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhi Wen
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuan-I Chang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA.,Institute of Physiology, National Yang-Ming University, Taipei City, Taiwan
| | - Shuiming Qian
- Wisconsin Institute for Discovery and Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Erik A Ranheim
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | | | | | - Yun Zhou
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Yangang Liu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Adhithi Rajagopalan
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Jingfang Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospital, University of California-San Francisco, San Francisco, CA, USA
| | - Mignon Loh
- Department of Pediatrics, Benioff Children's Hospital, University of California-San Francisco, San Francisco, CA, USA
| | - Inga Hofmann
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - David Yang
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Xuehua Zhong
- Wisconsin Institute for Discovery and Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric Padron
- Malignant Hematology, Moffitt Cancer Center, Tampa, FL, USA
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Warren S Pear
- Department of Pathology and Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
28
|
Costa-Machado LF, Martín-Hernández R, Sanchez-Luengo MÁ, Hess K, Vales-Villamarin C, Barradas M, Lynch C, de la Nava D, Diaz-Ruiz A, de Cabo R, Cañamero M, Martinez L, Sanchez-Carbayo M, Herranz D, Serrano M, Fernandez-Marcos PJ. Sirt1 protects from K-Ras-driven lung carcinogenesis. EMBO Rep 2018; 19:e43879. [PMID: 30021836 PMCID: PMC6123659 DOI: 10.15252/embr.201643879] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/18/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
The NAD+-dependent deacetylase SIRT1 can be oncogenic or tumor suppressive depending on the tissue. Little is known about the role of SIRT1 in non-small cell lung carcinoma (NSCLC), one of the deadliest cancers, that is frequently associated with mutated K-RAS Therefore, we investigated the effect of SIRT1 on K-RAS-driven lung carcinogenesis. We report that SIRT1 protein levels are downregulated by oncogenic K-RAS in a MEK and PI3K-dependent manner in mouse embryo fibroblasts (MEFs), and in human lung adenocarcinoma cell lines. Furthermore, Sirt1 overexpression in mice delays the appearance of K-RasG12V-driven lung adenocarcinomas, reducing the number and size of carcinomas at the time of death and extending survival. Consistently, lower levels of SIRT1 are associated with worse prognosis in human NSCLCs. Mechanistically, analysis of mouse Sirt1-Tg pneumocytes, isolated shortly after K-RasG12V activation, reveals that Sirt1 overexpression alters pathways involved in tumor development: proliferation, apoptosis, or extracellular matrix organization. Our work demonstrates a tumor suppressive role of SIRT1 in the development of K-RAS-driven lung adenocarcinomas in mice and humans, suggesting that the SIRT1-K-RAS axis could be a therapeutic target for NSCLCs.
Collapse
Affiliation(s)
- Luis Filipe Costa-Machado
- Bioactive Products and Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Roberto Martín-Hernández
- GENYAL Nutrigenomic Platform, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | | | - Katharina Hess
- Tumor Suppression Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Claudia Vales-Villamarin
- Bioactive Products and Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Marta Barradas
- Bioactive Products and Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Cian Lynch
- Tumor Suppression Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Daniel de la Nava
- Bioactive Products and Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Alberto Diaz-Ruiz
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Nutritional Interventions Group, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Nutritional Interventions Group, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Marta Cañamero
- Histopathology Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Pathology and Tissue Analysis, Pharma Research and Early Development Roche Innovation Centre, Munich, Germany
| | - Lola Martinez
- Flow Cytometry Unit, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Marta Sanchez-Carbayo
- Translational Oncology Lab, Lucio Lascaray Research Center, University of the Basque Country, Vitoria-Gasteiz, Spain
| | - Daniel Herranz
- Tumor Suppression Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Rutgers Cancer Institute of New Jersey and Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Manuel Serrano
- Tumor Suppression Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Pablo J Fernandez-Marcos
- Bioactive Products and Metabolic Syndrome Group - BIOPROMET, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
- Tumor Suppression Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| |
Collapse
|
29
|
Pine SR. Rethinking Gamma-secretase Inhibitors for Treatment of Non-small-Cell Lung Cancer: Is Notch the Target? Clin Cancer Res 2018; 24:6136-6141. [PMID: 30104200 DOI: 10.1158/1078-0432.ccr-18-1635] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/15/2018] [Accepted: 08/08/2018] [Indexed: 01/07/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths among men and women. γ-Secretase inhibitors, a class of small-molecule compounds that target the Notch pathway, have been tested to treat non-small-cell lung cancer (NSCLC) in preclinical and clinical trials. Although γ-secretase inhibitors elicit a response in some tumors as single agents and sensitize NSCLC to cytotoxic and targeted therapies, they have not yet been approved for NSCLC therapy. We discuss our recently published preclinical study using the γ-secretase inhibitor AL101, formerly BMS906024, on cell lines and PDX models of NSCLC, primarily lung adenocarcinoma. We propose that Notch pathway mutations may not be the most suitable biomarker for predicting NSCLC response to γ-secretase inhibitors. γ-Secretases have over 100 known γ-secretase cleavage substrates. Many of the γ-secretase substrates are directly involved in carcinogenesis or tumor progression, and are ideal candidates to be the "on-target" biomarkers for γ-secretase inhibitors. We propose the need to systematically test the γ-secretase and other targets as potential biomarkers for sensitivity before continuing clinical trials. Now that we have entered the postgenome/transcriptome era, this goal is easily attainable. Discovery of the biomarker(s) that predict sensitivity to γ-secretase inhibitors would guide selection of the responder population that is most likely to benefit and move the compounds closer to approval for therapeutic use in NSCLC.
Collapse
Affiliation(s)
- Sharon R Pine
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey. .,Departments of Pharmacology and Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, New Jersey.
| |
Collapse
|
30
|
Testa U, Castelli G, Pelosi E. Lung Cancers: Molecular Characterization, Clonal Heterogeneity and Evolution, and Cancer Stem Cells. Cancers (Basel) 2018; 10:E248. [PMID: 30060526 PMCID: PMC6116004 DOI: 10.3390/cancers10080248] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/21/2022] Open
Abstract
Lung cancer causes the largest number of cancer-related deaths in the world. Most (85%) of lung cancers are classified as non-small-cell lung cancer (NSCLC) and small-cell lung cancer (15%) (SCLC). The 5-year survival rate for NSCLC patients remains very low (about 16% at 5 years). The two predominant NSCLC histological phenotypes are adenocarcinoma (ADC) and squamous cell carcinoma (LSQCC). ADCs display several recurrent genetic alterations, including: KRAS, BRAF and EGFR mutations; recurrent mutations and amplifications of several oncogenes, including ERBB2, MET, FGFR1 and FGFR2; fusion oncogenes involving ALK, ROS1, Neuregulin1 (NRG1) and RET. In LSQCC recurrent mutations of TP53, FGFR1, FGFR2, FGFR3, DDR2 and genes of the PI3K pathway have been detected, quantitative gene abnormalities of PTEN and CDKN2A. Developments in the characterization of lung cancer molecular abnormalities provided a strong rationale for new therapeutic options and for understanding the mechanisms of drug resistance. However, the complexity of lung cancer genomes is particularly high, as shown by deep-sequencing studies supporting the heterogeneity of lung tumors at cellular level, with sub-clones exhibiting different combinations of mutations. Molecular studies performed on lung tumors during treatment have shown the phenomenon of clonal evolution, thus supporting the occurrence of a temporal tumor heterogeneity.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
31
|
Sosa Iglesias V, Giuranno L, Dubois LJ, Theys J, Vooijs M. Drug Resistance in Non-Small Cell Lung Cancer: A Potential for NOTCH Targeting? Front Oncol 2018; 8:267. [PMID: 30087852 PMCID: PMC6066509 DOI: 10.3389/fonc.2018.00267] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/29/2018] [Indexed: 12/14/2022] Open
Abstract
Drug resistance is a major cause for therapeutic failure in non-small cell lung cancer (NSCLC) leading to tumor recurrence and disease progression. Cell intrinsic mechanisms of resistance include changes in the expression of drug transporters, activation of pro-survival, and anti-apoptotic pathways, as well as non-intrinsic influences of the tumor microenvironment. It has become evident that tumors are composed of a heterogeneous population of cells with different genetic, epigenetic, and phenotypic characteristics that result in diverse responses to therapy, and underlies the emergence of resistant clones. This tumor heterogeneity is driven by subpopulations of tumor cells termed cancer stem cells (CSCs) that have tumor-initiating capabilities, are highly self-renewing, and retain the ability for multi-lineage differentiation. CSCs have been identified in NSCLC and have been associated with chemo- and radiotherapy resistance. Stem cell pathways are frequently deregulated in cancer and are implicated in recurrence after treatment. Here, we focus on the NOTCH signaling pathway, which has a role in stem cell maintenance in non-squamous non-small lung cancer, and we critically assess the potential for targeting the NOTCH pathway to overcome resistance to chemotherapeutic and targeted agents using both preclinical and clinical evidence.
Collapse
Affiliation(s)
- Venus Sosa Iglesias
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Lorena Giuranno
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Ludwig J Dubois
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Jan Theys
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Marc Vooijs
- Department of Radiation Oncology, GROW, School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| |
Collapse
|
32
|
Chammaa M, Malysa A, Redondo C, Jang H, Chen W, Bepler G, Fernandez-Valdivia R. RUMI is a novel negative prognostic marker and therapeutic target in non-small-cell lung cancer. J Cell Physiol 2018; 233:9548-9562. [PMID: 29953591 DOI: 10.1002/jcp.26858] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022]
Abstract
Recent comprehensive next-generation genome and transcriptome analyses in lung cancer patients, several clinical observations, and compelling evidence from mouse models of lung cancer have uncovered a critical role for Notch signaling in the initiation and progression of non-small-cell lung cancer (NSCLC). Notably, Rumi is a "protein O-glucosyltransferase" that regulates Notch signaling through O-glucosylation of Notch receptors, and is the only enzymatic regulator whose activity is required for both ligand-dependent and ligand-independent activation of Notch. We have conducted a detailed study on RUMI's involvement in NSCLC development and progression, and have further explored the therapeutic potential of its targeting in NSCLC. We have determined that Rumi is highly expressed in the alveolar and bronchiolar epithelia, including club cells and alveolar type II cells. Remarkably, RUMI maps to the region of chromosome 3q that corresponds to the major signature of neoplastic transformation in NSCLC, and is markedly amplified and overexpressed in NSCLC tumors. Notably, RUMI expression levels are predictive of poor prognosis and survival in NSCLC patients. Our data indicates that RUMI modulates Notch activity in NSCLC cells, and that its silencing dramatically decreases cell proliferation, migration, and survival. RUMI downregulation causes severe cell cycle S-phase arrest, increases genome instability, and induces late apoptotic-nonapoptotic cell death. Our studies demonstrate that RUMI is a novel negative prognostic factor with significant therapeutic potential in NSCLC, which embodies particular relevance especially when considering that, while current Notch inhibitory strategies target only ligand-dependent Notch activation, a large number of NSCLCs are driven by ligand-independent Notch activity.
Collapse
Affiliation(s)
- May Chammaa
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
| | - Agnes Malysa
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - Carlos Redondo
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan
| | - Hyejeong Jang
- Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Wei Chen
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan.,Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, Michigan.,Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Gerold Bepler
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, Michigan.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - Rodrigo Fernandez-Valdivia
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan.,Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan.,Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, Michigan.,Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| |
Collapse
|
33
|
Drosten M, Guerra C, Barbacid M. Genetically Engineered Mouse Models of K-Ras-Driven Lung and Pancreatic Tumors: Validation of Therapeutic Targets. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031542. [PMID: 28778964 DOI: 10.1101/cshperspect.a031542] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
K-RAS signaling has been intensely studied for over 40 years. Yet, as of today, no drugs have been approved to treat K-RAS mutant cancers. Since the turn of the century, scientists have used genetically engineered mouse (GEM) models to reproduce K-RAS mutant cancers in a laboratory setting to elucidate those molecular events responsible for the onset and progression of these tumors and to identify suitable therapies. In this review, we outline a brief description of available GEM models for two tumor types known to be driven by K-RAS mutations: lung adenocarcinoma and pancreatic ductal adenocarcinoma. In addition, we summarize a series of studies that have used these GEM tumor models to validate, either by genetic or pharmacological approaches, the therapeutic potential of a variety of targets, with the ultimate goal of translating these results to the clinical setting.
Collapse
Affiliation(s)
- Matthias Drosten
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), E-28029 Madrid, Spain
| | - Carmen Guerra
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), E-28029 Madrid, Spain
| | - Mariano Barbacid
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), E-28029 Madrid, Spain
| |
Collapse
|
34
|
Goriki A, Seiler R, Wyatt AW, Contreras-Sanz A, Bhat A, Matsubara A, Hayashi T, Black PC. Unravelling disparate roles of NOTCH in bladder cancer. Nat Rev Urol 2018; 15:345-357. [DOI: 10.1038/s41585-018-0005-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
miR-494-3p is a novel tumor driver of lung carcinogenesis. Oncotarget 2018; 8:7231-7247. [PMID: 27980227 PMCID: PMC5352317 DOI: 10.18632/oncotarget.13933] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/07/2016] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the leading cause of tumor-related death worldwide and more efforts are needed to elucidate lung carcinogenesis. Here we investigated the expression of 641 miRNAs in lung tumorigenesis in a K-Ras(+/LSLG12Vgeo);RERTn(ert/ert) mouse model and 113 human tumors. The conserved miRNA cluster on chromosome 12qF1 was significantly and progressively upregulated during murine lung carcinogenesis. In particular, miR-494-3p expression was correlated with lung cancer progression in mice and with worse survival in lung cancer patients. Mechanistically, ectopic expression of miR-494-3p in A549 lung cancer cells boosted the tumor-initiating population, enhanced cancer cell motility, and increased the expression of stem cell-related genes. Importantly, miR-494-3p improved the ability of A549 cells to grow and metastasize in vivo, modulating NOTCH1 and PTEN/PI3K/AKT signaling.Overall, these data identify miR-494-3p as a key factor in lung cancer onset and progression and possible therapeutic target.
Collapse
|
36
|
Nikolova O, Moser R, Kemp C, Gönen M, Margolin AA. Modeling gene-wise dependencies improves the identification of drug response biomarkers in cancer studies. Bioinformatics 2018; 33:1362-1369. [PMID: 28082455 DOI: 10.1093/bioinformatics/btw836] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/29/2016] [Indexed: 01/07/2023] Open
Abstract
Motivation In recent years, vast advances in biomedical technologies and comprehensive sequencing have revealed the genomic landscape of common forms of human cancer in unprecedented detail. The broad heterogeneity of the disease calls for rapid development of personalized therapies. Translating the readily available genomic data into useful knowledge that can be applied in the clinic remains a challenge. Computational methods are needed to aid these efforts by robustly analyzing genome-scale data from distinct experimental platforms for prioritization of targets and treatments. Results We propose a novel, biologically motivated, Bayesian multitask approach, which explicitly models gene-centric dependencies across multiple and distinct genomic platforms. We introduce a gene-wise prior and present a fully Bayesian formulation of a group factor analysis model. In supervised prediction applications, our multitask approach leverages similarities in response profiles of groups of drugs that are more likely to be related to true biological signal, which leads to more robust performance and improved generalization ability. We evaluate the performance of our method on molecularly characterized collections of cell lines profiled against two compound panels, namely the Cancer Cell Line Encyclopedia and the Cancer Therapeutics Response Portal. We demonstrate that accounting for the gene-centric dependencies enables leveraging information from multi-omic input data and improves prediction and feature selection performance. We further demonstrate the applicability of our method in an unsupervised dimensionality reduction application by inferring genes essential to tumorigenesis in the pancreatic ductal adenocarcinoma and lung adenocarcinoma patient cohorts from The Cancer Genome Atlas. Availability and Implementation : The code for this work is available at https://github.com/olganikolova/gbgfa. Contact : nikolova@ohsu.edu or margolin@ohsu.edu. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Olga Nikolova
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA
| | - Russell Moser
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Christopher Kemp
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mehmet Gönen
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA.,Department of Industrial Engineering, Koç University, İstanbul, Turkey
| | - Adam A Margolin
- Computational Biology Program, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
37
|
Zou B, Zhou XL, Lai SQ, Liu JC. Notch signaling and non-small cell lung cancer. Oncol Lett 2018; 15:3415-3421. [PMID: 29467866 PMCID: PMC5796339 DOI: 10.3892/ol.2018.7738] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 10/19/2017] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-associated mortality worldwide. Elucidation of the pathogenesis and biology of lung cancer is critical for the design of an effective treatment for patients. Non-small cell lung cancer (NSCLC) accounts for 80–85% of lung cancer cases. The abnormal expression of Notch signaling pathway members is a relatively frequent event in NSCLC. The Notch signaling pathway serves important roles in cell fate determination, proliferation, differentiation and apoptosis. Increasing evidence supports the association of Notch signaling dysregulation with various types of malignant tumor, including NSCLC. Several studies have demonstrated that members of the Notch signaling pathway may be potential biomarkers for predicting the progression and prognosis of patients with NSCLC. Furthermore, Notch signaling serves critical roles in the tumorigenesis and treatment resistance of NSCLC cells by promoting the proliferation or inhibiting the apoptosis of NSCLC cells. The present review provides a detailed summary of the roles of Notch signaling in NSCLC.
Collapse
Affiliation(s)
- Bin Zou
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xue-Liang Zhou
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Song-Qing Lai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ji-Chun Liu
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
38
|
High expression of MKP1/DUSP1 counteracts glioma stem cell activity and mediates HDAC inhibitor response. Oncogenesis 2017; 6:401. [PMID: 29284798 PMCID: PMC5865544 DOI: 10.1038/s41389-017-0003-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/09/2017] [Accepted: 09/07/2017] [Indexed: 12/12/2022] Open
Abstract
The elucidation of mechanisms involved in resistance to therapies is essential to improve the survival of patients with malignant gliomas. A major feature possessed by glioma cells that may aid their ability to survive therapy and reconstitute tumors is the capacity for self-renewal. We show here that glioma stem cells (GSCs) express low levels of MKP1, a dual-specificity phosphatase, which acts as a negative inhibitor of JNK, ERK1/2, and p38 MAPK, while induction of high levels of MKP1 expression are associated with differentiation of GSC. Notably, we find that high levels of MKP1 correlate with a subset of glioblastoma patients with better prognosis and overall increased survival. Gain of expression studies demonstrated that elevated MKP1 impairs self-renewal and induces differentiation of GSCs while reducing tumorigenesis in vivo. Moreover, we identified that MKP1 is epigenetically regulated and that it mediates the anti-tumor activity of histone deacetylase inhibitors (HDACIs) alone or in combination with temozolomide. In summary, this study identifies MKP1 as a key modulator of the interplay between GSC self-renewal and differentiation and provides evidence that the activation of MKP1, through epigenetic regulation, might be a novel therapeutic strategy to overcome therapy resistance in glioblastoma.
Collapse
|
39
|
Liao B, Zhou H, Liang H, Li C. Regulation of ERK and AKT pathways by hepatitis B virus X protein via the Notch1 pathway in hepatocellular carcinoma. Int J Oncol 2017; 51:1449-1459. [PMID: 29048612 PMCID: PMC5643068 DOI: 10.3892/ijo.2017.4126] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B virus (HBV) is the dominant risk factor for hepatocellular carcinoma (HCC). HBV X protein (HBx) plays crucial roles in HCC carcinogenesis. HBx interferes with several signaling pathways including the Notch1 pathway in HCC. In this study, we found that Notch1 was highly expressed in HCC, especially in large HCCs. Notch1 and HBx co-localized in HCC and their levels were positively correlated with each other. Notch1 expression was more elevated in HepG2.2.15 cells than that in HepG2 cells. HBx activated the Notch1 pathway in HepG2.2.15 cells. Suppression of HBx and the Notch1 pathway attenuated the growth of HepG2.2.15 cells. Notch1, ERK, and AKT pathways were inhibited after γ-secretase inhibitor treatment. Dual-specificity phosphatase 1 (DUSP1) and phosphatase and tensin homolog (PTEN) were upregulated after γ-secretase inhibitor treatment and Hes1 inhibition. Luciferase reporter assays showed that Hes1 suppressed the promoters of DUSP1 and PTEN genes, which was reversed by γ-secretase inhibitor treatment. Western blotting demonstrated that DUSP1 dephosphorylated pERK and PTEN dephosphorylated pAKT. Collectively, we found a link among HBx, the Notch1 pathway, DUSP1/PTEN, and ERK/AKT pathways, which influenced HCC cell survival and could be a therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Bo Liao
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, P.R. China
| | - Honghao Zhou
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, P.R. China
| | - Huifang Liang
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, P.R. China
| | - Changhai Li
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery; Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
40
|
Novikova SE, Kurbatov LK, Zavialova MG, Zgoda VG, Archakov AI. [Omics technologies in diagnostics of lung adenocarcinoma]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2017; 63:181-210. [PMID: 28781253 DOI: 10.18097/pbmc20176303181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To date lung adenocarcinoma (LAC) is the most common type of lung cancer. Numerous studies on LAC biology resulted in identification of crucial mutations in protooncogenes and activating neoplastic transformation pathways. Therapeutic approaches that significantly increase the survival rate of patients with LAC of different etiology have been developed and introduced into clinical practice. However, the main problem in the treatment of LAC is early diagnosis, taking into account both factors and mechanisms responsible in tumor initiation and progression. Identification of a wide biomarker repertoire with high specificity and reliability of detection appears to be a solution to this problem. In this context, proteins with differential expression in normal and pathological condition, suitable for detection in biological fluids are the most promising biomarkers. In this review we have analyzed literature data on studies aimed at search of LAC biomarkers. The major attention has been paid to protein biomarkers as the most promising and convenient subject of clinical diagnosis. The review also summarizes existing knowledge on posttranslational modifications, splice variants, isoforms, as well as model systems and transcriptome changes in LAC.
Collapse
Affiliation(s)
- S E Novikova
- Institute of Biomedical Chemistry, Moscow, Russia
| | - L K Kurbatov
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| | - A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
41
|
Forés M, Simón-Carrasco L, Ajuria L, Samper N, González-Crespo S, Drosten M, Barbacid M, Jiménez G. A new mode of DNA binding distinguishes Capicua from other HMG-box factors and explains its mutation patterns in cancer. PLoS Genet 2017; 13:e1006622. [PMID: 28278156 PMCID: PMC5344332 DOI: 10.1371/journal.pgen.1006622] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 02/08/2017] [Indexed: 11/19/2022] Open
Abstract
HMG-box proteins, including Sox/SRY (Sox) and TCF/LEF1 (TCF) family members, bind DNA via their HMG-box. This binding, however, is relatively weak and both Sox and TCF factors employ distinct mechanisms for enhancing their affinity and specificity for DNA. Here we report that Capicua (CIC), an HMG-box transcriptional repressor involved in Ras/MAPK signaling and cancer progression, employs an additional distinct mode of DNA binding that enables selective recognition of its targets. We find that, contrary to previous assumptions, the HMG-box of CIC does not bind DNA alone but instead requires a distant motif (referred to as C1) present at the C-terminus of all CIC proteins. The HMG-box and C1 domains are both necessary for binding specific TGAATGAA-like sites, do not function via dimerization, and are active in the absence of cofactors, suggesting that they form a bipartite structure for sequence-specific binding to DNA. We demonstrate that this binding mechanism operates throughout Drosophila development and in human cells, ensuring specific regulation of multiple CIC targets. It thus appears that HMG-box proteins generally depend on auxiliary DNA binding mechanisms for regulating their appropriate genomic targets, but that each sub-family has evolved unique strategies for this purpose. Finally, the key role of C1 in DNA binding also explains the fact that this domain is a hotspot for inactivating mutations in oligodendroglioma and other tumors, while being preserved in oncogenic CIC-DUX4 fusion chimeras associated to Ewing-like sarcomas.
Collapse
Affiliation(s)
- Marta Forés
- Institut de Biologia Molecular de Barcelona-CSIC, Barcelona, Spain
| | - Lucía Simón-Carrasco
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Leiore Ajuria
- Institut de Biologia Molecular de Barcelona-CSIC, Barcelona, Spain
| | - Núria Samper
- Institut de Biologia Molecular de Barcelona-CSIC, Barcelona, Spain
| | | | - Matthias Drosten
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Mariano Barbacid
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Gerardo Jiménez
- Institut de Biologia Molecular de Barcelona-CSIC, Barcelona, Spain
- ICREA, Barcelona, Spain
- * E-mail:
| |
Collapse
|
42
|
Abstract
The Notch signalling cascade is an evolutionarily conserved pathway that has a crucial role in regulating development and homeostasis in various tissues. The cellular processes and events that it controls are diverse, and continued investigation over recent decades has revealed how the role of Notch signalling is multifaceted and highly context dependent. Consistent with the far-reaching impact that Notch has on development and homeostasis, aberrant activity of the pathway is also linked to the initiation and progression of several malignancies, and Notch can in fact be either oncogenic or tumour suppressive depending on the tissue and cellular context. The Notch pathway therefore represents an important target for therapeutic agents designed to treat many types of cancer. In this Review, we focus on the latest developments relating specifically to the tumour-suppressor activity of Notch signalling and discuss the potential mechanisms by which Notch can inhibit carcinogenesis in various tissues. Potential therapeutic strategies aimed at restoring or augmenting Notch-mediated tumour suppression will also be highlighted.
Collapse
Affiliation(s)
- Craig S Nowell
- CMU, Department for Pathology and Immunology, University of Geneva, Rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research, Lausanne, Vaud 1015, Switzerland
| |
Collapse
|
43
|
Li S, Jiang Z, Xu W, Xie Y, Zhao L, Tang X, Wang F, Xin F. FIP-sch2, a new fungal immunomodulatory protein from Stachybotrys chlorohalonata, suppresses proliferation and migration in lung cancer cells. Appl Microbiol Biotechnol 2017; 101:3227-3235. [PMID: 28078399 DOI: 10.1007/s00253-016-8030-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 01/09/2023]
Abstract
Fungal immunomodulatory protein (FIP)-sch2, an immunomodulatory protein identified in the ascomycete Stachybotrys chlorohalonata by a sequence similarity search, is a novel member of the FIP family. FIP-sch2 shares high sequence identity, structure, and evolutionary conservation with previously reported FIPs. It was satisfactorily expressed in Escherichia coli with a glutathione S-transferase (GST) tag and purified by GST-affinity magnetic beads. To characterize the direct antitumor effects, human lung adenocarcinoma A549 cells were treated with different concentrations of recombinant FIP (rFIP)-sch2 in vitro, and the results showed that rFIP-sch2 could reduce cell viability dose-dependently with a half-maximal inhibitory concentration (IC50) of 9.48 μg/mL. Furthermore, rFIP-sch2 at 8 μg/mL could significantly induce apoptosis and interrupt migration in A549 cells. Notably, the antitumor effect of rFIP-sch2 was equivalent to that of rLZ-8 but was obviously increased compared to rFIP-fve. In addition, the exploration of the antitumor mechanism suggested that rFIP-sch2 induced lung cancer cell death by activating apoptosis and inhibiting migration. Our results indicated that rFIP-sch2 was a promising candidate for use in future cancer therapy.
Collapse
Affiliation(s)
- Shuying Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhonghao Jiang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Wenyi Xu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yingying Xie
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Leiming Zhao
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Xuanming Tang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Fengjiao Xin
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
44
|
Kurimoto R, Takiguchi Y. Targeting Epithelial-Mesenchymal Transition and Cancer Stem Cell. MOLECULAR TARGETED THERAPY OF LUNG CANCER 2017:295-307. [DOI: 10.1007/978-981-10-2002-5_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
45
|
Hernández J, Bechara E, Schlesinger D, Delgado J, Serrano L, Valcárcel J. Tumor suppressor properties of the splicing regulatory factor RBM10. RNA Biol 2016; 13:466-72. [PMID: 26853560 PMCID: PMC4841610 DOI: 10.1080/15476286.2016.1144004] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
RBM10 is an RNA binding protein and alternative splicing regulator frequently mutated in lung adenocarcinomas. Recent results indicate that RBM10 inhibits proliferation of lung cancer cells by promoting skipping of exon 9 of the gene NUMB, a frequent alternative splicing change in lung cancer generating a negative regulator of Notch signaling. Complementing these observations, we show that knock down of RBM10 in human cancer cells enhances growth of mouse tumor xenografts, confirming that RBM10 acts as a tumor suppressor, while knock down of an oncogenic mutant version of RBM10 reduces xenograft tumor growth. A RBM10 mutation found in lung cancer cells, V354E, disrupts RBM10-mediated regulation of NUMB alternative splicing, inducing the cell proliferation-promoting isoform. We now show that 2 natural RBM10 isoforms that differ by the presence or absence of V354 in the second RNA Recognition Motif (RRM2), display similar regulatory effects on NUMB alternative splicing, suggesting that V354E actively disrupts RBM10 activity. Structural modeling localizes V354 in the outside surface of one α-helix opposite to the RNA binding surface of RBM10, and we show that the mutation does not compromise binding of the RRM2 domain to NUMB RNA regulatory sequences. We further show that other RBM10 mutations found in lung adenocarcinomas also compromise regulation of NUMB exon 9. Collectively, our previous and current results reveal that RBM10 is a tumor suppressor that represses Notch signaling and cell proliferation through the regulation of NUMB alternative splicing.
Collapse
Affiliation(s)
- Jordi Hernández
- a Centre de Regulació Genòmica, The Barcelona Institute of Science and Technology , Dr. Aiguader 88, 08003 Barcelona , Spain.,b Universitat Pompeu Fabra , Dr. Aiguader 88, 08003 Barcelona , Spain
| | - Elias Bechara
- a Centre de Regulació Genòmica, The Barcelona Institute of Science and Technology , Dr. Aiguader 88, 08003 Barcelona , Spain.,b Universitat Pompeu Fabra , Dr. Aiguader 88, 08003 Barcelona , Spain
| | - Doerte Schlesinger
- a Centre de Regulació Genòmica, The Barcelona Institute of Science and Technology , Dr. Aiguader 88, 08003 Barcelona , Spain
| | - Javier Delgado
- a Centre de Regulació Genòmica, The Barcelona Institute of Science and Technology , Dr. Aiguader 88, 08003 Barcelona , Spain.,b Universitat Pompeu Fabra , Dr. Aiguader 88, 08003 Barcelona , Spain
| | - Luis Serrano
- a Centre de Regulació Genòmica, The Barcelona Institute of Science and Technology , Dr. Aiguader 88, 08003 Barcelona , Spain.,b Universitat Pompeu Fabra , Dr. Aiguader 88, 08003 Barcelona , Spain.,c Institució Catalana de Recerca i Estudis Avançats (ICREA) , Passeig Lluis Companys 23, 08010 Barcelona , Spain
| | - Juan Valcárcel
- a Centre de Regulació Genòmica, The Barcelona Institute of Science and Technology , Dr. Aiguader 88, 08003 Barcelona , Spain.,b Universitat Pompeu Fabra , Dr. Aiguader 88, 08003 Barcelona , Spain.,c Institució Catalana de Recerca i Estudis Avançats (ICREA) , Passeig Lluis Companys 23, 08010 Barcelona , Spain
| |
Collapse
|
46
|
Li S, Zhao L, Xu W, Jiang Z, Kang J, Wang F, Xin F. Identification and Characterisation of a Novel Protein FIP-sch3 from Stachybotrys chartarum. PLoS One 2016; 11:e0168436. [PMID: 27997578 PMCID: PMC5173029 DOI: 10.1371/journal.pone.0168436] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/01/2016] [Indexed: 11/19/2022] Open
Abstract
In this study, a novel FIP named FIP-sch3 has been identified and characterised. FIP-sch3 was identified in the ascomycete Stachybotrys chartarum, making it the second FIP to be identified outside the order of Basidiomycota. Recombinant FIP-sch3 (rFIP-shc3) was produced in Escherichia coli and purified using GST-affinity magnetic beads. The bioactive characteristics of FIP-sch3 were compared to those of well-known FIPs LZ-8 from Ganoderma lucidum and FIP-fve from Flammulina velutipes, which were produced and purified using the same method. The purified rFIP-sch3 exhibited a broad spectrum of anti-tumour activity in several types of tumour cells but had no cytotoxicity in normal human embryonic kidney 293 cells. Assays that were implemented to study these properties indicated that rFIP-sch3 significantly suppressed cell proliferation, induced apoptosis and inhibited cell migration in human lung adenocarcinoma A549 cells. The anti-tumour effects of rFIP-sch3 in A549 cells were comparable to those of rLZ-8, but they were significantly greater than those of rFIP-fve. Molecular assays that were built on real-time PCR further revealed potential mechanisms related to apoptosis and migration and that underlie phenotypic effects. These results indicate that FIP-shc3 has a unique anti-tumour bioactive profile, as do other FIPs, which provide a foundation for further studies on anti-tumour mechanisms. Importantly, this study also had convenient access to FIP-sch3 with potential human therapeutic applications.
Collapse
Affiliation(s)
- Shuying Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Leiming Zhao
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wenyi Xu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhonghao Jiang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jun Kang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (FW); (FX)
| | - Fengjiao Xin
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (FW); (FX)
| |
Collapse
|
47
|
Revandkar A, Perciato ML, Toso A, Alajati A, Chen J, Gerber H, Dimitrov M, Rinaldi A, Delaleu N, Pasquini E, D'Antuono R, Pinton S, Losa M, Gnetti L, Arribas A, Fraering P, Bertoni F, Nepveu A, Alimonti A. Inhibition of Notch pathway arrests PTEN-deficient advanced prostate cancer by triggering p27-driven cellular senescence. Nat Commun 2016; 7:13719. [PMID: 27941799 PMCID: PMC5159884 DOI: 10.1038/ncomms13719] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/27/2016] [Indexed: 12/17/2022] Open
Abstract
Activation of NOTCH signalling is associated with advanced prostate cancer and treatment resistance in prostate cancer patients. However, the mechanism that drives NOTCH activation in prostate cancer remains still elusive. Moreover, preclinical evidence of the therapeutic efficacy of NOTCH inhibitors in prostate cancer is lacking. Here, we provide evidence that PTEN loss in prostate tumours upregulates the expression of ADAM17, thereby activating NOTCH signalling. Using prostate conditional inactivation of both Pten and Notch1 along with preclinical trials carried out in Pten-null prostate conditional mouse models, we demonstrate that Pten-deficient prostate tumours are addicted to the NOTCH signalling. Importantly, we find that pharmacological inhibition of γ-secretase promotes growth arrest in both Pten-null and Pten/Trp53-null prostate tumours by triggering cellular senescence. Altogether, our findings describe a novel pro-tumorigenic network that links PTEN loss to ADAM17 and NOTCH signalling, thus providing the rational for the use of γ-secretase inhibitors in advanced prostate cancer patients.
Collapse
Affiliation(s)
- Ajinkya Revandkar
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
- Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne CH 1011, Switzerland
| | - Maria Luna Perciato
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Alberto Toso
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Abdullah Alajati
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Jingjing Chen
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
- Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne CH 1011, Switzerland
| | - Hermeto Gerber
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland
- Foundation Eclosion, Plan-Les-Ouates CH 1228, Switzerland
- Campus Biotech Innovation Park, Geneva CH 1202, Switzerland
| | - Mitko Dimitrov
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Nicolas Delaleu
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen 5021, Norway
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Rocco D'Antuono
- Institute for Research in Biomedicine, University of Italian Switzerland, Via Vincenzo Vela 6, Bellinzona 6500, Switzerland
| | - Sandra Pinton
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Marco Losa
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Letizia Gnetti
- Pathology Unit, University Hospital of Parma, Parma 43126, Italy
| | - Alberto Arribas
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Patrick Fraering
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH 1015, Switzerland
- Foundation Eclosion, Plan-Les-Ouates CH 1228, Switzerland
- Campus Biotech Innovation Park, Geneva CH 1202, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
| | - Alain Nepveu
- Rosalind and Morris Goodman Cancer Research Center, Department of Oncology, Biochemistry and Medicine, McGill University, Montreal, Quebec, Canada H3A1A3
| | - Andrea Alimonti
- Institute of Oncology Research (IOR) and Oncology Institute of Southern Switzerland (IOSI), Bellinzona CH 6500, Switzerland
- Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne CH 1011, Switzerland
| |
Collapse
|
48
|
Cimas FJ, Callejas-Valera JL, Pascual-Serra R, García-Cano J, Garcia-Gil E, De la Cruz-Morcillo MA, Ortega-Muelas M, Serrano-Oviedo L, Gutkind JS, Sánchez-Prieto R. MKP1 mediates chemosensitizer effects of E1a in response to cisplatin in non-small cell lung carcinoma cells. Oncotarget 2016; 6:44095-107. [PMID: 26689986 PMCID: PMC4792544 DOI: 10.18632/oncotarget.6574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/25/2015] [Indexed: 12/19/2022] Open
Abstract
The adenoviral gene E1a is known to enhance the antitumor effect of cisplatin, one of the cornerstones of the current cancer chemotherapy. Here we study the molecular basis of E1a mediated sensitivity to cisplatin in an experimental model of Non-small cell lung cancer. Our data show how E1a blocks the induction of autophagy triggered by cisplatin and promotes the apoptotic response in resistant cells. Interestingly, at the molecular level, we present evidences showing how the phosphatase MKP1 is a major determinant of cisplatin sensitivity and its upregulation is strictly required for the induction of chemosensitivity mediated by E1a. Indeed, E1a is almost unable to promote sensitivity in H460, in which the high expression of MKP1 remains unaffected by E1a. However, in resistant cell as H1299, H23 or H661, which display low levels of MKP1, E1a expression promotes a dramatic increase in the amount of MKP1 correlating with cisplatin sensitivity. Furthermore, effective knock down of MKP1 in H1299 E1a expressing cells restores resistance to a similar extent than parental cells. In summary, the present work reinforce the critical role of MKP1 in the cellular response to cisplatin highlighting the importance of this phosphatase in future gene therapy approach based on E1a gene.
Collapse
Affiliation(s)
- Francisco J Cimas
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | | | - Raquel Pascual-Serra
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Jesus García-Cano
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Elena Garcia-Gil
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Miguel A De la Cruz-Morcillo
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Marta Ortega-Muelas
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Leticia Serrano-Oviedo
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | | | - Ricardo Sánchez-Prieto
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| |
Collapse
|
49
|
Rajendran D, Zhang Y, Berry DM, McGlade CJ. Regulation of Numb isoform expression by activated ERK signaling. Oncogene 2016; 35:5202-13. [PMID: 27041567 DOI: 10.1038/onc.2016.69] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 12/23/2015] [Accepted: 01/25/2016] [Indexed: 12/29/2022]
Abstract
The endocytic adaptor protein Numb has a major role in development as an intrinsic regulator of cell fate determination and inhibitor of the Notch signaling pathway. In vertebrates, four protein isoforms of Numb are produced through alternative splicing (AS) of two cassette exons (exons 3 and 9). AS of coding exon 9 (E9) produces E9-included (p72/p71) and -excluded (p66/p65) protein products. Expression of Numb isoforms is developmentally regulated and E9-included products are expressed in progenitors, whereas E9-excluded isoforms are dominantly expressed in differentiated cells. Analyses of AS events in multiple cancers previously identified a switch in Numb transcript and protein expression from the E9-excluded to the E9-included isoform, suggesting that misregulation of the mechanisms that control E9 inclusion may have a role in tumorigenesis. Here we identify splicing factors ASF/SF2 and PTBP1 as regulators of E9 splicing and show that activation of the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway promotes E9 inclusion in cancer cells. Our evidence supports a mechanism by which Numb AS is regulated in response to oncogenic signaling pathways, and contributes to activation of downstream pathways to promote tumorigenesis.
Collapse
Affiliation(s)
- D Rajendran
- Program in Cell Biology, and The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital For Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Y Zhang
- Program in Cell Biology, and The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital For Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - D M Berry
- Program in Cell Biology, and The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital For Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
| | - C J McGlade
- Program in Cell Biology, and The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital For Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Ambrogio C, Nadal E, Villanueva A, Gómez-López G, Cash TP, Barbacid M, Santamaría D. KRAS-driven lung adenocarcinoma: combined DDR1/Notch inhibition as an effective therapy. ESMO Open 2016; 1:e000076. [PMID: 27843638 PMCID: PMC5070278 DOI: 10.1136/esmoopen-2016-000076] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 12/28/2022] Open
Abstract
Understanding the early evolution of cancer heterogeneity during the initial steps of tumorigenesis can uncover vulnerabilities of cancer cells that may be masked at later stages. We describe a comprehensive approach employing gene expression analysis in early lesions to identify novel therapeutic targets and the use of mouse models to test synthetic lethal drug combinations to treat human Kirsten rat sarcoma viral oncogene homologue (KRAS)-driven lung adenocarcinoma.
Collapse
Affiliation(s)
- Chiara Ambrogio
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ernest Nadal
- Department of Medical Oncology, Multidisciplinary Thoracic Oncology Unit , Catalan Institute of Oncology , Barcelona , Spain
| | - Alberto Villanueva
- Xenopat S.L., Business Bioincubator, Bellvitge Health Science Campus, Barcelona, Spain; Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit , Structural Biology and Biocomputing Programme, Centro Nacional de Investigaciones Oncológicas (CNIO) , Madrid , Spain
| | - Timothy P Cash
- Tumour Suppression, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO) , Madrid , Spain
| | - Mariano Barbacid
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO) , Madrid , Spain
| | - David Santamaría
- Experimental Oncology, Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO) , Madrid , Spain
| |
Collapse
|