1
|
Chen W, Wang F, Yu X, Qi J, Dong H, Cui B, Zhang Q, Wu Y, An J, Ni N, Liu C, Han Y, Zhang S, Schmitt CA, Deng J, Yu Y, Du J. LncRNA MIR31HG fosters stemness malignant features of non-small cell lung cancer via H3K4me1- and H3K27Ace-mediated GLI2 expression. Oncogene 2024; 43:1328-1340. [PMID: 37950038 PMCID: PMC11065682 DOI: 10.1038/s41388-023-02883-4] [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: 04/25/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Non-coding RNAs are responsible for oncogenesis and the development of stemness features, including multidrug resistance and metastasis, in various cancers. Expression of lncRNA MIR31HG in lung cancer tissues and peripheral sera of lung cancer patients were remarkably higher than that of healthy individuals and indicated a poor prognosis. Functional analysis showed that MIR31HG fosters stemness-associated malignant features of non-small cell lung cancer cells. Further mechanistic investigation revealed that MIR31HG modulated GLI2 expression via WDR5/MLL3/P300 complex-mediated H3K4me and H3K27Ace modification. In vivo MIR31HG repression with an antisense oligonucleotide attenuated tumor growth and distal organ metastasis, whereas MIR31HG promotion remarkably encouraged cellular invasion in lung and liver tissues. Our data suggested that MIR31HG is a potential diagnostic indicator and druggable therapeutic target to facilitate multiple strategic treatments for lung cancer patients.
Collapse
Affiliation(s)
- Weiwei Chen
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Fei Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Xinyuan Yu
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Jingjing Qi
- Department of Hematology and Internal Oncology, Johannes Kepler University Linz, Altenbergerstraße 69, 4040, Linz, Austria
| | - Hongliang Dong
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Bingjie Cui
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Qian Zhang
- Department of Pathology, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Yan Wu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Jiajia An
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, PR China
| | - Na Ni
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Cuilan Liu
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Yuchen Han
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, PR China
| | - Shuo Zhang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, 256600, PR China
| | - Clemens A Schmitt
- Johannes Kepler University, Altenbergerstraße 69, 4040, Linz, Austria
- Kepler University Hospital, Department of Hematology and Oncology, Krankenhausstraße 9, 4020, Linz, Austria
- Charité-Universitätsmedizin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medical Department of Hematology, Oncology and Tumor Immunology, and Molekulares Krebsforschungszentrum - MKFZ, Campus Virchow Klinikum, 13353, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
- Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner site, Berlin, Germany
| | - Jiong Deng
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China.
| | - Yong Yu
- Department of Hematology and Internal Oncology, Johannes Kepler University Linz, Altenbergerstraße 69, 4040, Linz, Austria.
| | - Jing Du
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, PR China.
- Department of Oncology, Binzhou Medical University Hospital, Binzhou, 256600, PR China.
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, 256600, PR China.
| |
Collapse
|
2
|
Zhang N, Shan W, Gao L, Kou SH, Lu C, Yang H, Peng B, Tam KY, Lee LTO, Zheng J. Repurposing the Hedgehog pathway inhibitor, BMS-833923, as a phosphatidylglycerol-selective membrane-disruptive colistin adjuvant against ESKAPE pathogens. Int J Antimicrob Agents 2023; 62:106888. [PMID: 37328075 DOI: 10.1016/j.ijantimicag.2023.106888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/18/2023]
Abstract
The rapid emergence and spread of multi-drug- or pan-drug-resistant bacterial pathogens, such as ESKAPE, pose a serious threat to global health. However, the development of novel antibiotics is hindered by difficulties in identifying new antibiotic targets and the rapid development of drug resistance. Drug repurposing is an effective alternative strategy for combating antibiotic resistance that both saves resources and extends the life of existing antibiotics in combination treatment regimens. Screening of a chemical compound library identified BMS-833923 (BMS), a smoothened antagonist that kills Gram-positive bacteria directly, and potentiates colistin to destroy various Gram-negative bacteria. BMS did not induce detectable antibiotic resistance in vitro, and showed effective activity against drug-resistant bacteria in vivo. Mechanistic studies revealed that BMS caused membrane disruption by targeting the membrane phospholipids phosphatidylglycerol and cardiolipin, promoting membrane dysfunction, metabolic disturbance, leakage of cellular components, and, ultimately, cell death. This study describes a potential strategy to enhance the efficacy of colistin and combat multi-drug-resistant ESKAPE pathogens.
Collapse
Affiliation(s)
- Nian Zhang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wenying Shan
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Liangliang Gao
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Si Hoi Kou
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Chang Lu
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Huilin Yang
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Bo Peng
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Leo Tsz On Lee
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Ministry of Education Frontiers Science Centre for Precision Oncology, University of Macau, Taipa, Macau, China; Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| | - Jun Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
3
|
Dong H, Zeng L, Chen W, Zhang Q, Wang F, Wu Y, Cui B, Qi J, Zhang X, Liu C, Deng J, Yu Y, Schmitt CA, Du J. N6-methyladenine-mediated aberrant activation of the lncRNA SOX2OT-GLI1 loop promotes non-small-cell lung cancer stemness. Cell Death Discov 2023; 9:149. [PMID: 37149646 PMCID: PMC10164154 DOI: 10.1038/s41420-023-01442-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
Despite the advent of precision medicine and immunotherapy, mortality due to lung cancer remains high. The sonic hedgehog (SHH) cascade and its key terminal factor, glioma-associated oncogene homolog 1 (GLI1), play a pivotal role in the stemness and drug resistance of lung cancer. Here, we investigated the molecular mechanism of non-canonical aberrant GLI1 upregulation. The SHH cascade was upregulated in stem spheres and chemo-resistant lung cancer cells and was accountable for drug resistance against multiple chemotherapy regimens. GLI1 and the long non-coding RNA SOX2OT were positively regulated, and the GLI1-SOX2OT loop mediated the proliferation of parental and stem-like lung cancer cells. Further mechanistic investigation revealed that SOX2OT facilitated METTL3/14/IGF2BP2-mediated m6A modification and stabilization of the GLI1 mRNA. Additionally, SOX2OT upregulated METTL3/14/IGF2BP2 by sponging miR-186-5p. Functional analysis corroborated that GLI1 acted as a downstream target of METTL3/14/IGF2BP2, and GLI1 silencing could block the oncogenicity of lung cancer stem-like cells. Pharmacological inhibition of the loop remarkably inhibited the oncogenesis of lung cancer cells in vivo. Compared with paired adjacent normal tissues, lung cancer specimens exhibited consistently upregulated GLI1/SOX2OT/METTL3/14/IGF2BP2. The m6A-modified GLI1-SOX2OT loop may serve as a potential therapeutic target and prognostic predictor for lung cancer therapy and diagnosis in the clinic.
Collapse
Affiliation(s)
- Hongliang Dong
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Lili Zeng
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Weiwei Chen
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Qian Zhang
- Department of Pathology, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Fei Wang
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Yan Wu
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
- Department of Oncology, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Bingjie Cui
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Jingjing Qi
- Johannes Kepler University, Altenbergerstraße 69, 4040, Linz, Austria
| | - Xin Zhang
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
- Department of Hematology, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Cuilan Liu
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Jiong Deng
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China
| | - Yong Yu
- Johannes Kepler University, Altenbergerstraße 69, 4040, Linz, Austria
| | - Clemens A Schmitt
- Johannes Kepler University, Altenbergerstraße 69, 4040, Linz, Austria.
- Kepler University Hospital, Department of Hematology and Oncology, Krankenhausstraße 9, 4020, Linz, Austria.
- Charité-Universitätsmedizin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medical Department of Hematology, Oncology and Tumor Immunology, and Molekulares Krebsforschungszentrum - MKFZ, Campus Virchow Klinikum, 13353, Berlin, Germany.
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße, 1013125, Berlin, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner site Berlin, Berlin, Germany.
| | - Jing Du
- Medical Research Center, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China.
- Department of Oncology, Binzhou Medical University Hospital, 256600, Binzhou, P. R. China.
| |
Collapse
|
4
|
Rowbotham SP, Goruganthu MUL, Arasada RR, Wang WZ, Carbone DP, Kim CF. Lung Cancer Stem Cells and Their Clinical Implications. Cold Spring Harb Perspect Med 2022; 12:a041270. [PMID: 34580078 PMCID: PMC9121890 DOI: 10.1101/cshperspect.a041270] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
It is now widely accepted that stem cells exist in various cancers, including lung cancer, which are referred to as cancer stem cells (CSCs). CSCs are defined in this context as the subset of tumor cells with the ability to form tumors in serial transplantation and cloning assays and form tumors at metastatic sites. Mouse models of lung cancer have shown that lung CSCs reside in niches that are essential for the maintenance of stemness, plasticity, enable antitumor immune evasion, and provide metastatic potential. Similar to normal lung stem cells, Notch, Wnt, and the Hedgehog signaling cascades have been recruited by the CSCs to regulate stemness and also provide therapy-driven resistance in lung cancer. Compounds targeting β-catenin and Sonic hedgehog (Shh) activity have shown promising anti-CSC activity in preclinical murine models of lung cancer. Understanding CSCs and their niches in lung cancer can answer fundamental questions pertaining to tumor maintenance and associated immune regulation and escape that appear important in the quest to develop novel lung cancer therapies and enhance sensitivity to currently approved chemo-, targeted-, and immune therapeutics.
Collapse
Affiliation(s)
- Samuel P Rowbotham
- Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mounika U L Goruganthu
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio 43210, USA
| | - Rajeswara R Arasada
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, Ohio 43210, USA
| | - Walter Z Wang
- James Thoracic Oncology Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - David P Carbone
- James Thoracic Oncology Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Carla F Kim
- Stem Cell Program, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
5
|
Ma C, Hu K, Ullah I, Zheng QK, Zhang N, Sun ZG. Molecular Mechanisms Involving the Sonic Hedgehog Pathway in Lung Cancer Therapy: Recent Advances. Front Oncol 2022; 12:729088. [PMID: 35433472 PMCID: PMC9010822 DOI: 10.3389/fonc.2022.729088] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 03/03/2022] [Indexed: 12/09/2022] Open
Abstract
According to the latest statistics from the International Agency for Research on Cancer (IARC), lung cancer is one of the most lethal malignancies in the world, accounting for approximately 18% of all cancer-associated deaths. Yet, even with aggressive interventions for advanced lung cancer, the five-year survival rate remains low, at around 15%. The hedgehog signaling pathway is highly conserved during embryonic development and is involved in tissue homeostasis as well as organ development. However, studies have documented an increasing prevalence of aberrant activation of HH signaling in lung cancer patients, promoting malignant lung cancer progression with poor prognostic outcomes. Inhibitors targeting the HH pathway have been widely used in tumor therapy, however, they still cannot avoid the occurrence of drug resistance. Interestingly, natural products, either alone or in combination with chemotherapy, have greatly improved overall survival outcomes for lung cancer patients by acting on the HH signaling pathway because of its unique and excellent pharmacological properties. In this review, we elucidate on the underlying molecular mechanisms through which the HH pathway promotes malignant biological behaviors in lung cancer, as well as the potential of inhibitors or natural compounds in targeting HH signaling for clinical applications in lung cancer therapy.
Collapse
Affiliation(s)
- Chao Ma
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Kang Hu
- School of Clinical Medicine, Weifang Medical University, Weifang, China
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Irfan Ullah
- Department of Surgery, Khyber Medical University Peshawar, Peshawar, Pakistan
| | - Qing-Kang Zheng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Nan Zhang
- Breast Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Zhi-Gang Sun, ; Nan Zhang,
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Zhi-Gang Sun, ; Nan Zhang,
| |
Collapse
|
6
|
Zárate AM, Espinosa-Bustos C, Guerrero S, Fierro A, Oyarzún-Ampuero F, Quest AFG, Di Marcotullio L, Loricchio E, Caimano M, Calcaterra A, González-Quiroz M, Aguirre A, Meléndez J, Salas CO. A New Smoothened Antagonist Bearing the Purine Scaffold Shows Antitumour Activity In Vitro and In Vivo. Int J Mol Sci 2021; 22:8372. [PMID: 34445078 PMCID: PMC8395040 DOI: 10.3390/ijms22168372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
The Smoothened (SMO) receptor is the most druggable target in the Hedgehog (HH) pathway for anticancer compounds. However, SMO antagonists such as vismodegib rapidly develop drug resistance. In this study, new SMO antagonists having the versatile purine ring as a scaffold were designed, synthesised, and biologically tested to provide an insight to their mechanism of action. Compound 4s was the most active and the best inhibitor of cell growth and selectively cytotoxic to cancer cells. 4s induced cell cycle arrest, apoptosis, a reduction in colony formation and downregulation of PTCH and GLI1 expression. BODIPY-cyclopamine displacement assays confirmed 4s is a SMO antagonist. In vivo, 4s strongly inhibited tumour relapse and metastasis of melanoma cells in mice. In vitro, 4s was more efficient than vismodegib to induce apoptosis in human cancer cells and that might be attributed to its dual ability to function as a SMO antagonist and apoptosis inducer.
Collapse
Affiliation(s)
- Ana María Zárate
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile; (A.M.Z.); (A.F.)
| | - Christian Espinosa-Bustos
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile;
| | - Simón Guerrero
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone 1007, Independencia, Santiago 8380492, Chile; (S.G.); (F.O.-A.); (A.F.G.Q.)
- Instituto de Investigación Interdisciplinar en Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad SEK (I3CBSEK), Fernando Manterola 0789, Providencia, Santiago 7520317, Chile
| | - Angélica Fierro
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile; (A.M.Z.); (A.F.)
| | - Felipe Oyarzún-Ampuero
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone 1007, Independencia, Santiago 8380492, Chile; (S.G.); (F.O.-A.); (A.F.G.Q.)
- Departamento de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago 8380494, Chile
| | - Andrew F. G. Quest
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Sergio Livingstone 1007, Independencia, Santiago 8380492, Chile; (S.G.); (F.O.-A.); (A.F.G.Q.)
- Laboratorio de Comunicaciones Celulares, Centro de Estudios en Ejercicio, Metabolismo y Cáncer (CEMC), Program of Cellular and Molecular Biology, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile
| | - Lucia Di Marcotullio
- Laboratory Affiliated to Insituto Pasteur Italia, Fondazione Cenci Bognetti, Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Elena Loricchio
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Miriam Caimano
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy;
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Matías González-Quiroz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Universidad de Chile, Independencia 1027, Santiago 8380453, Chile;
| | - Adam Aguirre
- Laboratorio de Medicina Traslacional, Fundación Arturo López Pérez, Rancagua 878, Lower Fifth Floor, Providencia, Santiago 8320000, Chile;
| | - Jaime Meléndez
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile;
| | - Cristian O. Salas
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago 702843, Chile; (A.M.Z.); (A.F.)
| |
Collapse
|
7
|
Sun J, Jia J, Yuan W, Liu S, Wang W, Ge L, Ge L, Liu XJ. LncRNA BLACAT1 Accelerates Non-small Cell Lung Cancer Through Up-Regulating the Activation of Sonic Hedgehog Pathway. Front Oncol 2021; 11:625253. [PMID: 33937028 PMCID: PMC8080024 DOI: 10.3389/fonc.2021.625253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Recently, increasing evidence has displayed that lncRNAs can exhibit crucial function in cancer progression, including lung cancer. LncRNA bladder cancer-associated transcript 1 (BLACAT1) is reported to participate in various cancers. The aim of our current study was to investigate the function of BLACAT1 in non-small cell lung cancer progression and study the functional pathway. Here, we reported BLACAT1 was significantly up-regulated in lung cancer tissues in comparison to the adjacent normal tissues, which suggested BLACAT1 might act as an oncogene in lung cancer. Then, A549 and PC9 cells were infected with BLACAT1 overexpression plasmid and shRNA. As shown, we proved up-regulation of BLACAT1 greatly induced the growth of non-small cell lung cancer cells. Reversely, knockdown of BLACAT1 reduced A549 and PC9 cell proliferation, migration and invasion. Sonic hedgehog (shh) signaling is able to exert a significant role in carcinogenesis, including lung cancer. Currently, we proved that up-regulation of BLACAT1 activated shh signaling pathway, via inducing shh, Gli-1 and Smo expression. shh pathway inhibitor GANT-61 reversed the effect of overexpression of BLACAT1 on non-small cell lung cancer. Moreover, we manifested that loss of BLACAT1 remarkably reduced the in vivo growth and metastasis of A549 cells via enhancing infiltrating CD3+ T cells. In conclusion, our research revealed a critical role of BLACAT1 in the modulation of non-small cell lung cancer via modulating shh pathway.
Collapse
Affiliation(s)
- Jiwei Sun
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Jingzhou Jia
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Wuying Yuan
- Department of Thoracic Surgery, Henan Provincial Chest Hospital, Zhengzhou, China
| | - Shu Liu
- Department of Respiratory, Huai'an Second People's Hospital and the Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Wei Wang
- Department of Oncology, Huai'an Second People's Hospital and the Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Lili Ge
- Department of Clinical Laboratory, Huai'an Second People's Hospital and the Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Liyue Ge
- Department of Oncology, Huai'an Second People's Hospital and the Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Xiao-Jun Liu
- Outpatient Department of External Injury and Wound, Huai'an Second People's Hospital and the Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
8
|
Chen WW, Gong KK, Yang LJ, Dai JJ, Zhang Q, Wang F, Li XL, Xi SC, Du J. Scutellariabarbata D. Don extraction selectively targets stemness-prone NSCLC cells by attenuating SOX2/SMO/GLI1 network loop. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113295. [PMID: 32841701 DOI: 10.1016/j.jep.2020.113295] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/02/2020] [Accepted: 08/16/2020] [Indexed: 05/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellariabarbata D. Don extraction (SBE), a traditional Chinese medicine, has been proved effective against various malignant disorders in clinics with tolerable side-effects when administered alone or in combination with conventional chemotherapeutic regimens. AIM OF THIS STUDY Multi-drug resistance of cancer is attributed to existence of cancer stemness-prone cells that harbor aberrantly high activation of Sonic Hedgehog (SHH) cascade. Our previous study has demonstrated that SBE sensitized non-small cell lung cancer (NSCLC) cells to Cisplatin (DDP) treatment by downregulating SHH pathway. Yet, whether SBE could prohibit proliferation of cancer stemness-prone cells and its underlying molecular mechanisms remain to be investigated. In this article, we further investigated intervention of SBE on NSCLC cell stemness-associated phenotypes and its potential mode of action. MATERIALS AND METHODS CCK-8 and clonal formation detection were used to measure the anti-proliferative potency of SBE against NSCLC and normal epithelial cells. Sphere formation assay and RQ-PCR were used to detect proliferation of cancer stemness cells and associated marker expression upon SBE incubation. Mechanistically, DARTS-WB and SPR were used to unveil binding target of SBE. Immunodeficient mice were implanted with patient derived tumor bulk for in vivo validation of anti-cancer effect of SBE. RESULTS SBE selectively attenuated proliferation and stemness-like phenotypes of NSCLC cells rather than bronchial normal epithelial cells. Drug-protein interaction analysis revealed that SBE could directly bind with stem cell-specific transcription factor sex determining region Y-box 2 (SOX2) and interfere with the SOX2/SMO/GLI1 positive loop. In vivo assay using patient-derived xenografts (PDXs) model further proved that SBE diminished tumor growth and SOX2 expression in vivo. CONCLUSION Our data indicate that SBE represses stemness-related features of NSCLC cells via targeting SOX2 and may serve as an alternative therapeutic option for clinic treatment.
Collapse
Affiliation(s)
- Wei-Wei Chen
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Kai-Kai Gong
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Li-Juan Yang
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Juan-Juan Dai
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Qian Zhang
- Department of Pathology, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Feng Wang
- Department of Oncology, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Xue-Lin Li
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China
| | - Si-Chuan Xi
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China.
| | - Jing Du
- Center Research Institute, Binzhou Medical University Hospital, 256600, Binzhou, PR China.
| |
Collapse
|
9
|
Qi H, Li W, Zhang J, Chen J, Peng J, Liu Y, Yang S, Du J, Long X, Ng CSH, Li MY, Chen GG. Glioma-associated oncogene homolog 1 stimulates FOXP3 to promote non-small cell lung cancer stemness. Am J Transl Res 2020; 12:1839-1850. [PMID: 32509180 PMCID: PMC7270000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/20/2020] [Indexed: 06/11/2023]
Abstract
Glioma-associated oncogene homolog 1 (GLI1), an oncogenic molecule in non-small cell lung cancer (NSCLC), promotes the growth of NSCLC by enhancing lung cancer stem cells (LCSCs). However, the mechanism responsible remains unknown. FOXP3 is known to maintain LCSCs. The aim of this study was to explore whether GLI1 enhanced LCSCs via stimulating FOXP3. Experiments were performed in NSCLC tissue samples, cell lines and the animal tumor model. The expression of GLI1- and LCSC-related molecules was assessed at protein and mRNA levels. Relevant cell functions were also determined. A tumor xenograft mouse model was established to confirm the oncogenic role of GLI1. We confirmed that the expression of GLI1 was up-regulated in the tumor tissues of NSCLC compared with adjacent non-tumor tissues. But no significant association between GLI1 and clinicopathological characteristics was found. GLI1 expression was positively correlated with FOXP3 and it could promote FOXP3 expression likely via acting on the promoter of FOXP3. Along with the upregulation of FOXP3, GLI1 increased the expression of LCSC markers, ALDH1A1 and OCT4A, and the formation of tumor spheres, whereas the inhibition of GLI1 decreased the above features. We also found the involvement of Notch1 activation in GLI1-mediated FOXP3 pathway. The In vivo mouse tumor model verified the positive role of GLI1 in the growth of the tumor. Collectively, this study has demonstrated that GLI1 stimulates FOXP3 to promote LCSCs.
Collapse
Affiliation(s)
- Haolong Qi
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan UniversityWuhan, Hubei, China
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales HospitalShatin, NT, Hong Kong, China
| | - Wende Li
- Guangdong Key Laboratory of Laboratory Animal, Guangdong Laboratory Animals Monitoring InstituteGuangzhou, Guangdong, China
| | - Jie Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan UniversityWuhan, Hubei, China
| | - Jianxin Chen
- Department of Surgery, Jiangxia District Hospital of Traditional Chinese MedicineWuhan, Hubei, China
| | - Jia Peng
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales HospitalShatin, NT, Hong Kong, China
| | - Yi Liu
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales HospitalShatin, NT, Hong Kong, China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical UniversityZhanjiang, Guangdong, China
| | - Shucai Yang
- Department of Clinical Laboratory, Pingshan District People’s Hospital of ShenzhenShenzhen, Guangdong, China
| | - Jing Du
- Peking University Shenzhen HospitalShenzhen, Guangdong, China
| | - Xiang Long
- Peking University Shenzhen HospitalShenzhen, Guangdong, China
| | - Calvin SH Ng
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales HospitalShatin, NT, Hong Kong, China
| | - Ming-Yue Li
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales HospitalShatin, NT, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, Guangdong, China
- Guangzhou Regenerative Medicine and Health Guangdong LaboratoryGuangzhou, Guangdong, China
| | - George G Chen
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales HospitalShatin, NT, Hong Kong, China
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical UniversityZhanjiang, Guangdong, China
- Shenzhen Research Institute, The Chinese University of Hong KongShenzhen, Guangdong, China
| |
Collapse
|
10
|
Xu Y, Wang J, Ding H. Regulation of epithelial-mesenchymal transition via sonic hedgehog/glioma-associated oncogene homolog 1 signaling pathway in peritoneal mesothelial cells. Cell Biol Int 2020; 44:1691-1700. [PMID: 32298032 DOI: 10.1002/cbin.11363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/24/2020] [Accepted: 04/11/2020] [Indexed: 02/06/2023]
Abstract
Sonic hedgehog (Shh) signaling regulating epithelial-mesenchymal transition (EMT) in cultured rat peritoneal mesothelial cells (PMCs) remains an under-investigated topic. The current study aimed to elucidate the role of Shh signaling in the regulation of EMT in PMCs to attenuate peritoneal injury, with the view of enhancing the efficacy of peritoneal dialysis (PD). PMCs were initially extracted from male Wistar rats using pancreatic enzyme digestion. The expression of Shh and glioma-associated oncogene homolog (Gli1) was quantitatively analyzed using the reverse-transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. Migration of PMCs was determined using Transwell assay. The expression of Shh, Gli1, and EMT markers including α-smooth muscle actin (α-SMA), fibronectin, collagen I, snail1, and E-cadherin was examined by RT-qPCR, western blot analysis, and immunofluorescence respectively. High glucose induction was identified to promote cell migration and increase the expression of Shh and Gli1 in a dose- and time-dependent manner in rat PMCs. Cyclopamine (CPN) was observed to block the Shh signaling induced by high glucose, accompanied by cell migration inhibition, decreased expression of α-SMA, fibronectin, collagen I and snail1 as well as increased expression of E-cadherin. Altogether, overexpression of Gli1 by transfected Gli1 plasmid promotes cell migration and upregulates α-SMA, fibronectin, Snail1, and collagen I expression, while downregulating E-cadherin expression. Shh/Gli1 signaling is important in mediating EMT in rat PMCs, which provides a potential novel therapeutic approach for clinical investigation on renal failure treatment.
Collapse
Affiliation(s)
- Yanyan Xu
- Department of Nephrology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Wang
- Department of Nephrology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hong Ding
- Department of Nephrology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
11
|
Zhang Z, Li Z, Liu Z, Zhang X, Yu N, Xu Z. Identification of microenvironment-related genes with prognostic value in clear cell renal cell carcinoma. J Cell Biochem 2020; 121:3606-3615. [PMID: 31961022 DOI: 10.1002/jcb.29654] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 12/19/2019] [Indexed: 12/27/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney tumor. Previous studies have shown that the interaction between tumor cells and microenvironment has an important impact on prognosis. Immune and stromal cells are two vital components of the tumor microenvironment. Our study aimed to better understand and explore the genes involved in immune/stromal cells on prognosis. We used the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data algorithm to calculate immune/stromal scores. According to the scores, we divided ccRCC patients from The Cancer Genome Atlas database into low and high groups and identified the genes which were differentially expressed and significantly associated with prognosis. The result of functional enrichment analysis and protein-protein interaction networks indicated that these genes mainly were involved in extracellular matrix and regulation of cellular activities. Then another independent cohort from the International Cancer Genome Consortium database was used to validate these genes. Finally, we acquired a list of microenvironment-related genes that can predict prognosis for ccRCC patients.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Zeyan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiang Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Nengwang Yu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhonghua Xu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
12
|
He S, Ma C, Zhang L, Bai J, Wang X, Zheng X, Zhang J, Xin W, Li Y, Jiang Y, Wang S, Zhu D. GLI1-mediated pulmonary artery smooth muscle cell pyroptosis contributes to hypoxia-induced pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 318:L472-L482. [PMID: 31868509 DOI: 10.1152/ajplung.00405.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pulmonary hypertension (PH) is a clinically common malignant cardiovascular disease. Pyroptosis is a new form of inflammatory cell death that is involved in many disease processes. Glioma-associated oncogene family zinc finger 1 (GLI1) is a transcriptional activator that participates in many diseases, but its role has never been explored in inducing pyroptosis and the progress of PH. In this study, we used an animal model and cell molecular biology to determine the effect of GLI1 on chronic hypoxia-mediated PH progression and pulmonary artery smooth muscle cell (PASMC) pyroptosis. The major findings of the present study are as follows: Hypoxia induced aberrant expression of GLI1. The inhibition of GLI1 attenuated hypoxia-induced PH and PASMC pyroptosis. Meanwhile, GLI1 enhanced apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) expression by binding with its promoter. GLI1 may promote PASMC pyroptosis through ASC to affect the progression of PH. These findings may identify novel targets for molecular therapy of PH.
Collapse
Affiliation(s)
- Siyu He
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, People's Republic of China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, People's Republic of China
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Xiaoying Wang
- College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Xiaodong Zheng
- Department of Genetics and Cell Biology, Harbin Medical University (Daqing), Daqing, People's Republic of China
| | - Junting Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Wei Xin
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yiying Li
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yuan Jiang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Sen Wang
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, People's Republic of China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, People's Republic of China.,College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China.,State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Daqing, People's Republic of China.,Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
13
|
Maity G, Ghosh A, Gupta V, Haque I, Sarkar S, Das A, Dhar K, Bhavanasi S, Gunewardena SS, Von Hoff DD, Mallik S, Kambhampati S, Banerjee SK, Banerjee S. CYR61/CCN1 Regulates dCK and CTGF and Causes Gemcitabine-resistant Phenotype in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2019; 18:788-800. [PMID: 30787177 DOI: 10.1158/1535-7163.mct-18-0899] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/30/2018] [Accepted: 01/30/2019] [Indexed: 02/03/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) develops extrinsic- and intrinsic-resistant phenotypes to prevent chemotherapies from entering into the cells by promoting desmoplastic reactions (DR) and metabolic malfunctions of the drugs. It is well established that these responses are also associated with pancreatic cancer cells' gemcitabine resistance. However, the mechanism by which these resistant pathways function in the pancreatic cancer cells remains poorly understood. In these studies, we show that CYR61/CCN1 signaling plays a vital role in making pancreatic cancer cells resistant to gemcitabine in vitro and also in a tumor xenograft model. We proved that the catastrophic effect of gemcitabine could significantly be increased in gemcitabine-resistant PDAC cells when CYR61/CCN1 is depleted, while this effect can be suppressed in gemcitabine-sensitive neoplastic cells by treating them with CYR61/CCN1 recombinant protein. Ironically, nontransformed pancreatic cells, which are sensitive to gemcitabine, cannot be resistant to gemcitabine by CYR61/CCN1 protein treatment, showing a unique feature of CYR61/CCN signaling that only influences PDAC cells to become resistant. Furthermore, we demonstrated that CYR61/CCN1 suppresses the expression of the gemcitabine-activating enzyme deoxycytidine kinase (dCK) while it induces the expression of a DR-promoting factor CTGF (connective tissue growth factor) in pancreatic cancer cells in vitro and in vivo Thus, the previously described mechanisms (dCK and CTGF pathways) for gemcitabine resistance may be two novel targets for CYR61/CCN1 to protect pancreatic cancer cells from gemcitabine. Collectively, these studies reveal a novel paradigm in which CYR61/CCN1regulates both extrinsic and intrinsic gemcitabine resistance in PDAC cells by employing unique signaling pathways.
Collapse
Affiliation(s)
- Gargi Maity
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Vijayalaxmi Gupta
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
- Department of Ob/Gyn, University of Kansas Medical Center, Kansas City, Kansas
| | - Inamul Haque
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Sandipto Sarkar
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Amlan Das
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
| | - Kakali Dhar
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
| | - Sneha Bhavanasi
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
| | - Sumedha S Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Daniel D Von Hoff
- The Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Suman Kambhampati
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri
- The Sarah Cannon Cancer Center at HCA Midwest Health, Kansas City, Missouri
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
14
|
Triptonide inhibits lung cancer cell tumorigenicity by selectively attenuating the Shh-Gli1 signaling pathway. Toxicol Appl Pharmacol 2019; 365:1-8. [DOI: 10.1016/j.taap.2019.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 12/20/2018] [Accepted: 01/01/2019] [Indexed: 12/16/2022]
|
15
|
Guo J, Wu Y, Du J, Yang L, Chen W, Gong K, Dai J, Miao S, Jin D, Xi S. Deregulation of UBE2C-mediated autophagy repression aggravates NSCLC progression. Oncogenesis 2018; 7:49. [PMID: 29904125 PMCID: PMC6002383 DOI: 10.1038/s41389-018-0054-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/12/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023] Open
Abstract
The roles of aberrantly regulated autophagy in human malignancy and the mechanisms that initiate and sustain the repression of autophagy in carcinogenesis are less well defined. Activation of the oncogene UBE2C and repression of autophagy are concurrently underlying the initiation, progression, and metastasis of lung cancer and exploration of essential association of UBE2C with autophagy will confer more options in searching novel molecular therapeutic targets in lung cancer. Here we report that aberrant activation of UBE2C in lung tumors from patients associates with adverse prognosis and enhances cell proliferation, clonogenicity, and invasive growth of NSCLC. UBE2C selectively represses autophagy in NSCLC and disruption of UBE2C-mediated autophagy repression attenuates cell proliferation, clonogenicity, and invasive growth of NSCLC. Autophagy repression is essentially involved in UBE2C-induced cell proliferation, clonogenicity, and invasive growth of NSCLC. Interference of UBE2C-autophagy repression axis by Norcantharidin arrests NSCLC progression. UBE2C is repressed post-transcriptionally via tumor suppressor miR-381 and epitranscriptionally stabilized with maintenance of lower m6A level within its mature RNAs due to the upregulation of m6A demethylase ALKBH5 in NSCLC. Collectively, our results indicated that deregulated UBE2C-autophagy repression axis drives NSCLC progression which renders varieties of potential molecular targets in cancer therapy of NSCLC.
Collapse
Affiliation(s)
- Jiwei Guo
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Yan Wu
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Jing Du
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Lijuan Yang
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Weiwei Chen
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Kaikai Gong
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Juanjuan Dai
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Shuang Miao
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Dan Jin
- Department of Pain Ward, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China
| | - Sichuan Xi
- Cancer Research Institute, Binzhou Medical University Hospital, 256603, Binzhou, P.R. China.
| |
Collapse
|
16
|
Mastrangelo E, Milani M. Role and inhibition of GLI1 protein in cancer. LUNG CANCER-TARGETS AND THERAPY 2018; 9:35-43. [PMID: 29628779 PMCID: PMC5877502 DOI: 10.2147/lctt.s124483] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
GLI1 is a transcriptional regulator involved in the development of different types of cancer. GLI1 transcriptional activity is regulated within the Hedgehog pathway (canonical activity), but can also be controlled independently (non-canonical activity) in the context of other signaling pathways. Experimental evidences show GLI1 involvement in both small- and non–small-cell lung cancers. Direct inhibition of the protein, in combination with other chemotherapeutic agents, represents a promising strategy for the treatment of different malignancies.
Collapse
Affiliation(s)
- Eloise Mastrangelo
- CNR - Biophysics Institute, c/o Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Mario Milani
- CNR - Biophysics Institute, c/o Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
17
|
Sodium selenite attenuates lung adenocarcinoma progression by repressing SOX2-mediated stemness. Cancer Chemother Pharmacol 2018; 81:885-895. [PMID: 29546459 DOI: 10.1007/s00280-018-3561-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/11/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Sodium selenite (SS) has been widely reported to induce apoptosis in various cancer cell types. However, the underlying molecular mechanisms governing SS-mediated repression of lung cancer stem cells remain largely undefined. METHODS In vitro assays of cell proliferation, clonal formation, apoptosis, migration and cancer stemness cell sphere formation were performed to examine the inhibitory effects of SS on lung adenocarcinoma (LAD) cells with or without the overexpression of SRY-related high-mobility-group box 2 (SOX2). RESULTS SS significantly inhibited cell growth and induced apoptosis in LAD cells in a dose-dependent manner with marginal effects on normal epithelial cell HBEC. SS dramatically repressed expression of SOX2 and its upstream regulator GLI1 and strongly decreased stemness sphere formation in LAD cells at 10 µM. Forced expression of SOX2 significantly buffered anti-cancer effects of SS. CONCLUSIONS Our results demonstrate that SS attenuates lung adenocarcinoma progression by repressing SOX2 and its upstream regulator GLI1, which suggests that SS may be a potential therapeutic drug candidate for lung cancer patients.
Collapse
|