1
|
Jiang P, Ye S, Fan X, Tian Y, Zhang D, Pan W. Schistosoma japonicum infection-mediated downregulation of lncRNA Malat1 contributes to schistosomiasis hepatic fibrosis by the Malat1/miR-96/Smad7 pathway. Parasit Vectors 2024; 17:413. [PMID: 39363237 PMCID: PMC11451255 DOI: 10.1186/s13071-024-06499-9] [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: 07/25/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Schistosoma japonicum infection causes hepatic fibrosis, a primary cause of morbidity and mortality associated with the disease, and effective treatments are still lacking. Long non-coding RNAs (lncRNAs) have been implicated in the pathogenic process of various tissue fibroses. However, the role of lncRNAs in schistosomiasis hepatic fibrosis (HF) is poorly understood. Understanding the role of lncRNAs in schistosomiasis HF will enhance knowledge of disease processes and aid in the discovery of therapeutic targets and diagnostic biomarkers. METHODS Differentially expressed lncRNA profiles in primary hepatic stellate cells (HSCs) of mice infected with S. japonicum were identified using high-throughput lncRNA sequencing. Primary HSCs were isolated from infected mice using collagenase digestion and density-gradient centrifugation, cultured in DMEM with 10% fetal bovine serum. Dual-luciferase reporter assays, nuclear cytoplasm fractionation and RIP assays were employed to assess the relationship between Malat1 and miRNA-96. Malat1 lentivirus and ASO-Malat1 were constructed for forced expression and downregulated expression of Malat1. The Malat1-KO mouse was constructed by CRISPR/Cas9 technology. Pathological features of the liver were evaluated by hematoxylin-eosin (HE), Masson's trichrome staining and immunohistochemistry (IHC). The expression levels of fibrosis-related genes were determined by quantitative real-time PCR (qRT-PCR) and Western blot. RESULTS A total of 1561 differentially expressed lncRNAs were identified between infected and uninfected primary HSCs. Among the top altered lncRNAs, the downregulated Malat1 was observed in infected HSCs and verified by qPCR. Treatment of infected mice with praziquantel (PZQ) significantly increased the Malat1 expression. Elevated Malat1 expression in infected primary HSC reduced the expressions of profibrogenic genes, whereas Malat1 knockdown had the opposite effect. Moreover, Malat1 was found to interact with miR-96, a profibrotic miRNA, by targeting Smad7. Forced Malat1 expression reduced miR-96 levels in infected primary HSCs, attenuating fibrogenesis and showing negative correlation between Malat1 expression and the expression levels of miR-96 and profibrogenic genes α-SMA and Col1α1. Notably, in Malat1-KO mice, knockout of Malat1 aggravates schistosomiasis HF, while restored Malat1 expression in the infected HSCs reduced the expression of profibrogenic genes. CONCLUSIONS We demonstrate that lncRNA is involved in regulation of schistosomiasis HF. Elevated lncRNA Malat1 expression in infected HSCs reduces fibrosis via the Malat1/miR-96/Smad7 pathway, thus providing a novel therapeutic target for schistosomiasis HF. Furthermore, Malat1 expression is sensitive to PZQ treatment, thus offering a potential biomarker for assessing the response to chemotherapy.
Collapse
Affiliation(s)
- Pengyue Jiang
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, China
| | - Shengyu Ye
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| | - Xiaobin Fan
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| | - Yini Tian
- Department of Tropical Diseases, Naval Medical University, Shanghai, China
| | - Dongmei Zhang
- Department of Tropical Diseases, Naval Medical University, Shanghai, China.
| | - Weiqing Pan
- Department of Tropical Diseases, Naval Medical University, Shanghai, China.
- Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Huang L, Liu X, Chen Q, Yang J, Zhang D, Zhao Y, Xu L, Li Z, Liu X, Shao S, Li D, Song Y, Liu X, Zhan Q. TGF-β-induced lncRNA TBUR1 promotes EMT and metastasis in lung adenocarcinoma via hnRNPC-mediated GRB2 mRNA stabilization. Cancer Lett 2024; 600:217153. [PMID: 39102940 DOI: 10.1016/j.canlet.2024.217153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 08/07/2024]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway is pivotal in inducing epithelial-mesenchymal transition (EMT) and promoting cancer metastasis. Long non-coding RNAs (lncRNAs) have emerged as significant players in these processes, yet their precise mechanisms remain elusive. Here, we demonstrate that TGF-β-upregulated lncRNA 1 (TBUR1) is significantly activated by TGF-β via Smad3/4 signaling in lung adenocarcinoma (LUAD) cells. Functionally, TBUR1 triggers EMT, enhances LUAD cell migration and invasion in vitro, and promotes metastasis in nude mice. Mechanistically, TBUR1 interacts with heterogeneous nuclear ribonucleoprotein C (hnRNPC) to stabilize GRB2 mRNA in an m6A-dependent manner. Clinically, TBUR1 is upregulated in LUAD tissues and correlates with poor prognosis, highlighting its potential as a prognostic biomarker and therapeutic target for LUAD. Taken together, our findings underscore the crucial role of TBUR1 in mediating TGF-β-induced EMT and metastasis in LUAD, providing insights for future therapeutic interventions.
Collapse
Affiliation(s)
- Lijie Huang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Xiaoxu Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Qiuying Chen
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Jingyu Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Dongdong Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yabing Zhao
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Lele Xu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Zhangfu Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xinyuan Liu
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Shujuan Shao
- Liaoning Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Dan Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xuefeng Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China.
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Soochow University Cancer Institute, Suzhou 215000, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
3
|
Aftabi S, Barzegar Behrooz A, Cordani M, Rahiman N, Sadeghdoust M, Aligolighasemabadi F, Pistorius S, Alavizadeh SH, Taefehshokr N, Ghavami S. Therapeutic targeting of TGF-β in lung cancer. FEBS J 2024. [PMID: 39083441 DOI: 10.1111/febs.17234] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Transforming growth factor-β (TGF-β) plays a complex role in lung cancer pathophysiology, initially acting as a tumor suppressor by inhibiting early-stage tumor growth. However, its role evolves in the advanced stages of the disease, where it contributes to tumor progression not by directly promoting cell proliferation but by enhancing epithelial-mesenchymal transition (EMT) and creating a conducive tumor microenvironment. While EMT is typically associated with enhanced migratory and invasive capabilities rather than proliferation per se, TGF-β's influence on this process facilitates the complex dynamics of tumor metastasis. Additionally, TGF-β impacts the tumor microenvironment by interacting with immune cells, a process influenced by genetic and epigenetic changes within tumor cells. This interaction highlights its role in immune evasion and chemoresistance, further complicating lung cancer therapy. This review provides a critical overview of recent findings on TGF-β's involvement in lung cancer, its contribution to chemoresistance, and its modulation of the immune response. Despite the considerable challenges encountered in clinical trials and the development of new treatments targeting the TGF-β pathway, this review highlights the necessity for continued, in-depth investigation into the roles of TGF-β. A deeper comprehension of these roles may lead to novel, targeted therapies for lung cancer. Despite the intricate behavior of TGF-β signaling in tumors and previous challenges, further research could yield innovative treatment strategies.
Collapse
Affiliation(s)
- Sajjad Aftabi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Canada
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Iran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University, Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Iran
| | - Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Farnaz Aligolighasemabadi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Canada
| | - Stephen Pistorius
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Canada
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Iran
| | - Nima Taefehshokr
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, University of Manitoba, Winnipeg, Canada
- Faculty Academy of Silesia, Faculty of Medicine, Katowice, Poland
- Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
4
|
Bracken CP, Goodall GJ, Gregory PA. RNA regulatory mechanisms controlling TGF-β signaling and EMT in cancer. Semin Cancer Biol 2024; 102-103:4-16. [PMID: 38917876 DOI: 10.1016/j.semcancer.2024.06.001] [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: 12/15/2023] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a major contributor to metastatic progression and is prominently regulated by TGF-β signalling. Both EMT and TGF-β pathway components are tightly controlled by non-coding RNAs - including microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) - that collectively have major impacts on gene expression and resulting cellular states. While miRNAs are the best characterised regulators of EMT and TGF-β signaling and the miR-200-ZEB1/2 feedback loop plays a central role, important functions for lncRNAs and circRNAs are also now emerging. This review will summarise our current understanding of the roles of non-coding RNAs in EMT and TGF-β signaling with a focus on their functions in cancer progression.
Collapse
Affiliation(s)
- Cameron P Bracken
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA 5000, Australia.
| | - Gregory J Goodall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA 5000, Australia.
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
5
|
Wang M, Ding X, Fang X, Xu J, Chen Y, Qian Y, Zhang J, Yu D, Zhang X, Ma X, Zhu T, Gu J, Zhang X. Circ6834 suppresses non-small cell lung cancer progression by destabilizing ANHAK and regulating miR-873-5p/TXNIP axis. Mol Cancer 2024; 23:128. [PMID: 38890620 PMCID: PMC11184876 DOI: 10.1186/s12943-024-02038-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) play important roles in cancer progression and metastasis. However, the expression profiles and biological roles of circRNAs in non-small cell lung cancer (NSCLC) remain unclear. METHODS In this study, we identified a novel circRNA, hsa_circ_0006834 (termed circ6834), in NSCLC by RNA-seq and investigated the biological role of circ6834 in NSCLC progression in vitro and in vivo. Finally, the molecular mechanism of circ6834 was revealed by tagged RNA affinity purification (TRAP), western blot, RNA immunoprecipitation, dual luciferase reporter gene assays and rescue experiments. RESULTS Our results showed that circ6834 was downregulated in NSCLC tumor tissues and cell lines. Circ6834 overexpression inhibited NSCLC cell growth and metastasis both in vitro and in vivo, while circ6834 knockdown had the opposite effect. We found that TGF-β treatment decreased circ6834 expression, which was associated with the QKI reduction in NSCLC cells and circ6834 antagonized TGF-β-induced EMT and metastasis in NSCLC cells. Mechanistically, circ6834 bound to AHNAK protein, a key regulator of TGF-β/Smad signaling, and inhibited its stability by enhancing TRIM25-mediated ubiquitination and degradation. In addition, circ6834 acted as a miRNA sponge for miR-873-5p and upregulated TXNIP gene expression, which together inactivated the TGF-β/Smad signaling pathway in NSCLC cells. CONCLUSION In conclusion, circ6834 is a tumor-suppressive circRNA that inhibits NSCLC progression by forming a negative regulatory feedback loop with the TGF-β/Smad signaling pathway and represents a novel therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Maoye Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoge Ding
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xinjian Fang
- Department of Oncology, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, 211300, China
| | - Jing Xu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yanke Chen
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Qian
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Jiahui Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Dan Yu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiaoxin Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Xiuqin Ma
- Department of Pulmonary and Critical Care Medicine, Yixing Hospital affiliated to Jiangsu University, Yixing, 214200, China
| | - Taofeng Zhu
- Department of Pulmonary and Critical Care Medicine, Yixing Hospital affiliated to Jiangsu University, Yixing, 214200, China.
| | - Jianmei Gu
- Departmemt of Clinical Laboratory Medicine, Nantong Tumor Hospital/Affiliated Tumor Hospital of Nantong University, Nantong, 226300, China.
| | - Xu Zhang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
6
|
Alhajlah S. The molecular mechanisms of various long non-coding RNA (lncRNA) in human lung tumors: Shedding light on the molecular mechanisms. Pathol Res Pract 2024; 256:155253. [PMID: 38513578 DOI: 10.1016/j.prp.2024.155253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024]
Abstract
Although it is still mostly incomplete, unraveling the gene expression networks controlling the initiation and progression of cancer is crucial. The rapid identification and characterization of long noncoding RNAs (lncRNAs) is made possible by advancements in computational biology and RNA-seq technology. According to recent research, lncRNAs are involved in several stages in the genesis of lung cancer. These lncRNAs interact with DNA, RNA, protein molecules, and/or their combinations. They play a crucial role in transcriptional and post-transcriptional regulation, as well as chromatin architecture. Their misexpression gives cancer cells the ability to start, grow, and spread tumors. This review will focus on their abnormal expression and function in lung cancer, as well as their involvement in cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia.
| |
Collapse
|
7
|
Song B, Wei F, Peng J, Wei X, Liu M, Nie Z, Ma Y, Peng T. Icariin Regulates EMT and Stem Cell-Like Character in Breast Cancer through Modulating lncRNA NEAT1/TGFβ/SMAD2 Signaling Pathway. Biol Pharm Bull 2024; 47:399-410. [PMID: 38220208 DOI: 10.1248/bpb.b23-00668] [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] [Indexed: 01/16/2024]
Abstract
Metastases and drug resistance are the major risk factors associated with breast cancer (BC), which is the most common type of tumor affecting females. Icariin (ICA) is a traditional Chinese medicine compound that possesses significant anticancer properties. Long non-coding RNAs (lncRNAs) are involved in a wide variety of biological and pathological processes and have been shown to modulate the effectiveness of certain drugs in cancer. The purpose of this study was to examine the potential effect of ICA on epithelial mesenchymal transition (EMT) and stemness articulation in BC cells, as well as the possible relationship between its inhibitory action on EMT and stemness with the NEAT1/transforming growth factor β (TGFβ)/SMAD2 pathway. The effect of ICA on the proliferation (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and colony assays), EMT (Western blotting, immunofluorescence, and wound healing), and stemness (mammosphere formation assays, Western blotting) of BC cells were examined. According to the findings, ICA suppressed the proliferation, EMT, and stem cell-like in MDA-MB-231 cells, and exerted its inhibitory impact by downregulating the TGFβ/SMAD2 signaling pathway. ICA could significantly downregulate the expression of lncRNA NEAT1, and silencing NEAT1 enhanced the effect of ICA in suppressing EMT and expression of different stem cell markers. In addition, silencing NEAT1 was found to attenuate the TGFβ/SMAD2 signaling pathway, thereby improving the inhibitory impact of ICA on stemness and EMT in BC cells. In conclusion, ICA can potentially inhibit the metastasis of BC via affecting the NEAT1/TGFβ/SMAD2 pathway, which provides a theoretical foundation for understanding the mechanisms involved in potential application of ICA for BC therapy.
Collapse
Affiliation(s)
- Bo Song
- School of Third Clinical Medicine, Shanxi University of Chinese Medicine
| | - Fuxia Wei
- School of Third Clinical Medicine, Shanxi University of Chinese Medicine
| | - Jiehao Peng
- School of Third Clinical Medicine, Shanxi University of Chinese Medicine
| | - Xiuhong Wei
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine
| | - Mingran Liu
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine
| | - Zhongbiao Nie
- Pharmaceutical Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University
| | - Yanmiao Ma
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine
| | - Tao Peng
- Famous Chinese Medicine Studio, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Hospital of Integrated Traditional Chinese and Western Medicine
| |
Collapse
|
8
|
Huang L, Wang Z, Liao C, Zhao Z, Gao H, Huang R, Chen J, Wu F, Zeng F, Zhang Y, Jiang T, Hu H. PVT1 promotes proliferation and macrophage immunosuppressive polarization through STAT1 and CX3CL1 regulation in glioblastoma multiforme. CNS Neurosci Ther 2024; 30:e14566. [PMID: 38287522 PMCID: PMC10805395 DOI: 10.1111/cns.14566] [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: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/30/2023] [Indexed: 01/31/2024] Open
Abstract
AIMS This study aimed to investigate the role of plasmacytoma variant translocation 1 (PVT1), a long non-coding RNA, in glioblastoma multiforme (GBM) and its impact on the tumor microenvironment (TME). METHODS We assessed aberrant PVT1 expression in glioma tissues and its impact on GBM cell growth in vitro and in vivo. Additionally, we investigated PVT1's role in influencing glioma-associated macrophages. To understand PVT1's role in cell growth and the immunosuppressive TME, we performed a series of comprehensive experiments. RESULTS PVT1 was overexpressed in GBM due to copy number amplification, correlating with poor prognosis. Elevated PVT1 promoted GBM cell proliferation, while its downregulation inhibited growth in vitro and in vivo. PVT1 inhibited type I interferon-stimulated genes (ISGs), with STAT1 as the central hub. PVT1 correlated with macrophage enrichment and regulated CX3CL1 expression, promoting recruitment and M2 phenotype polarization of macrophages. PVT1 localized to the cell nucleus and bound to DHX9, enriching at the promoter regions of STAT1 and CX3CL1, modulating ISGs and CX3CL1 expression. CONCLUSION PVT1 plays a significant role in GBM, correlating with poor prognosis, promoting cell growth, and shaping an immunosuppressive TME via STAT1 and CX3CL1 regulation. Targeting PVT1 may hold therapeutic promise for GBM patients.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Pathophysiology, Beijing Neurosurgical InstituteCapital Medical UniversityChina
| | - Zheng Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Chihyi Liao
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Hua Gao
- Department of Cell Biology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Ruoyu Huang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Jing Chen
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Fan Zeng
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Ying Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Center of Brain TumorBeijing Institute for Brain DisordersBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Chinese Glioma Genome Atlas Network and Asian Glioma Genome Atlas NetworkBeijingChina
| | - Huimin Hu
- Department of Molecular Neuropathology, Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
| |
Collapse
|
9
|
Li C, Fu Y, He Y, Huang N, Yue J, Miao Y, Lv J, Xiao Y, Deng R, Zhang C, Huang M. Knockdown of LINC00511 enhances radiosensitivity of lung adenocarcinoma via regulating miR-497-5p/SMAD3. Cancer Biol Ther 2023; 24:2165896. [PMID: 36861928 PMCID: PMC9988350 DOI: 10.1080/15384047.2023.2165896] [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] [Indexed: 03/03/2023] Open
Abstract
As the most common histological subtype of primary lung cancer, lung adenocarcinoma (LUAD) causes enormous cancer deaths worldwide. Radiotherapy has been frequently used in LUAD cases, and radiosensitivity is vital for LUAD therapy. This research sought to explore the genetic factors affecting radiosensitivity in LUAD and inner mechanisms. LINC00511, miR-497-5p, and SMAD3 expression in LUAD cells were detected via qRT-PCR and western blot. CCK-8 assays, colony formation, and flow cytometry assays were employed to explore the cell viability, apoptosis, and radiosensitivity in PC-9 and A549 cells. The targeting relationship between LINC00511, miR-497-5p, and SMAD3 was verified by dual luciferase reporter assay. Furthermore, xenograft experiments were performed for the in vivo verification. In conclusion, LINC00511 was overexpressed in LUAD cells, which downregulated downstream miR-497-5p expression and mediately led to SMAD3 activation. LINC00511 downregulation suppressed cell viability while enhanced apoptosis rate in LUAD cells. Also, LINC00511 and SMAD3 were overexpressed, while miR-497-5p was downregulated in LUAD cells exposed to 4Gy irradiation treatment. Moreover, LINC00511 inhibition could block SMAD3 expression and promoted the radiosensitivity both in vitro and in vivo. These findings uncover LINC00511 knockdown promoted miR-497-5p expression and subsequently led to lower SMAD3 level, which enhanced radiosensitivity in LUAD cells. LINC00511/miR-497-5p/SMAD3 axis could be of considerable potential to enhance radiosensitivity in LUAD.
Collapse
Affiliation(s)
- Chongxin Li
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Yanyan Fu
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Yongmei He
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Nan Huang
- Department of Pulmonary, the Shizong Hospital of First People’s Hospital in Qujing, Qujing, P.R. China
| | - Jun Yue
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Yi Miao
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Jialing Lv
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Youchuan Xiao
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Ruoyu Deng
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
| | - Chao Zhang
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
- CONTACT Chao zhang
| | - Meifang Huang
- Department of Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, P.R. China
- Meifang Huang Department of Surgical Oncology, the First People’s Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan655000, P.R. China
| |
Collapse
|
10
|
Ao YQ, Gao J, Jiang JH, Wang HK, Wang S, Ding JY. Comprehensive landscape and future perspective of long noncoding RNAs in non-small cell lung cancer: it takes a village. Mol Ther 2023; 31:3389-3413. [PMID: 37740493 PMCID: PMC10727995 DOI: 10.1016/j.ymthe.2023.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/01/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are a distinct subtype of RNA that lack protein-coding capacity but exert significant influence on various cellular processes. In non-small cell lung cancer (NSCLC), dysregulated lncRNAs act as either oncogenes or tumor suppressors, contributing to tumorigenesis and tumor progression. LncRNAs directly modulate gene expression, act as competitive endogenous RNAs by interacting with microRNAs or proteins, and associate with RNA binding proteins. Moreover, lncRNAs can reshape the tumor immune microenvironment and influence cellular metabolism, cancer cell stemness, and angiogenesis by engaging various signaling pathways. Notably, lncRNAs have shown great potential as diagnostic or prognostic biomarkers in liquid biopsies and therapeutic strategies for NSCLC. This comprehensive review elucidates the significant roles and diverse mechanisms of lncRNAs in NSCLC. Furthermore, we provide insights into the clinical relevance, current research progress, limitations, innovative research approaches, and future perspectives for targeting lncRNAs in NSCLC. By summarizing the existing knowledge and advancements, we aim to enhance the understanding of the pivotal roles played by lncRNAs in NSCLC and stimulate further research in this field. Ultimately, unraveling the complex network of lncRNA-mediated regulatory mechanisms in NSCLC could potentially lead to the development of novel diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Yong-Qiang Ao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Gao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia-Hao Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hai-Kun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Shuai Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jian-Yong Ding
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Zefferino R, Conese M. A Vaccine against Cancer: Can There Be a Possible Strategy to Face the Challenge? Possible Targets and Paradoxical Effects. Vaccines (Basel) 2023; 11:1701. [PMID: 38006033 PMCID: PMC10674257 DOI: 10.3390/vaccines11111701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Is it possible to have an available vaccine that eradicates cancer? Starting from this question, this article tries to verify the state of the art, proposing a different approach to the issue. The variety of cancers and different and often unknown causes of cancer impede, except in some cited cases, the creation of a classical vaccine directed at the causative agent. The efforts of the scientific community are oriented toward stimulating the immune systems of patients, thereby preventing immune evasion, and heightening chemotherapeutic agents effects against cancer. However, the results are not decisive, because without any warning signs, metastasis often occurs. The purpose of this paper is to elaborate on a vaccine that must be administered to a patient in order to prevent metastasis; metastasis is an event that leads to death, and thus, preventing it could transform cancer into a chronic disease. We underline the fact that the field has not been studied in depth, and that the complexity of metastatic processes should not be underestimated. Then, with the aim of identifying the target of a cancer vaccine, we draw attention to the presence of the paradoxical actions of different mechanisms, pathways, molecules, and immune and non-immune cells characteristic of the tumor microenvironment at the primary site and pre-metastatic niche in order to exclude possible vaccine candidates that have opposite effects/behaviors; after a meticulous evaluation, we propose possible targets to develop a metastasis-targeting vaccine. We conclude that a change in the current concept of a cancer vaccine is needed, and the efforts of the scientific community should be redirected toward a metastasis-targeting vaccine, with the increasing hope of eradicating cancer.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
12
|
Fan C, González-Prieto R, Kuipers TB, Vertegaal ACO, van Veelen PA, Mei H, Ten Dijke P. The lncRNA LETS1 promotes TGF-β-induced EMT and cancer cell migration by transcriptionally activating a TβR1-stabilizing mechanism. Sci Signal 2023; 16:eadf1947. [PMID: 37339182 DOI: 10.1126/scisignal.adf1947] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/25/2023] [Indexed: 06/22/2023]
Abstract
Transforming growth factor-β (TGF-β) signaling is a critical driver of epithelial-to-mesenchymal transition (EMT) and cancer progression. In SMAD-dependent TGF-β signaling, activation of the TGF-β receptor complex stimulates the phosphorylation of the intracellular receptor-associated SMADs (SMAD2 and SMAD3), which translocate to the nucleus to promote target gene expression. SMAD7 inhibits signaling through the pathway by promoting the polyubiquitination of the TGF-β type I receptor (TβRI). We identified an unannotated nuclear long noncoding RNA (lncRNA) that we designated LETS1 (lncRNA enforcing TGF-β signaling 1) that was not only increased but also perpetuated by TGF-β signaling. Loss of LETS1 attenuated TGF-β-induced EMT and migration in breast and lung cancer cells in vitro and extravasation of the cells in a zebrafish xenograft model. LETS1 potentiated TGF-β-SMAD signaling by stabilizing cell surface TβRI, thereby forming a positive feedback loop. Specifically, LETS1 inhibited TβRI polyubiquitination by binding to nuclear factor of activated T cells (NFAT5) and inducing the expression of the gene encoding the orphan nuclear receptor 4A1 (NR4A1), a component of a destruction complex for SMAD7. Overall, our findings characterize LETS1 as an EMT-promoting lncRNA that potentiates signaling through TGF-β receptor complexes.
Collapse
Affiliation(s)
- Chuannan Fan
- Department of Cell and Chemical Biology, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
- Oncode Institute, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
| | - Román González-Prieto
- Department of Cell and Chemical Biology, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
- Genome Proteomics Laboratory, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Seville, Américo Vespucio 24, 41092 Seville, Spain
- Department of Cell Biology, University of Seville, Américo Vespucio 24, 41092 Seville, Spain
| | - Thomas B Kuipers
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
| | - Alfred C O Vertegaal
- Department of Cell and Chemical Biology, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
| | - Hailiang Mei
- Department of Biomedical Data Sciences, Sequencing Analysis Support Core, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
- Oncode Institute, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, Netherlands
| |
Collapse
|
13
|
Entezari M, Taheriazam A, Paskeh MDA, Sabouni E, Zandieh MA, Aboutalebi M, Kakavand A, Rezaei S, Hejazi ES, Saebfar H, Salimimoghadam S, Mirzaei S, Hashemi M, Samarghandian S. The pharmacological and biological importance of EZH2 signaling in lung cancer. Biomed Pharmacother 2023; 160:114313. [PMID: 36738498 DOI: 10.1016/j.biopha.2023.114313] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Up to 18% of cancer-related deaths worldwide are attributed to lung tumor and global burden of this type of cancer is ascending. Different factors are responsible for development of lung cancer such as smoking, environmental factors and genetic mutations. EZH2 is a vital protein with catalytic activity and belongs to PCR2 family. EZH2 has been implicated in regulating gene expression by binding to promoter of targets. The importance of EZH2 in lung cancer is discussed in current manuscript. Activation of EZH2 significantly elevates the proliferation rate of lung cancer. Furthermore, metastasis and associated molecular mechanisms including EMT undergo activation by EZH2 in enhancing the lung cancer progression. The response of lung cancer to therapy can be significantly diminished due to EZH2 upregulation. Since EZH2 increases tumor progression, anti-cancer agents suppressing its expression reduce malignancy. In spite of significant effort in understanding modulatory function of EZH2 on other pathways, it appears that EZH2 can be also regulated and controlled by other factors that are described in current review. Therefore, translating current findings to clinic can improve treatment and management of lung cancer patients.
Collapse
Affiliation(s)
- Maliheh Entezari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Eisa Sabouni
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Maryam Aboutalebi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirabbas Kakavand
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shamin Rezaei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elahe Sadat Hejazi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Saebfar
- European University Association, League of European Research Universities, university of milan, Italy
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
14
|
Hu Z, Liu Y, Liu M, Zhang Y, Wang C. Roles of TGF‑β signalling pathway‑related lncRNAs in cancer (Review). Oncol Lett 2023; 25:107. [PMID: 36817052 PMCID: PMC9932718 DOI: 10.3892/ol.2023.13693] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are a class of RNAs that are >200 nucleotides in length that do not have the ability to be translated into protein but are associated with numerous diseases, including cancer. The involvement of lncRNAs in the signalling of certain signalling pathways can promote tumour progression; these pathways include the transforming growth factor (TGF)-β signalling pathway, which is related to tumour development. The expression of lncRNAs in various tumour tissues is specific, and their interaction with the TGF-β signalling pathway indicates that they may serve as new tumour markers and therapeutic targets. The present review summarized the role of TGF-β pathway-associated lncRNAs in regulating tumorigenesis in different types of cancer and their effects on the TGF-β signalling pathway.
Collapse
Affiliation(s)
- Zhizhong Hu
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yitong Liu
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Meiqi Liu
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yang Zhang
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China,Correspondence to: Dr Yang Zhang or Dr Chengkun Wang, Cancer Research Institute, Medical School, University of South China, 28 Chang Sheng Xi Avenue, Hengyang, Hunan 421001, P.R. China, E-mail:
| | - Chengkun Wang
- Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China,Correspondence to: Dr Yang Zhang or Dr Chengkun Wang, Cancer Research Institute, Medical School, University of South China, 28 Chang Sheng Xi Avenue, Hengyang, Hunan 421001, P.R. China, E-mail:
| |
Collapse
|
15
|
A Regulatory Network Analysis of the Importance of USP15 in Breast Cancer Metastasis and Prognosis. JOURNAL OF ONCOLOGY 2022; 2022:1427726. [PMID: 36213818 PMCID: PMC9536986 DOI: 10.1155/2022/1427726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/04/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022]
Abstract
Background Ubiquitin-specific protease15(USP15), is the 16th identified protease in the USP family and is a key protein in tumorigenesis. However, the predictive value and regulatory mechanism of USP15 in breast cancer are unclear. Methods The GEPIA, UALCAN, GeneMANIA, and STRING databases were applied to explore the expression of USP15 in breast cancer and associated proteins. In addition, the TIMER database was evaluated for immune infiltration patterns. Moreover, protein immunoblotting assay, cell scratching assay, small compartment invasion assay, 3D stromal gel assay, immunoprecipitation assay, and immunohistochemistry (IHC) were used to USP15 regulatory mechanisms in breast cancer. Results In BRCA, several databases, including GEPIA and UALCAN, describe the upregulation of total protein levels and USP15 phosphorylation. In addition, the expression of USP15 was significantly correlated with gender and clinical stage. Overall survival (OS) was lower in patients with high USP15 expression. Functional network analysis showed that USP15 is involved in tumor-associated pathways, DNA replication, and cell cycle signaling through TGFβRI. In addition, USP15 expression was positively correlated with immune infiltration, including immune score, mesenchymal score, and several tumor-infiltrating lymphocytes (TIL). In addition, IHC results further confirmed the high expression of USP15 in breast cancer and its prognostic potential. Conclusions These findings demonstrate that high USP15 expression indicates poor prognosis in BRCA and reveal potential regulatory networks and the positive relationship with immune infiltration. Thus, USP15 may be an attractive predictor for BRCA.
Collapse
|
16
|
LINC00152 induced by TGF-β promotes metastasis via HuR in lung adenocarcinoma. Cell Death Dis 2022; 13:772. [PMID: 36071042 PMCID: PMC9452677 DOI: 10.1038/s41419-022-05164-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023]
Abstract
Lung adenocarcinoma (LUAD) is one of the main causes of cancer-related mortality, with a strong tendency to metastasize early. Transforming growth factor-β (TGF-β) signaling is a powerful regulator to promote metastasis of LUAD. Here, we screened long non-coding RNAs (lncRNAs) responsive to TGF-β and highly expressed in LUAD cells, and finally obtained our master molecular LINC00152. We proved that the TGF-β promoted transcription of LINC00152 through the classical TGF-β/SMAD3 signaling pathway and maintained its stability through the RNA-binding protein HuR. Moreover, LINC00152 increased ZEB1, SNAI1 and SNAI2 expression via increasing the interactions of HuR and these transcription factors, ultimately promoting epithelial-mesenchymal transition of LUAD cell and enhancing LUAD metastasis in vivo. These data provided evidence that LINC00152 induced by TGF-β promotes metastasis depending HuR in lung adenocarcinoma. Designing targeting LINC00152 and HuR inhibitors may therefore be an effective therapeutic strategy for LUAD treatment.
Collapse
|
17
|
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial–mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm (Beijing) 2022; 3:e144. [PMID: 35601657 PMCID: PMC9115588 DOI: 10.1002/mco2.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a program wherein epithelial cells lose their junctions and polarity while acquiring mesenchymal properties and invasive ability. Originally defined as an embryogenesis event, EMT has been recognized as a crucial process in tumor progression. During EMT, cell–cell junctions and cell–matrix attachments are disrupted, and the cytoskeleton is remodeled to enhance mobility of cells. This transition of phenotype is largely driven by a group of key transcription factors, typically Snail, Twist, and ZEB, through epigenetic repression of epithelial markers, transcriptional activation of matrix metalloproteinases, and reorganization of cytoskeleton. Mechanistically, EMT is orchestrated by multiple pathways, especially those involved in embryogenesis such as TGFβ, Wnt, Hedgehog, and Hippo, suggesting EMT as an intrinsic link between embryonic development and cancer progression. In addition, redox signaling has also emerged as critical EMT modulator. EMT confers cancer cells with increased metastatic potential and drug resistant capacity, which accounts for tumor recurrence in most clinic cases. Thus, targeting EMT can be a therapeutic option providing a chance of cure for cancer patients. Here, we introduce a brief history of EMT and summarize recent advances in understanding EMT mechanisms, as well as highlighting the therapeutic opportunities by targeting EMT in cancer treatment.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Chengwei Zhou
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Lin Liu
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| |
Collapse
|
18
|
Duan C. LncRNA SLC16A1-AS1 contributes to the progression of hepatocellular carcinoma cells by modulating miR-411/MITD1 axis. J Clin Lab Anal 2022; 36:e24344. [PMID: 35293026 PMCID: PMC8993651 DOI: 10.1002/jcla.24344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/23/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignancy with high morbidity. The current study aimed to explore the molecular mechanism of lncRNA SLC16A1‐AS1 in the tumorigenesis of HCC. Material and methods The expression of SLC16A1‐AS1 and miR‐411 was examined in clinical HCC tissues. HCC cell lines Hep3B and Huh‐7 were employed and transfected with si‐SLC16A1‐AS1. The correlation between SLC16A1‐AS1 and miR‐411 was verified by luciferase reporter assay. Cell viability was detected by CCK‐8 assay. Cell migration and invasion capacity were examined by transwell assay. The protein level of MITD1 was analyzed by western blotting. Results The expression of SLC16A1‐AS1 markedly increased in HCC tissues and cell lines. Subsequent studies identified SLC16A1‐AS1 as a downstream target of miR‐411. In addition, SLC16A1‐AS1 knockdown and miR‐411 overexpression significantly stagnated the progression of HCC cells. SLC16A1‐AS1 knockdown also downregulated MITD1 levels. Conclusion Our findings showed that SLC16A1‐AS1 was overexpressed in HCC cells and tissues. SLC16A1‐AS1 promoted the malignant characteristics of HCC cells and acted as an oncogene. Its regulatory effect may be associated with miR‐411/MITD1 axis. Therefore, SLC16A1‐AS1 has the potential to be used as a biomarker or therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Chun Duan
- Department of Infectious Diseases, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
19
|
Downregulation of LNMAS orchestrates partial EMT and immune escape from macrophage phagocytosis to promote lymph node metastasis of cervical cancer. Oncogene 2022; 41:1931-1943. [PMID: 35152264 PMCID: PMC8956512 DOI: 10.1038/s41388-022-02202-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/29/2021] [Accepted: 01/19/2022] [Indexed: 12/19/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is an essential step to drive the metastatic cascade to lymph nodes (LNs) in cervical cancer cells. However, few of them metastasize successfully partially due to increased susceptibility to immunosurveillance conferred by EMT. The precise mechanisms of cancer cells orchestrate EMT and immune evasion remain largely unexplored. In this study, we identified a lncRNA termed lymph node metastasis associated suppressor (LNMAS), which was downregulated in LN-positive cervical cancer patients and correlated with LN metastasis and prognosis. Functionally, LNMAS suppressed cervical cancer cells metastasis in vitro and in vivo. Mechanistically, LNMAS exerts its metastasis suppressive activity by competitively interacting with HMGB1 and abrogating the chromatin accessibility of TWIST1 and STC1, inhibiting TWIST1-mediated partial EMT and STC1-dependent immune escape from macrophage phagocytosis. We further demonstrated that the CpG sites in the promoter region of LNMAS was hypermethylated and contributed to the downregulation of LNMAS. Taken together, our results reveal the essential role of LNMAS in the LN metastasis of cervical cancer and provide mechanistic insights into the regulation of LNMAS in EMT and immune evasion.
Collapse
|
20
|
Wang T, Li Z, Yan L, Yan F, Shen H, Tian X. Long Non-Coding RNA Neighbor of BRCA1 Gene 2: A Crucial Regulator in Cancer Biology. Front Oncol 2021; 11:783526. [PMID: 34926299 PMCID: PMC8674783 DOI: 10.3389/fonc.2021.783526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are involved in fundamental biochemical and cellular processes. The neighbor of BRCA1 gene 2 (NBR2) is a long intergenic non-coding RNA (lincRNA) whose gene locus is adjacent to the tumor suppressor gene breast cancer susceptibility gene 1 (BRCA1). In human cancers, NBR2 expression is dysregulated and correlates with clinical outcomes. Moreover, NBR2 is crucial for glucose metabolism and affects the proliferation, survival, metastasis, and therapeutic resistance in different types of cancer. Here, we review the precise molecular mechanisms underlying NBR2-induced changes in cancer. In addition, the potential application of NBR2 in the diagnosis and treatment of cancer is also discussed, as well as the challenges of exploiting NBR2 for cancer intervention.
Collapse
Affiliation(s)
- Ting Wang
- Department of Laboratory Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaosheng Li
- Department of Laboratory Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Liujia Yan
- Department of Laboratory Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Yan
- Department of Laboratory Medicine, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xinyu Tian
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
21
|
Harnessing Carcinoma Cell Plasticity Mediated by TGF-β Signaling. Cancers (Basel) 2021; 13:cancers13143397. [PMID: 34298613 PMCID: PMC8307280 DOI: 10.3390/cancers13143397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary This review describes mechanisms driving epithelial plasticity in carcinoma mediated by transforming growth factor beta (TGF-β) signaling. Plasticity in carcinoma is frequently induced through epithelial–mesenchymal transition (EMT), an evolutionary conserved process in the development of multicellular organisms. The review explores the multifaceted functions of EMT, particularly focusing on the intermediate stages, which provide more adaptive responses of carcinoma cells in their microenvironment. The review critically considers how different intermediate or hybrid EMT stages confer carcinoma cells with stemness, refractoriness to therapies, and ability to execute all steps of the metastatic cascade. Finally, the review provides examples of therapeutic interventions based on the EMT concept. Abstract Epithelial cell plasticity, a hallmark of carcinoma progression, results in local and distant cancer dissemination. Carcinoma cell plasticity can be achieved through epithelial–mesenchymal transition (EMT), with cells positioned seemingly indiscriminately across the spectrum of EMT phenotypes. Different degrees of plasticity are achieved by transcriptional regulation and feedback-loops, which confer carcinoma cells with unique properties of tumor propagation and therapy resistance. Decoding the molecular and cellular basis of EMT in carcinoma should enable the discovery of new therapeutic strategies against cancer. In this review, we discuss the different attributes of plasticity in carcinoma and highlight the role of the canonical TGFβ receptor signaling pathway in the acquisition of plasticity. We emphasize the potential stochasticity of stemness in carcinoma in relation to plasticity and provide data from recent clinical trials that seek to target plasticity.
Collapse
|