1
|
Zhang J, Yang H, Zhang X, Chen J, Luo H, Li C, Chen X. Prognostic significance of copper metabolism-related genes as risk markers in bladder urothelial carcinoma. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 44:598-616. [PMID: 39120157 DOI: 10.1080/15257770.2024.2387783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Bladder urothelial carcinoma (BLCA), a prevalent malignant neoplasm affecting the human urinary system, is frequently linked with an unfavorable prognosis in a significant proportion of individuals. More effective and sensitive markers are needed to provide a reference for prognostic judgment. We obtained RNA sequencing data and clinical information of individuals from TCGA, and 133 copper metabolism-related genes from literature. Prognostic genes were evaluated by univariate/multivariate Cox regression analysis and LASSO analysis, and a risk-scoring model was established and validated in the GEO dataset. The CIBERSORT method was utilized to explore immune cell infiltration in BLCA individuals. In addition, tumor immune dysfunction and exclusion (TIDE) and immunophenoscore (IPS) were utilized to verify whether the model can foretell the response of BLCA individuals to immunotherapy. We successfully constructed an 8-gene risk scoring model to foretell individuals' overall survival, and the model performed well in TCGA training and GEO validation cohorts. Lastly, a nomogram containing clinical parameters and risk scores was constructed to help individualized result prediction for individuals. Calibration curves demonstrated a high degree of concordance between the observed and projected survival durations, attesting to its exceptional predictive accuracy. Analysis utilizing CIBERSORT unveiled elevated levels of immune cell infiltration in individuals classified as low risk. TIDE and IPS analyses substantiated that low-risk individuals exhibited a more favorable response to immunotherapy. In summary, the model held immense potential for stratifying the risk of survival and guiding tailored treatment approaches for individuals with BLCA, thereby offering valuable insights for personalized therapeutic interventions.
Collapse
Affiliation(s)
- Jiamo Zhang
- Department of Urology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Houwei Yang
- Department of Urology, Sinopharm Chongqing Southwest Aluminum Hospital, Chongqing, China
| | - Xuan Zhang
- Department of Urology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Jiangchuan Chen
- Department of Urology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Huaming Luo
- Department of Urology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Changlong Li
- Department of Urology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Chen
- Department of Urology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Yang C, Liu Y, Lv C, Xu M, Xu K, Shi J, Tan T, Zhou W, Lv D, Li Y, Xu J, Shao T. CanCellVar: A database for single-cell variants map in human cancer. Am J Hum Genet 2024; 111:1420-1430. [PMID: 38838674 PMCID: PMC11267512 DOI: 10.1016/j.ajhg.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024] Open
Abstract
Numerous variants, including both single-nucleotide variants (SNVs) in DNA and A>G RNA edits in mRNA as essential drivers of cellular proliferation and tumorigenesis, are commonly associated with cancer progression and growth. Thus, mining and summarizing single-cell variants will provide a refined and higher-resolution view of cancer and further contribute to precision medicine. Here, we established a database, CanCellVar, which aims to provide and visualize the comprehensive atlas of single-cell variants in tumor microenvironment. The current CanCellVar identified ∼3 million variants (∼1.4 million SNVs and ∼1.4 million A>G RNA edits) involved in 2,754,531 cells of 5 major cell types across 37 cancer types. CanCellVar provides the basic annotation information as well as cellular and molecular function properties of variants. In addition, the clinical relevance of variants can be obtained including tumor grade, treatment, metastasis, and others. Several flexible tools were also developed to aid retrieval and to analyze cell-cell interactions, gene expression, cell-development trajectories, regulation, and molecular structure affected by variants. Collectively, CanCellVar will serve as a valuable resource for investigating the functions and characteristics of single-cell variations and their roles in human tumor evolution and treatment.
Collapse
Affiliation(s)
- Changbo Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Yujie Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Chongwen Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Mengjia Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Kang Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Jingyi Shi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Tingting Tan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Weiwei Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Dezhong Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, Heilongjiang Province 150081, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China.
| | - Tingting Shao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
3
|
Merlano MC, Paccagnella M, Denaro N, Abbona A, Galizia D, Sangiolo D, Gammaitoni L, Fiorino E, Minei S, Bossi P, Licitra L, Garrone O. Baseline Values of Circulating IL-6 and TGF-β Might Identify Patients with HNSCC Who Do Not Benefit from Nivolumab Treatment. Cancers (Basel) 2023; 15:5257. [PMID: 37958430 PMCID: PMC10649732 DOI: 10.3390/cancers15215257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND The immunotherapy of head and neck cancer induces a limited rate of long-term survivors at the cost of treating many patients exposed to toxicity without benefit, regardless of PD-L1 expression. The identification of better biomarkers is warranted. We analyzed a panel of cytokines, chemokines and growth factors, hereinafter all referred to as 'cytokines', as potential biomarkers in patients with head and neck cancer treated with nivolumab. MATERIALS AND METHODS A total of 18 circulating cytokines were analyzed. Samples were gathered at baseline (T0) and after 3 courses of nivolumab (T1) in patients with relapsed/metastatic disease. The data extracted at T0 were linked to survival; the comparison of T0-T1 explored the effect of immunotherapy. RESULTS A total of 22 patients were accrued: 64% current heavy smokers, 36% female and 14% had PS = 2. At T0, ROC analysis showed that IL-6, IL-8, IL-10 and TGF-β were higher in patients with poor survival. Cox analysis demonstrated that only patients with the IL-6 and TGF-β discriminate had good or poor survival, respectively. Longitudinal increments of CCL-4, IL-15, IL-2 and CXCL-10 were observed in all patients during nivolumab treatment. CONCLUSION In this small population with poor clinical characteristics, this study highlights the prognostic role of IL-6 and TGF-β. Nivolumab treatment is associated with a positive modulation of some Th1 cytokines, but it does not correlate with the outcome.
Collapse
Affiliation(s)
- Marco Carlo Merlano
- Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy; (M.C.M.); (D.G.); (L.G.)
| | | | - Nerina Denaro
- Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (O.G.)
| | - Andrea Abbona
- Translational Oncology ARCO Foundation, 12100 Cuneo, Italy;
| | - Danilo Galizia
- Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy; (M.C.M.); (D.G.); (L.G.)
| | - Dario Sangiolo
- Department of Oncology, University of Turin, 10060 Torino, Italy; (D.S.); (E.F.)
| | - Loretta Gammaitoni
- Candiolo Cancer Institute, FPO-IRCCS Candiolo, 10060 Torino, Italy; (M.C.M.); (D.G.); (L.G.)
| | - Erika Fiorino
- Department of Oncology, University of Turin, 10060 Torino, Italy; (D.S.); (E.F.)
| | - Silvia Minei
- Post-Graduate School of Specialization Medical Oncology, University of Bari “A. Moro”, 70120 Bari, Italy;
- Medical Oncology, A.U.O. Consorziale Policlinico di Bari, 70120 Bari, Italy
| | - Paolo Bossi
- Medical Oncology, Department of Medical and Surgical Specialties, Radiological Sciences, Public Health, University of Brescia, 25123 Brescia, Italy;
| | - Lisa Licitra
- Fondazione IRCCS Istituto Nazionale dei Tumori, University of Milan, 20133 Milan, Italy;
| | - Ornella Garrone
- Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (N.D.); (O.G.)
| |
Collapse
|
4
|
Zhu J, Jiang Q. Twist1‑mediated transcriptional activation of Claudin‑4 promotes cervical cancer cell migration and invasion. Oncol Lett 2023; 26:335. [PMID: 37427351 PMCID: PMC10326656 DOI: 10.3892/ol.2023.13921] [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: 02/18/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Claudin-4, a member of the claudin multigene family, participates in events associated with mesenchymal-like activity of cancerous cells. Claudin-4 expression is upregulated in cervical cancer tissue compared with that in adjoining non-neoplastic tissue. However, the mechanisms that regulate Claudin-4 expression in cervical cancer are poorly understood. Moreover, whether Claudin-4 contributes to the migration and invasion of cervical cancer cells remains unclear. By western blotting, reverse transcription-qPCR, bioinformatics analysis, dual-luciferase reporter assay, chromatin immunoprecipitation assay, wound healing assay and Transwell migration/invasion assay, the present study confirmed that Claudin-4 was a downstream target of Twist1, a helix-loop-helix transcriptional factor, the activity of which has a positive correlation with Claudin-4 expression. Mechanistically, Twist1 directly binds to Claudin-4 promoter, resulting in the transactivation of expression. The depletion of the Twist1-binding E-Box1 domain on Claudin-4 promoter via CRISPR-Cas9 knockout system downregulates Claudin-4 expression and suppresses the ability of cervical cancer cells to migrate and invade by elevating E-cadherin levels and lowering N-cadherin levels. Following activation by transforming growth factor-β, Twist1 induces Claudin-4 expression, thus enhancing migration and invasion of cervical cancer cells. In summary, the present data suggested that Claudin-4 was a direct downstream target of Twist1 and served a critical role in promoting Twist1-mediated cervical cancer cell migration and invasion.
Collapse
Affiliation(s)
- Jiaqi Zhu
- Department of Gynecology, Beilun People's Hospital, Beilun Branch of The First Affiliated Hospital, School of Medicine, Zhejiang University, Ningbo, Zhejiang 315826, P.R. China
| | - Qi Jiang
- Department of Obstetrics, Beilun People's Hospital, Beilun Branch of The First Affiliated Hospital, School of Medicine, Zhejiang University, Ningbo, Zhejiang 315826, P.R. China
| |
Collapse
|
5
|
Zanotti LC, Malizia F, Cesatti Laluce N, Avila A, Mamberto M, Anselmino LE, Menacho-Márquez M. Synuclein Proteins in Cancer Development and Progression. Biomolecules 2023; 13:980. [PMID: 37371560 PMCID: PMC10296229 DOI: 10.3390/biom13060980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Synucleins are a family of small, soluble proteins mainly expressed in neural tissue and in certain tumors. Since their discovery, tens of thousands of scientific reports have been published about this family of proteins as they are associated with severe human diseases. Although the physiological function of these proteins is still elusive, their relationship with neurodegeneration and cancer has been clearly described over the years. In this review, we summarize data connecting synucleins and cancer, going from the structural description of these molecules to their involvement in tumor-related processes, and discuss the putative use of these proteins as cancer molecular biomarkers.
Collapse
Affiliation(s)
- Lucía C. Zanotti
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 3100, Argentina
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| | - Florencia Malizia
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 3100, Argentina
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| | - Nahuel Cesatti Laluce
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 3100, Argentina
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| | - Aylén Avila
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| | - Macarena Mamberto
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 3100, Argentina
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| | - Luciano E. Anselmino
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 3100, Argentina
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| | - Mauricio Menacho-Márquez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 3100, Argentina
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 3100, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina
- Centro de Investigación del Cáncer de Rosario, Red de Investigación del Cáncer de Rosario (RICaR), 37007 Salamanca, Spain
| |
Collapse
|
6
|
Wang T, Rho O, Eguiarte-Solomon F, DiGiovanni J. Twist1 as a target for prevention of cutaneous squamous cell carcinoma. Mol Carcinog 2023; 62:62-76. [PMID: 36373194 PMCID: PMC9772054 DOI: 10.1002/mc.23482] [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: 08/17/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
Cutaneous squamous cell carcinoma (cSCC) represents an important clinical problem requiring novel approaches for both prevention and treatment. The transcription factor, Twist-related protein 1 (Twist1), has been identified as having a key mechanistic role in the development and progression of cSCC. Studies in relevant mouse models of cSCC have shown that Twist1 regulates epithelial-mesenchymal transition (EMT) and stemness driving progression and metastasis of cSCC. In addition, further research has shown that Twist1 regulates the balance between keratinocyte proliferation and differentiation and therefore impacts earlier stages of cSCC development. Through use of keratinocyte specific Twist1 knockout models, a role for this gene in keratinocyte stem cell homeostasis has been revealed. As a transcription factor, Twist1 regulates a large number of genes both in a positive, as well as a negative manner across several interdependent pathways. Studies in keratinocyte specific knockout models have shown that Twist1 upregulates the expression of genes involved in proliferation, stemness, and EMT while downregulating the expression of genes associated with differentiation. Furthermore, a number of compounds, including naturally occurring compounds, have been identified that target Twist1 and can block its effects in cancer cells and in keratinocytes in vivo. Collectively, the current understanding of Twist1 function in cSCC development and progression suggests that it represents a potential target for prevention and treatment of cSCC.
Collapse
Affiliation(s)
- Tingzeng Wang
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
| | - Okkyung Rho
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
| | - Fernando Eguiarte-Solomon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78723, United States
- Center for Molecular Carcinogenesis and Toxicology, The University of Texas at Austin, Austin, TX 78723, United States
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78723, United States
| |
Collapse
|
7
|
Zhang G, Wang Z, Song P, Zhan X. DNA and histone modifications as potent diagnostic and therapeutic targets to advance non-small cell lung cancer management from the perspective of 3P medicine. EPMA J 2022; 13:649-669. [PMID: 36505890 PMCID: PMC9727004 DOI: 10.1007/s13167-022-00300-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
Abstract
Lung cancer has a very high mortality in females and males. Most (~ 85%) of lung cancers are non-small cell lung cancers (NSCLC). When lung cancer is diagnosed, most of them have either local or distant metastasis, with a poor prognosis. In order to achieve better outcomes, it is imperative to identify the molecular signature based on genetic and epigenetic variations for different NSCLC subgroups. We hypothesize that DNA and histone modifications play significant roles in the framework of predictive, preventive, and personalized medicine (PPPM; 3P medicine). Epigenetics has a significant impact on tumorigenicity, tumor heterogeneity, and tumor resistance to chemotherapy, targeted therapy, and immunotherapy. An increasing interest is that epigenomic regulation is recognized as a potential treatment option for NSCLC. Most attention has been paid to the epigenetic alteration patterns of DNA and histones. This article aims to review the roles DNA and histone modifications play in tumorigenesis, early detection and diagnosis, and advancements and therapies of NSCLC, and also explore the connection between DNA and histone modifications and PPPM, which may provide an important contribution to improve the prognosis of NSCLC. We found that the success of targeting DNA and histone modifications is limited in the clinic, and how to combine the therapies to improve patient outcomes is necessary in further studies, especially for predictive diagnostics, targeted prevention, and personalization of medical services in the 3P medicine approach. It is concluded that DNA and histone modifications are potent diagnostic and therapeutic targets to advance non-small cell lung cancer management from the perspective of 3P medicine.
Collapse
Affiliation(s)
- Guodong Zhang
- Thoracic Surgery Department, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Shandong 250117 Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| | - Zhengdan Wang
- Thoracic Surgery Department, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Shandong 250117 Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| | - Pingping Song
- Thoracic Surgery Department, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Shandong 250117 Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Thoracic Surgery Department, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Shandong 250117 Jinan, People’s Republic of China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingdao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
8
|
Manni W, Min W. Signaling pathways in the regulation of cancer stem cells and associated targeted therapy. MedComm (Beijing) 2022; 3:e176. [PMID: 36226253 PMCID: PMC9534377 DOI: 10.1002/mco2.176] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/07/2022] Open
Abstract
Cancer stem cells (CSCs) are defined as a subpopulation of malignant tumor cells with selective capacities for tumor initiation, self-renewal, metastasis, and unlimited growth into bulks, which are believed as a major cause of progressive tumor phenotypes, including recurrence, metastasis, and treatment failure. A number of signaling pathways are involved in the maintenance of stem cell properties and survival of CSCs, including well-established intrinsic pathways, such as the Notch, Wnt, and Hedgehog signaling, and extrinsic pathways, such as the vascular microenvironment and tumor-associated immune cells. There is also intricate crosstalk between these signal cascades and other oncogenic pathways. Thus, targeting pathway molecules that regulate CSCs provides a new option for the treatment of therapy-resistant or -refractory tumors. These treatments include small molecule inhibitors, monoclonal antibodies that target key signaling in CSCs, as well as CSC-directed immunotherapies that harness the immune systems to target CSCs. This review aims to provide an overview of the regulating networks and their immune interactions involved in CSC development. We also address the update on the development of CSC-directed therapeutics, with a special focus on those with application approval or under clinical evaluation.
Collapse
Affiliation(s)
- Wang Manni
- Department of Biotherapy, Cancer Center, West China HospitalSichuan UniversityChengduP. R. China
| | - Wu Min
- Department of Biomedical Sciences, School of Medicine and Health SciencesUniversity of North DakotaGrand ForksNorth DakotaUSA
| |
Collapse
|
9
|
El-Helkan B, Emam M, Mohanad M, Fathy S, Zekri AR, Ahmed OS. Long non-coding RNAs as novel prognostic biomarkers for breast cancer in Egyptian women. Sci Rep 2022; 12:19498. [PMID: 36376369 PMCID: PMC9663553 DOI: 10.1038/s41598-022-23938-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer (BC), the most common type of malignant tumor, is the leading cause of death, having the highest incidence rate among women. The lack of early diagnostic tools is one of the clinical obstacles for BC treatment. The current study was designed to evaluate a panel of long non-coding RNAs (lncRNAs) BC040587, HOTAIR, MALAT1, CCAT1, CCAT2, PVT1, UCA1, SPRY4-IT1, PANDAR, and AK058003-and two mRNAs (SNCG, BDNF) as novel prognostic biomarkers for BC. This study was ethically approved by the Institutional Review Board of the National Cancer Institute, Cairo University. Our study included 75 women recently diagnosed with BC and 25 healthy women as normal controls. Patients were divided into three groups: 24 with benign breast diseases, 28 with metastatic breast cancer (MBC, stage IV), and 23 with non-metastatic breast cancer (NMBC, stage III). LncRNA and mRNA expression levels were measured in patient plasma using quantitative real-time PCR. We found that 10 lncRNAs (BCO40587, HOTAIR, PVT1, CCAT2, PANDAR, CCAT1, UCA1, SPRY4-IT1, AK058003, and MALAT1) and both mRNAs demonstrated at least a 2-fold change in expression with a more than 95% probability of significance. BCO40587 and SNCG were significantly up-regulated in MBC and NMBC patients (3.2- and 4-fold, respectively) compared with normal controls. The expression of UCA1 was repressed by 1.78-fold in MBC and NMBC patients compared with those with benign diseases. SPRY4-IT1 was down-regulated by 1.45-fold in MBC patients compared with NMBC and benign disease patients. Up-regulation of lncRNAs plays an important role in BC development. SNCG and BCO40587 may be potential prognostic markers for BC.The organization number is IORG0003381 (IRB No: IRB00004025).
Collapse
Affiliation(s)
- Basma El-Helkan
- grid.7269.a0000 0004 0621 1570Department of Biochemistry, Faculty of Science-Ain Shams University, Cairo, Egypt
| | - Manal Emam
- grid.7269.a0000 0004 0621 1570Department of Biochemistry, Faculty of Science-Ain Shams University, Cairo, Egypt
| | - Marwa Mohanad
- grid.440875.a0000 0004 1765 2064College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October ,Giza, Egypt
| | - Shadia Fathy
- grid.7269.a0000 0004 0621 1570Department of Biochemistry, Faculty of Science-Ain Shams University, Cairo, Egypt
| | - Abdel Rahman Zekri
- grid.7776.10000 0004 0639 9286Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ola S. Ahmed
- grid.7776.10000 0004 0639 9286Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
10
|
Fernández‐Nogales M, López‐Cascales MT, Murcia‐Belmonte V, Escalante A, Fernández‐Albert J, Muñoz‐Viana R, Barco A, Herrera E. Multiomic Analysis of Neurons with Divergent Projection Patterns Identifies Novel Regulators of Axon Pathfinding. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200615. [PMID: 35988153 PMCID: PMC9561852 DOI: 10.1002/advs.202200615] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/30/2022] [Indexed: 06/15/2023]
Abstract
Axon pathfinding is a key step in neural circuits formation. However, the transcriptional mechanisms regulating its progression remain poorly understood. The binary decision of crossing or avoiding the midline taken by some neuronal axons during development represents a robust model to investigate the mechanisms that control the selection of axonal trajectories. Here, to identify novel regulators of axon guidance, this work compares the transcriptome and chromatin occupancy profiles of two neuronal subpopulations, ipsilateral (iRGC) and contralateral retinal ganglion cells (cRGC), with similar functions but divergent axon trajectories. These analyses retrieved a number of genes encoding for proteins not previously implicated in axon pathfinding. In vivo functional experiments confirm the implication of some of these candidates in axonal navigation. Among the candidate genes, γ-synuclein is identified as essential for inducing midline crossing. Footprint and luciferase assays demonstrate that this small-sized protein is regulated by the transcription factor (TF) Pou4f1 in cRGCs. It is also shown that Lhx2/9 are specifically expressed in iRGCs and control a program that partially overlaps with that regulated by Zic2, previously described as essential for iRGC specification. Overall, the analyses identify dozens of new molecules potentially involved in axon guidance and reveal the regulatory logic behind the selection of axonal trajectories.
Collapse
Affiliation(s)
- Marta Fernández‐Nogales
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Maria Teresa López‐Cascales
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Verónica Murcia‐Belmonte
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Augusto Escalante
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Jordi Fernández‐Albert
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Rafael Muñoz‐Viana
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Angel Barco
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| | - Eloísa Herrera
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas ‐Universidad Miguel Hernández de Elche, CSIC‐UMH)San Juan de AlicanteAv. Santiago Ramón y Cajal s/nAlicante03550Spain
| |
Collapse
|
11
|
Zheng L, Guan Z, Xue M. TGF-β Signaling Pathway-Based Model to Predict the Subtype and Prognosis of Head and Neck Squamous Cell Carcinoma. Front Genet 2022; 13:862860. [PMID: 35586572 PMCID: PMC9108263 DOI: 10.3389/fgene.2022.862860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/31/2022] [Indexed: 01/07/2023] Open
Abstract
Background: Although immunotherapy with immune checkpoint therapy has been used to treat head and neck squamous cell carcinoma (HNSCC), response rates and treatment sensitivity remain limited. Recent studies have indicated that transforming growth factor-β (TGF-β) may be an important target for novel cancer immunotherapies. Materials and methods: We collected genomic profile data from The Cancer Genome Atlas and Gene Expression Omnibus. The least absolute shrinkage and selection operator method and Cox regression were used to establish a prognostic model. Gene set enrichment analysis was applied to explore biological functions. Tracking of indels by decomposition and subclass mapping algorithms were adopted to evaluate immunotherapy efficiency. Result: We established a seven TGF-β pathway-associated gene signature with good prediction efficiency. The high-risk score subgroup mainly showed enrichment in tumor-associated signaling such as hypoxia and epithelial-mesenchymal transition (EMT) pathways; This subgroup was also associated with tumor progression. The low-risk score subgroup was more sensitive to immunotherapy and the high-risk score subgroup to cisplatin, erlotinib, paclitaxel, and crizotinib. Conclusion: The TGF-β pathway signature gene model provides a novel perspective for evaluating effectiveness pre-immunotherapy and may guide further studies of precision immuno-oncology.
Collapse
Affiliation(s)
- Lian Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Guan
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhenjie Guan, ; Miaomiao Xue,
| | - Miaomiao Xue
- Department of General Dentistry, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Zhenjie Guan, ; Miaomiao Xue,
| |
Collapse
|
12
|
TGF-β induces GBM mesenchymal transition through upregulation of CLDN4 and nuclear translocation to activate TNF-α/NF-κB signal pathway. Cell Death Dis 2022; 13:339. [PMID: 35418179 PMCID: PMC9008023 DOI: 10.1038/s41419-022-04788-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/06/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
Glioblastoma (GBM) is the most common and aggressive primary malignant brain tumor. The unregulated expression of Claudin-4 (CLDN4) plays an important role in tumor progression. However, the biological role of CLDN4 in GBM is still unknown. This study aimed to determine whether CLDN4 mediates glioma malignant progression, if so, it would further explore the molecular mechanisms of carcinogenesis. Our results revealed that CLDN4 was significantly upregulated in glioma specimens and cells. The inhibition of CLND4 expression could inhibit mesenchymal transformation, cell invasion, cell migration and tumor growth in vitro and in vivo. Moreover, combined with in vitro analysis, we found that CLDN4 can modulate tumor necrosis factor-α (TNF-α) signal pathway. Meanwhile, we also validated that the transforming growth factor-β (TGF-β) signal pathway can upregulate the expression of CLDN4, and promote the invasion ability of GBM cells. Conversely, TGF-β signal pathway inhibitor ITD-1 can downregulate the expression of CLDN4, and inhibit the invasion ability of GBM cells. Furthermore, we found that TGF-β can promote the nuclear translocation of CLDN4. In summary, our findings indicated that the TGF-β/CLDN4/TNF-α/NF-κB signal axis plays a key role in the biological progression of glioma. Disrupting the function of this signal axis may represent a new treatment strategy for patients with GBM.
Collapse
|
13
|
Hoang PH, Landi MT. DNA Methylation in Lung Cancer: Mechanisms and Associations with Histological Subtypes, Molecular Alterations, and Major Epidemiological Factors. Cancers (Basel) 2022; 14:cancers14040961. [PMID: 35205708 PMCID: PMC8870477 DOI: 10.3390/cancers14040961] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/14/2021] [Accepted: 02/11/2022] [Indexed: 01/27/2023] Open
Abstract
Lung cancer is the major leading cause of cancer-related mortality worldwide. Multiple epigenetic factors-in particular, DNA methylation-have been associated with the development of lung cancer. In this review, we summarize the current knowledge on DNA methylation alterations in lung tumorigenesis, as well as their associations with different histological subtypes, common cancer driver gene mutations (e.g., KRAS, EGFR, and TP53), and major epidemiological risk factors (e.g., sex, smoking status, race/ethnicity). Understanding the mechanisms of DNA methylation regulation and their associations with various risk factors can provide further insights into carcinogenesis, and create future avenues for prevention and personalized treatments. In addition, we also highlight outstanding questions regarding DNA methylation in lung cancer to be elucidated in future studies.
Collapse
|
14
|
Zhao Z, Wang Z, Wang P, Liu S, Li Y, Yang X. EPDR1, Which Is Negatively Regulated by miR-429, Suppresses Epithelial Ovarian Cancer Progression via PI3K/AKT Signaling Pathway. Front Oncol 2021; 11:751567. [PMID: 35004274 PMCID: PMC8733570 DOI: 10.3389/fonc.2021.751567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 11/30/2021] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the main pathological type of ovarian cancer. In this study, we found that ependymin-related 1 (EPDR1) was remarkably downregulated in EOC tissues, and low EPDR1 expression was associated with International Federation of Gynecology and Obstetrics (FIGO) stage, metastasis, and poor prognosis. We confirmed that EPDR1 overexpression dramatically suppressed EOC cell proliferation, migration, and invasion in vitro and in vivo. Mechanistically, EPDR1 inhibited EOC tumorigenesis and progression, at least in part, through the repression of the PI3K (Phosphoinositide 3-kinase)/AKT (AKT Serine/Threonine Kinase 1) signaling pathway. Furthermore, the expression and function of EPDR1 were regulated by miR-429, as demonstrated by luciferase reporter assays and rescue experiments. In conclusion, our study validated that EPDR1, negatively regulated by miR-429, played an important role as a tumor-suppressor gene in EOC development via inhibition of the PI3K/AKT pathway. The miR-429/EPDR1 axis might provide novel therapeutic targets for individualized treatment of EOC patients in the future.
Collapse
Affiliation(s)
- Zhendan Zhao
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Zhiling Wang
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Pengling Wang
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Shujie Liu
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, China
- Department of Obstetrics and Gynecology, Zibo Spring Hospital Co., Ltd., Zibo, China
| | - Yingwei Li
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, China
| | - Xingsheng Yang
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
15
|
Xie Y, Shangguan W, Chen Z, Zheng Z, Chen Y, Zhong Q, Zhang Y, Yang J, Zhu D, Xie W. Establishment of Sunitinib-Resistant Xenograft Model of Renal Cell Carcinoma and the Identification of Drug-Resistant Hub Genes and Pathways. Drug Des Devel Ther 2021; 15:5061-5074. [PMID: 34938069 PMCID: PMC8687523 DOI: 10.2147/dddt.s343718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/06/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Yingwei Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Wentai Shangguan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Zhiliang Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Zaosong Zheng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yuqing Chen
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Qiyu Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Yishan Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Jingying Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
| | - Dingjun Zhu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510120, People’s Republic of China
| | - Wenlian Xie
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510120, People’s Republic of China
- Correspondence: Wenlian Xie Email
| |
Collapse
|
16
|
Cheung WW, Hao S, Zheng R, Wang Z, Gonzalez A, Zhou P, Hoffman HM, Mak RH. Targeting interleukin-1 for reversing fat browning and muscle wasting in infantile nephropathic cystinosis. J Cachexia Sarcopenia Muscle 2021; 12:1296-1311. [PMID: 34196133 PMCID: PMC8517356 DOI: 10.1002/jcsm.12744] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/05/2021] [Accepted: 06/08/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Ctns-/- mice, a mouse model of infantile nephropathic cystinosis, exhibit hypermetabolism with adipose tissue browning and profound muscle wasting. Inflammatory cytokines such as interleukin (IL)-1 trigger inflammatory cascades and may be an important cause for cachexia. We employed genetic and pharmacological approaches to investigate the effects of IL-1 blockade in Ctns-/- mice. METHODS We generated Ctns-/- Il1β-/- mice, and we treated Ctns-/- and wild-type control mice with IL-1 receptor antagonist, anakinra (2.5 mg/kg/day, IP) or saline as vehicle for 6 weeks. In each of these mouse lines, we characterized the cachexia phenotype consisting of anorexia, loss of weight, fat mass and lean mass, elevation of metabolic rate, and reduced in vivo muscle function (rotarod activity and grip strength). We quantitated energy homeostasis by measuring the protein content of uncoupling proteins (UCPs) and adenosine triphosphate in adipose tissue and skeletal muscle. We measured skeletal muscle fiber area and intramuscular fatty infiltration. We also studied expression of molecules regulating adipose tissue browning and muscle mass metabolism. Finally, we evaluated the impact of anakinra on the muscle transcriptome in Ctns-/- mice. RESULTS Skeletal muscle expression of IL-1β was significantly elevated in Ctns-/- mice relative to wild-type control mice. Cachexia was completely normalized in Ctns-/- Il1β-/- mice relative to Ctns-/- mice. We showed that anakinra attenuated the cachexia phenotype in Ctns-/- mice. Anakinra normalized UCPs and adenosine triphosphate content of adipose tissue and muscle in Ctns-/- mice. Anakinra attenuated aberrant expression of beige adipose cell biomarkers (UCP-1, CD137, Tmem26, and Tbx1) and molecules implicated in adipocyte tissue browning (Cox2/Pgf2α, Tlr2, Myd88, and Traf6) in inguinal white adipose tissue in Ctns-/- mice. Moreover, anakinra normalized gastrocnemius weight and fiber size and attenuated muscle fat infiltration in Ctns-/- mice. This was accompanied by correction of the increased muscle wasting signalling pathways (increased protein content of ERK1/2, JNK, p38 MAPK, and nuclear factor-κB p65 and mRNA expression of Atrogin-1 and Myostatin) and the decreased myogenesis process (decreased mRNA expression of MyoD and Myogenin) in the gastrocnemius muscle of Ctns-/- mice. Previously, we identified the top 20 differentially expressed skeletal muscle genes in Ctns-/- mice by RNAseq. Aberrant expression of these 20 genes have been implicated in muscle wasting, increased energy expenditure, and lipolysis. We showed that anakinra attenuated 12 of those top 20 differentially expressed muscle genes in Ctns-/- mice. CONCLUSIONS Anakinra may provide a targeted novel therapy for patients with infantile nephropathic cystinosis.
Collapse
Affiliation(s)
- Wai W. Cheung
- Division of Pediatric Nephrology, Department of Pediatrics, Rady Children's Hospital San DiegoUniversity of California, San DiegoLa JollaCAUSA
| | - Sheng Hao
- Department of Nephrology and Rheumatology, Shanghai Children's HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Ronghao Zheng
- Department of Pediatric Nephrology, Rheumatology, and Immunology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhen Wang
- Department of Pediatrics, Shanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Alex Gonzalez
- Division of Pediatric Nephrology, Department of Pediatrics, Rady Children's Hospital San DiegoUniversity of California, San DiegoLa JollaCAUSA
| | - Ping Zhou
- Sichuan Provincial Hospital for Women and ChildrenAffiliated Women and Children's Hospital of Chengdu Medical CollegeChengduChina
| | - Hal M. Hoffman
- Department of PediatricsUniversity of California, San DiegoLa JollaCAUSA
| | - Robert H. Mak
- Division of Pediatric Nephrology, Department of Pediatrics, Rady Children's Hospital San DiegoUniversity of California, San DiegoLa JollaCAUSA
| |
Collapse
|
17
|
Takemura Y, Ojima H, Oshima G, Shinoda M, Hasegawa Y, Kitago M, Yagi H, Abe Y, Hori S, Fujii‐Nishimura Y, Kubota N, Masuda Y, Hibi T, Sakamoto M, Kitagawa Y. Gamma-synuclein is a novel prognostic marker that promotes tumor cell migration in biliary tract carcinoma. Cancer Med 2021; 10:5599-5613. [PMID: 34245137 PMCID: PMC8366101 DOI: 10.1002/cam4.4121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Gamma‐synuclein (SNCG) promotes invasive behavior and is reportedly a prognostic factor in a range of cancers. However, its role in biliary tract carcinoma (BTC) remains unknown. Consequently, we investigated the clinicopathological significance and function of SNCG in BTC. Using resected BTC specimens from 147 patients with adenocarcinoma (extrahepatic cholangiocarcinoma [ECC, n = 96]; intrahepatic cholangiocarcinoma [ICC, n = 51]), we immunohistochemically evaluated SNCG expression and investigated its correlation with clinicopathological factors and outcomes. Furthermore, cell lines with high SNCG expression were selected from 16 BTC cell lines and these underwent cell proliferation and migration assays by siRNAs. In the results, SNCG expression was present in 22 of 96 (22.9%) ECC patients and in 10 of 51 (19.6%) ICC patients. SNCG expression was significantly correlated with poorly differentiated tumor in both ECC and ICC (p = 0.01 and 0.03, respectively) and with perineural invasion and lymph node metastases in ECC (p = 0.04 and 0.003, respectively). Multivariate analyses revealed that SNCG expression was an independent poor prognostic factor in both OS and RFS in both ECC and ICC. In vitro analyses showed high SNCG expression in three BTC cell lines (NCC‐BD1, NCC‐BD3, and NCC‐CC6‐1). Functional analysis revealed that SNCG silencing could suppress cell migration in NCC‐BD1 and NCC‐CC6‐1 and downregulate cell proliferation in NCC‐CC6‐1 significantly. In conclusion, SNCG may promote tumor cell activity and is potentially a novel prognostic marker in BTC.
Collapse
Affiliation(s)
- Yusuke Takemura
- Department of SurgeryKeio University School of MedicineTokyoJapan
- Department of PathologyKeio University School of MedicineTokyoJapan
| | - Hidenori Ojima
- Department of PathologyKeio University School of MedicineTokyoJapan
| | - Go Oshima
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Masahiro Shinoda
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yasushi Hasegawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Minoru Kitago
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Hiroshi Yagi
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yuta Abe
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Shutaro Hori
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Yoko Fujii‐Nishimura
- Department of PathologyKeio University School of MedicineTokyoJapan
- Department of PathologyInternational University of Health and Welfare School of MedicineChibaJapan
| | - Naoto Kubota
- Department of PathologyKeio University School of MedicineTokyoJapan
| | - Yuki Masuda
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Taizo Hibi
- Department of SurgeryKeio University School of MedicineTokyoJapan
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Michiie Sakamoto
- Department of PathologyKeio University School of MedicineTokyoJapan
| | - Yuko Kitagawa
- Department of SurgeryKeio University School of MedicineTokyoJapan
| |
Collapse
|
18
|
Liu C, Mo LH, Feng BS, Jin QR, Li Y, Lin J, Shu Q, Liu ZG, Liu Z, Sun X, Yang PC. Twist1 contributes to developing and sustaining corticosteroid resistance in ulcerative colitis. Am J Cancer Res 2021; 11:7797-7812. [PMID: 34335965 PMCID: PMC8315068 DOI: 10.7150/thno.62256] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/16/2021] [Indexed: 12/27/2022] Open
Abstract
Rationale: Corticosteroid resistance (CR) is a serious drawback to steroid therapy in patients with ulcerative colitis (UC); the underlying mechanism is incompletely understood. Twist1 protein (TW1) is an apoptosis inhibitor and has immune regulatory functions. This study aims to elucidate the roles of TW1 in inducing and sustaining the CR status in UC. Methods: Surgically removed colon tissues of patients with ulcerative colitis (UC) were collected, from which neutrophils were isolated by flow cytometry. The inflammation-related gene activities in neutrophils were analyzed by RNA sequencing. A CR colitis mouse model was developed with the dextran sulfate sodium approach in a hypoxia environment. Results: Higher TW1 gene expression was detected in neutrophils isolated from the colon tissues of UC patients with CR and the CR mouse colon tissues. TW1 physically interacted with glucocorticoid receptor (GR)α in CR neutrophils that prevented GRα from interacting with steroids; which consequently abrogated the effects of steroids on regulating the cellular activities of neutrophils. STAT3 (Signal Transducer and Activator of Transcription-3) interacted with Ras protein activator like 1 to sustain the high TW1 expression in colon mucosal neutrophils of CR patients and CR mice. Inhibition of TW1 restored the sensitivity to corticosteroid of neutrophils in the colon tissues of a CR murine model. Conclusions: UC patients at CR status showed high TW1 expression in neutrophils. TW1 prevented steroids from regulating neutrophil activities. Inhibition of TW1 restored the sensitivity to corticosteroids in the colon tissues at the CR status.
Collapse
|
19
|
Zhang G, Wu P, Zhou K, He M, Zhang X, Qiu C, Li T, Zhang T, Xie K, Dai G, Wang J. Study on the transcriptome for breast muscle of chickens and the function of key gene RAC2 on fibroblasts proliferation. BMC Genomics 2021; 22:157. [PMID: 33676413 PMCID: PMC7937270 DOI: 10.1186/s12864-021-07453-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Growth performance is significant in broiler production. In the growth process of broilers, gene expression varies at different growth stages. However, limited research has been conducted on the molecular mechanisms of muscle growth and development in yellow-feathered male chickens. RESULTS In the study, we used RNA-seq to study the transcriptome of the breast muscle of male Jinghai yellow chickens at 4 (M4F), 8 (M8F) and 12 weeks (M12F) of age. The results showed that 4608 differentially expressed genes (DEGs) were obtained by comparison in pairs of the three groups with Fold Change (FC) ≥ 2 and False Discovery Rate (FDR) ≤ 0.05, and 83, 3445 and 3903 DEGs were obtained separately from M4FvsM8F, M4FvsM12F and M8FvsM12F. Six genes were found as co-differentially expressed in the three age groups, namely SNCG, MYH1A, ARHGDIB, ENSGALG00000031598, ENSGALG00000035660 and ENSGALG00000030559. The GO analysis showed that 0, 304 and 408 biological process (BP) were significantly enriched in M4FvsM8F, M4FvsM12F and M8FvsM12F groups, respectively. KEGG pathway enrichment showed that 1, 2, 4 and 4 pathways were significantly enriched in M4FvsM8F, M4FvsM12F, M8FvsM12F and all DEGs, respectively. They were steroid biosynthesis, carbon metabolism, focal adhesion, cytokine-cytokine receptor interaction, biosynthesis of amino acids and salmonella infection. We constructed short hairpin RNA (shRNA) to interfere the differentially expressed gene RAC2 in DF-1 cells and detected mRNA and protein expression of the downstream genes PAK1 and MAPK8. Results of qPCR showed that RAC2, PAK1 and MAPK8 mRNA expression significantly decreased in the shRAC2-2 group compared with the negative control (NC) group. Western Blot (WB) results showed that the proteins of RAC2, PAK1 and MAPK8 also decreased in the shRAC2-2 group. Cell Counting Kit-8 (CCK-8) and 5-Ethynyl-2'-deoxyuridine (EdU) assay both showed that the proliferation of DF-1 cells was significantly inhibited after transfection of shRAC2-2. CONCLUSIONS The results of RNA-seq revealed genes, BP terms and KEGG pathways related to growth and development of male Jinghai yellow chickens, and they would have important guiding significance to our production practice. Further research suggested that RAC2 might regulate cell proliferation by regulating PAKs/MAPK8 pathway and affect growth of chickens.
Collapse
Affiliation(s)
- Genxi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Pengfei Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China.
| | - Kaizhi Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Mingliang He
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Xinchao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Cong Qiu
- Jiangsu Jinghai Poultry Group Co. Ltd., Nantong, 226100, China
| | - Tingting Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Kaizhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Guojun Dai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| | - Jinyu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou, 225009, China
| |
Collapse
|
20
|
Yang S, Zhang H, Yang H, Zhang J, Wang J, Luo T, Jiang Y, Hua H. SEPHS1 promotes SMAD2/3/4 expression and hepatocellular carcinoma cells invasion. Exp Hematol Oncol 2021; 10:17. [PMID: 33622411 PMCID: PMC7903722 DOI: 10.1186/s40164-021-00212-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the common cancers that are very aggressive. The secreted cytokine transforming growth factor-β (TGF-β) promotes cancer metastasis by multiple mechanisms such as epithelial-mesenchymal transition and immune evasion. The canonical TGF-β signaling is largely mediated by smooth muscle actin/mothers against decapentaplegic (SMAD) proteins. The current study aims to explore the regulation of TGF-β/SMAD signaling by selenophosphate synthetase 1 (SEPHS1). METHODS Immunohistochemistry was used to detect the expression of SEPHS1 in HCC and adjacent liver tissues. Western blotting and quantitative reverse-transcription PCR were used to detect the protein and mRNA levels in HCC cell lines. Cell migration and invasion were determined by transwell assay. Bioinformatic analysis was conducted to determine SEPHS1 expression in HCC and its correlation with the survival of HCC patients. RESULTS Here we report that SEPHS1 is a positive regulator of SMAD proteins. SEPHS1 expression is up-regulated in HCC compared with adjacent liver tissues. SEPHS1 knockdown leads to decreased expression of SMAD2/3/4 and mesenchymal markers including snail, slug and N-cadherin in HCC cells. Furthermore, SEPHS1 knockdown results in a decrease in HCC cells migration and invasion, and suppresses the stimulation of HCC cells migration and invasion by TGF-β. Overexpression of SEPHS1 in HCC cells promotes cell invasion, which can be abrogated by SMAD3 knockdown. Lastly, higher expression of SEPHS1 is correlated with poor prognosis in HCC patients, as manifested by decreased overall survival and disease-free survival. CONCLUSIONS SEPHS1 is a positive regulator of TGF-β/SMAD signaling that is up-regulated in HCC. Increased SEPHS1 expression may indicate poor prognosis for patients with HCC.
Collapse
Affiliation(s)
- Shu Yang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Department of Abdominal Oncology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Hongying Zhang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hua Yang
- Department of Pathology, the Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jin Zhang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ting Luo
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangfu Jiang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
21
|
Wei C, Zhao X, Wang L, Zhang H. TRIP suppresses cell proliferation and invasion in choroidal melanoma via promoting the proteasomal degradation of Twist1. FEBS Lett 2020; 594:3170-3181. [PMID: 32640040 DOI: 10.1002/1873-3468.13882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
Choroidal melanoma (CM) remains the most prevalent form of intraocular malignancy, and the prognosis of affected patients is poor. While the E3 ubiquitin ligase TRAF-interacting protein (TRIP) is known to play key regulatory roles in multiple diseases, its relevance in CM remains uncertain. In the present study, we found that TRIP overexpression is sufficient to inhibit the proliferation, invasion, and epithelial-mesenchymal transition (EMT) of CM cells in vitro, whereas the opposite phenotypes are observed following TRIP knockdown. We further determined that TRIP is able to promote the K48-polyubiquitination of EMT-associated transcription factor Twist-related protein 1, thereby suppressing EMT progression. Together, our results suggest that TRIP plays an important role in regulating the progression of CM and that it may therefore be an important therapeutic target for the treatment of this disease.
Collapse
Affiliation(s)
- Chao Wei
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofei Zhao
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lei Wang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Han Zhang
- Department of Ophthalmology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
22
|
Xiao J, Zhou N, Li Y, Xiao Y, Chen W, Ye J, Ma T, Zhang Y. PEITC inhibits the invasion and migration of colorectal cancer cells by blocking TGF-β-induced EMT. Biomed Pharmacother 2020; 130:110743. [PMID: 34321176 DOI: 10.1016/j.biopha.2020.110743] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/30/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
AIM Tumor metastasis is the leading cause of death in patients with colorectal cancer (CRC), in which epithelial-mesenchymal transition(EMT) plays a vital role. However, the exact mechanisms of this process remain largely unknown. The aim of the present study was to determine the role of phenethyl isothiocyanate (PEITC) in CRC metastasis by regulating EMT. MAIN METHODS Wound healing assays and Transwell matrix assays were used to evaluate the potential of PEITC to inhibit CRC cells invasion and migration in vitro. Western blotting, light microscopy and immunofluorescence assays were used to detect the occurrence of EMT. Luciferase activity assay, real time-PCR and western blotting were used to investigate TGF-β1/Smad signaling activity. KEY FINDINGS We observed that PEITC, an isothiocyanate compound from crucifer with chemopreventive potential, inhibited the invasion and migration of CRC cells. Moreover, we showed that PEITC regulated the EMT of CRC cells. Additionally, we demonstrated that PEITC blocked the activation of the TGF-β1/Smad pathway and significantly suppressed TGF-β1-induced EMT. SIGNIFICANCE Our results suggested that PEITC plays a crucial role in inhibiting the invasion and migration of CRC cells by regulating TGF-β1-induced EMT. The results of the present study provide a theoretical basis for the use of PEITC to treat CRC.
Collapse
Affiliation(s)
- Jian Xiao
- Department of Medicine Oncology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| | - Ningning Zhou
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yin Li
- Department of Endoscopy, Sun Yat-sen University Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yunyun Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wei Chen
- Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Junwen Ye
- Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Tenghui Ma
- Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, Guangzhou, 510655, China
| | - Yan Zhang
- Department of Medicine Oncology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
| |
Collapse
|
23
|
Zhao L, Liu Y, Liu Y, Zhang M, Zhang X. Microfluidic Control of Tumor and Stromal Cell Spheroids Pairing and Merging for Three-Dimensional Metastasis Study. Anal Chem 2020; 92:7638-7645. [PMID: 32374153 DOI: 10.1021/acs.analchem.0c00408] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Three-dimensional cell culture provides an efficient way to simulate the in vivo tumorigenic microenvironment where tumor-stroma interaction intrinsically plays a pivotal role. Conventional three-dimensional (3D) culture is inadequate to address precise coexistential heterogeneous pairing and quantitative measurement in a parallel algorithm format. Herein, we implemented a set of microwell array microfluidic devices to study the cell spheroids-based tumor-stromal metastatic process in vitro. This approach enables accurate one-to-one pairing between tumor and fibroblast spheroid for dissecting 3D tumor invasion in the manner of high-content imaging. On one single device, 240 addressable tumor-stroma pairings can be formed with convenient pipetting and centrifugation within a small area of 1 cm2. Consequential confocal imaging analysis disclosed that the tumor spheroid could envelop the fibroblast spheroid. Specific chemicals can effectively hamper or promote this 3D metastasis. Due to the addressable time-resolved measurements of the merging process of hundreds of doublets, our approach allows us to decipher the metastatic phenotype between different tumor spheroids. Compared with traditional protocols, massive heterogeneous cellular spheroids pairing and merging using this method is well-defined with microfluidic control, which leads to a favorable high-content tumor-stroma doublet metastasis analysis. This simple technique will be a useful tool for investigating heterotypic spheroid-spheroid interactions.
Collapse
Affiliation(s)
- Liang Zhao
- Institute of Precision Medicine and Health, Research Center for Bioengineering and Sensing Technology, Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China, 100083
| | - Yingying Liu
- Institute of Precision Medicine and Health, Research Center for Bioengineering and Sensing Technology, Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China, 100083
| | - Yang Liu
- Institute of Precision Medicine and Health, Research Center for Bioengineering and Sensing Technology, Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China, 100083
| | - Meiqin Zhang
- Institute of Precision Medicine and Health, Research Center for Bioengineering and Sensing Technology, Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China, 100083
| | - Xueji Zhang
- Institute of Precision Medicine and Health, Research Center for Bioengineering and Sensing Technology, Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China, 100083
| |
Collapse
|
24
|
Zhang J, Liu XH, Li C, Wu XX, Chen YL, Li WW, Li X, Gong F, Tang Q, Jiang D. SNCG promotes the progression and metastasis of high-grade serous ovarian cancer via targeting the PI3K/AKT signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:79. [PMID: 32381034 PMCID: PMC7204046 DOI: 10.1186/s13046-020-01589-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
Background The poor prognosis of patients with ovarian cancer is mainly due to cancer progression. γ-Synuclein (SNCG) has reported as a critical player in cancer metastasis. However, its biological roles and mechanism are yet incompletely understood in ovarian cancer, especially in high-grade serous ovarian cancer (HGSOC). Methods This is a retrospective study of 312 patients with ovarian cancer at a single center between 2006 and 2016. Ovarian cancer tissues were stained by immunohistochemistry to analyze the relationship between SNCG expression and clinicopathologic factors. The clinical outcomes versus SNCG expression level were evaluated by Kaplan–Meier method and multiple Cox regression analysis. Next, systematical functional experiments were given to examine the proliferation and metastatic abilities of SNCG both in vitro and in vivo using loss- and gain- of function approaches. Furthermore, the mechanisms of SNCG overexpression were examined by human phospho-kinase array kit and western blot analysis. Results Clinically, the expression of SNCG was significantly upregulated in ovarian cancer compared with the borderline and benign tumor, normal ovary, and fallopian tube. Notably, the high level of SNCG correlated with high-risk clinicopathologic features and showed poor survival for patients with HGSOC, indicating an independent prognostic factor for these patients. Functionally, we observed that overexpression of SNCG promoted cell proliferation, tumor formation, migration, and invasion both in vitro and in vivo. Mechanistically, we identified that SNCG promoted cancer cell metastasis through activating the PI3K/AKT signaling pathway. Conclusions Our results reveal SNCG up-regulation contributes to the poor clinical outcome of patients with HGSOC and highlight the metastasis-promoting function of SNCG via activating the PI3K/Akt signaling pathway in HGSOC.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xiao-Han Liu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cong Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiao-Xing Wu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yan-Lin Chen
- Department of Pathology, Jinshan Hospital, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 401122, China
| | - Wen-Wen Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xian Li
- Department of Pathology, Faculty of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Fan Gong
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qin Tang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Dan Jiang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
25
|
Shen S, Yang C, Liu X, Zheng J, Liu Y, Liu L, Ma J, Ma T, An P, Lin Y, Cai H, Wang D, Li Z, Zhao L, Xue Y. RBFOX1 Regulates the Permeability of the Blood-Tumor Barrier via the LINC00673/MAFF Pathway. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:138-152. [PMID: 32322670 PMCID: PMC7163051 DOI: 10.1016/j.omto.2020.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/25/2020] [Indexed: 12/20/2022]
Abstract
The blood-tumor barrier limits the delivery of therapeutic drugs to brain tumor tissues. Selectively opening the blood-tumor barrier is considered crucial for effective chemotherapy of glioma. RNA-binding proteins have emerged as crucial regulators in various biologic processes. This study found that RNA-binding Fox-1 homolog 1 (RBFOX1) was downregulated in glioma vascular endothelial cells derived from glioma tissues, and in glioma endothelial cells obtained by co-culturing endothelial cells with glioma cells. Overexpression of RBFOX1 impaired the integrity of the blood-tumor barrier and increased its permeability. Additionally, RBFOX1 overexpression decreased the expression of tight junction proteins ZO-1, occludin, and claudin-5. Subsequent analysis of the mechanism indicated that the overexpression of RBFOX1 increased musculoaponeurotic fibrosarcoma protein basic leucine zipper [bZIP] transcription factor F (MAFF) expression by downregulating LINC00673, which stabilized MAFF messenger RNA (mRNA) through Staufen1-mediated mRNA decay. Moreover, MAFF could bind to the promoter region and inhibit the promoter activities of ZO-1, occludin, and claudin-5, which reduced its expression. The combination of RBFOX1 upregulation and LINC00673 downregulation promoted doxorubicin delivery across the blood-tumor barrier, resulting in apoptosis of glioma cells. In conclusion, this study indicated that overexpression of RBFOX1 increased blood-tumor barrier permeability through the LINC00673/MAFF pathway, which might provide a new useful target for future enhancement of blood-tumor barrier permeability.
Collapse
Affiliation(s)
- Shuyuan Shen
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Chunqing Yang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Xiaobai Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Jian Zheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Libo Liu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Jun Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Ping An
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Yang Lin
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| | - Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Di Wang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.,Liaoning Clinical Medical Research Center in Nervous System Disease, Shenyang 110004, People's Republic of China.,Key Laboratory of Neuro-oncology in Liaoning Province, Shenyang 110004, People's Republic of China
| | - Lini Zhao
- Department of Pharmacology, Shenyang Medical College, Shenyang 110034, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Cell Biology, Ministry of Public Health of China, China Medical University, Shenyang 110122, People's Republic of China.,Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, People's Republic of China
| |
Collapse
|
26
|
Hong E, Park S, Ooshima A, Hong CP, Park J, Heo JS, Lee S, An H, Kang JM, Park SH, Park JO, Kim SJ. Inhibition of TGF-β signalling in combination with nal-IRI plus 5-Fluorouracil/Leucovorin suppresses invasion and prolongs survival in pancreatic tumour mouse models. Sci Rep 2020; 10:2935. [PMID: 32076068 PMCID: PMC7031242 DOI: 10.1038/s41598-020-59893-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies. TGF-β is strongly expressed in both the epithelial and stromal compartments of PDAC, and dysregulation of TGF-β signalling is a frequent molecular disturbance in PDAC progression and metastasis. In this study, we investigated whether blockade of TGF-β signalling synergizes with nal-IRI/5-FU/LV, a chemotherapy regimen for malignant pancreatic cancer, in an orthotopic pancreatic tumour mouse model. Compared to nal-IRI/5-FU/LV treatment, combining nal-IRI/5-FU/LV with vactosertib, a TGF-β signalling inhibitor, significantly improved long-term survival rates and effectively suppressed invasion to surrounding tissues. Through RNA-sequencing analysis, we identified that the combination treatment results in robust abrogation of tumour-promoting gene signatures and positive enrichment of tumour-suppressing and apoptotic gene signatures. Particularly, the expression of tumour-suppressing gene Ccdc80 was induced by vactosertib and further induced by vactosertib in combination with nal-IRI/5-FU/LV. Ectopic expression of CCDC80 suppressed migration and colony formation concomitant with decreased expression of epithelial-to-mesenchymal transition (EMT) markers in pancreatic cancer cells. Collectively, these results indicate that combination treatment of vactosertib with nal-IRI/5-FU/LV improves overall survival rates in a mouse model of pancreatic cancer by suppressing invasion through CCDC80. Therefore, combination therapy of nal-IRI/5-FU/LV with vactosertib could provide clinical benefits to pancreatic cancer patients.
Collapse
Affiliation(s)
- Eunji Hong
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea.,Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea
| | - Sujin Park
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea.
| | - Akira Ooshima
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Chang Pyo Hong
- TheragenEtex Bio Institute, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Jinah Park
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Jin Sun Heo
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Siyoung Lee
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Haein An
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Jin Muk Kang
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea
| | - Seok Hee Park
- Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Gyeonggi-do, Republic of Korea
| | - Joon Oh Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seong-Jin Kim
- Precision Medicine Research Center, Advanced Institute of Convergence Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Suwon, Gyeonggi-do, 16229, Republic of Korea.,TheragenEtex Bio Institute, Suwon, Gyeonggi-do, 16229, Republic of Korea.,Medpacto Inc., Seoul, Republic of Korea
| |
Collapse
|
27
|
Wang H, Chen Z, Wang S, Gao X, Qian M, Qiu W, Zhang Z, Zhang S, Qi Y, Sun X, Xue H, Guo X, Zhao R, Li G. TGFβ1-induced beta-site APP-cleaving enzyme 2 upregulation promotes tumorigenesis through the NF-κB signalling pathway in human gliomas. Mol Oncol 2020; 14:407-425. [PMID: 31856384 PMCID: PMC6998390 DOI: 10.1002/1878-0261.12623] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 01/05/2023] Open
Abstract
Gliomas are the most common primary malignant tumours of the central nervous system, and new molecular biomarkers are urgently needed for diagnosis and targeted therapy. Here, we report that increased beta-site APP-cleaving enzyme 2 (BACE2) expression is associated with increases in the grade of human glioma, the incidence of the mesenchymal molecular glioblastoma multiforme subtype and the likelihood of poor prognoses for patients. BACE2 knockdown suppressed cell invasion, cell migration and tumour growth both in vitro and in vivo, while BACE2 overexpression promoted the mesenchymal transition and cell proliferation. Furthermore, TGFβ1 stimulated BACE2 expression through Smad-dependent signalling, which modulated TNF-α-induced NF-κB activity through the PP1A/IKK pathway to promote tumorigenesis in both U87MG and U251 cells. Our study indicated that BACE2 plays a significant role in glioma development. Therefore, BACE2 is a potential therapeutic target for human gliomas due to its function and ability to be regulated.
Collapse
Affiliation(s)
- Huizhi Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Zihang Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Shaobo Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Xiao Gao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Mingyu Qian
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Wei Qiu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Zongpu Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Shouji Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Yanhua Qi
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Xiaopeng Sun
- Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China.,Department of Neurosurgery, Dezhou People's Hospital, Dezhou, Shandong Province, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Xing Guo
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China.,Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, Shandong Province, China.,Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
28
|
Ahmadi A, Najafi M, Farhood B, Mortezaee K. Transforming growth factor-β signaling: Tumorigenesis and targeting for cancer therapy. J Cell Physiol 2018; 234:12173-12187. [PMID: 30537043 DOI: 10.1002/jcp.27955] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 11/19/2018] [Indexed: 02/06/2023]
Abstract
Transforming growth factor (TGF)-β is a multitasking cytokine such that its aberrant expression is related to cancer progression and metastasis. TGF-β is produced by a variety of cells within the tumor microenvironment (TME), and it is responsible for regulation of the activity of cells within this milieu. TGF-β is a main inducer of epithelial-mesenchymal transition (EMT), immune evasion, and metastasis during cancer progression. TGF-β exerts most of its functions by acting on TβRI and TβRII receptors in canonical (Smad-dependent) or noncanonical (Smad-independent) pathways. Members of mitogen-activated protein kinase, phosphatidylinositol 3-kinase/protein kinase B, and nuclear factor κβ are involved in the non-Smad TGF-β pathway. TGF-β acts by complex signaling, and deletion in one of the effectors in this pathway may influence the outcome in a diverse way by taking even an antitumor role. The stage and the type of tumor (contextual cues from cancer cells and/or the TME) and the concentration of TGF-β are other important factors determining the fate of cancer (progression or repression). There are a number of ways for targeting TGF-β signaling in cancer, among them the special focus is on TβRII suppression.
Collapse
Affiliation(s)
- Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
29
|
Li Y, Zhong W, Zhu M, Hu S, Su X. Nodal regulates bladder cancer cell migration and invasion via the ALK/Smad signaling pathway. Onco Targets Ther 2018; 11:6589-6597. [PMID: 30323631 PMCID: PMC6178944 DOI: 10.2147/ott.s177514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Bladder cancer is the most common malignant tumor of the urinary tract. We aimed to explore the biological role and molecular mechanism of Nodal in bladder cancer. Materials and methods The expression of Nodal in bladder cancer tissues and cells was determined by quantitative real-time polymerase chain reaction. The effect of silencing of Nodal on cell proliferation, clone formation, and migration and invasion was evaluated by MTT cell proliferation assay, colony formation, and transwell assays, respectively. Western blot analysis was employed to detect the expression of proliferation- and invasion-related proteins and proteins involved in ALK/Smad signaling. Results We found that the expression of Nodal was significantly increased in bladder cancer tissues and cell lines. Downregulation of Nodal effectively weakened cell proliferation, clone formation, and cell migration and invasion abilities. The protein expression levels of CDC6, E-cadherin, MMP-2, and MMP-9 were also altered by downregulation of Nodal. Knockdown of Nodal also blocked the expression of ALK4, ALK7, Smad2, and Smad4, which are involved in ALK/Smad signaling. Additionally, the ALK4/7 receptor blocker SB431542 reversed the promotive effects of Nodal overexpression on bladder cancer cell proliferation, migration, and invasion. Conclusion Our study indicated that Nodal functions as an oncogene by regulating cell proliferation, migration, and invasion in bladder cancer via the ALK/Smad signaling pathway, thereby providing novel insights into its role in bladder cancer treatment.
Collapse
Affiliation(s)
- Youkong Li
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| | - Wen Zhong
- Department of Endocrine, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China
| | - Min Zhu
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| | - Shengguo Hu
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| | - Xiaokang Su
- Department of Urology, Jingzhou Central Hospital and The Second Clinical Medical College, Yangtze University, Jingzhou 434020, People's Republic of China,
| |
Collapse
|