1
|
Lin J, Zhou J, Xie G, Xie X, Luo Y, Liu J. Retracted article: Functional analysis of ceRNA network of lncRNA TSIX/miR-34a-5p/RBP2 in acute myocardial infarction based on GEO database. Bioengineered 2024; 15:2006865. [PMID: 34784842 PMCID: PMC10841007 DOI: 10.1080/21655979.2021.2006865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022] Open
Abstract
Jiezhong Lin, Jianyi Zhou, Guiting Xie, Xiongwei Xie, Yanfang Luo and Jinguang Liu. Functional analysis of ceRNA network of lncRNA TSIX/miR-34a-5p/RBP2 in acute myocardial infarction based on GEO database. 2021 Oct. doi: 10.1080/21655979.2021.2006865.Since publication, significant concerns have been raised about the compliance with ethical policies for human research and the integrity of the data reported in the article.When approached for an explanation, the authors provided some original data but were not able to provide all the necessary supporting information. As verifying the validity of published work is core to the scholarly record's integrity, we are retracting the article. All authors listed in this publication have been informed.We have been informed in our decision-making by our editorial policies and the COPE guidelines.The retracted article will remain online to maintain the scholarly record, but it will be digitally watermarked on each page as 'Retracted.'
Collapse
Affiliation(s)
- Jiezhong Lin
- Department of Cardiology, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Jianyi Zhou
- Department of Cardiology, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Guiting Xie
- Department of Cardiology, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Xiongwei Xie
- Department of Cardiology, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Yanfang Luo
- Department of Cardiology, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Jinguang Liu
- Department of Cardiology, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| |
Collapse
|
2
|
Andersen GT, Ianevski A, Resell M, Pojskic N, Rabben HL, Geithus S, Kodama Y, Hiroyuki T, Kainov D, Grønbech JE, Hayakawa Y, Wang TC, Zhao CM, Chen D. Multi-bioinformatics revealed potential biomarkers and repurposed drugs for gastric adenocarcinoma-related gastric intestinal metaplasia. NPJ Syst Biol Appl 2024; 10:127. [PMID: 39496635 PMCID: PMC11535201 DOI: 10.1038/s41540-024-00455-0] [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: 06/05/2024] [Accepted: 10/13/2024] [Indexed: 11/06/2024] Open
Abstract
Biomarkers associated with the progression from gastric intestinal metaplasia (GIM) to gastric adenocarcinoma (GA), i.e., GA-related GIM, could provide valuable insights into identifying patients with increased risk for GA. The aim of this study was to utilize multi-bioinformatics to reveal potential biomarkers for the GA-related GIM and predict potential drug repurposing for GA prevention in patients. The multi-bioinformatics included gene expression matrix (GEM) by microarray gene expression (MGE), ScType (a fully automated and ultra-fast cell-type identification based solely on a given scRNA-seq data), Ingenuity Pathway Analysis, PageRank centrality, GO and MSigDB enrichments, Cytoscape, Human Protein Atlas and molecular docking analysis in combination with immunohistochemistry. To identify GA-related GIM, paired surgical biopsies were collected from 16 GIM-GA patients who underwent gastrectomy, yielding 64 samples (4 biopsies per stomach x 16 patients) for MGE. Co-analysis was performed by including scRNAseq and immunohistochemistry datasets of endoscopic biopsies of 37 patients. The results of the present study showed potential biomarkers for GA-related GIM, including GEM of individual patients, individual genes (such as RBP2 and CD44), signaling pathways, network of molecules, and network of signaling pathways with key topological nodes. Accordingly, potential treatment targets with repurposed drugs were identified including epidermal growth factor receptor, proto-oncogene tyrosine-protein kinase Src, paxillin, transcription factor Jun, breast cancer type 1 susceptibility protein, cellular tumor antigen p53, mouse double minute 2, and CD44.
Collapse
Affiliation(s)
- Gøran Troseth Andersen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
- Department of Surgery, Namsos Hospital, Namsos, Norway
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Mathilde Resell
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Naris Pojskic
- Laboratory for Bioinformatics and Biostatistics, University of Sarajevo - Institute for Genetic Engineering and Biotechnology, Sarajevo, Bosnia and Herzegovina
| | - Hanne-Line Rabben
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Synne Geithus
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Yosuke Kodama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tomita Hiroyuki
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jon Erik Grønbech
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Surgery, St. Olav's Hospital, Trondheim, Norway
| | - Yoku Hayakawa
- Department of Gastroenterology, Tokyo University Hospital, Tokyo, Japan
| | - Timothy C Wang
- Department of Digestive and Liver Diseases and Herbert Iring Comprehensive Cancer Center, Columbia University Medical Center, New York, USA
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Duan Chen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
3
|
Nasimi Shad A, Akhlaghipour I, Alshakarchi HI, Saburi E, Moghbeli M. Role of microRNA-363 during tumor progression and invasion. J Physiol Biochem 2024; 80:481-499. [PMID: 38691273 DOI: 10.1007/s13105-024-01022-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/05/2024] [Indexed: 05/03/2024]
Abstract
Recent progresses in diagnostic and therapeutic methods have significantly improved prognosis in cancer patients. However, cancer is still considered as one of the main causes of human deaths in the world. Late diagnosis in advanced tumor stages can reduce the effectiveness of treatment methods and increase mortality rate of cancer patients. Therefore, investigating the molecular mechanisms of tumor progression can help to introduce the early diagnostic markers in these patients. MicroRNA (miRNAs) has an important role in regulation of pathophysiological cellular processes. Due to their high stability in body fluids, they are always used as the non-invasive markers in cancer patients. Since, miR-363 deregulation has been reported in a wide range of cancers, we discussed the role of miR-363 during tumor progression and metastasis. It has been reported that miR-363 has mainly a tumor suppressor function through the regulation of transcription factors, apoptosis, cell cycle, and structural proteins. MiR-363 also affected the tumor progression via regulation of various signaling pathways such as WNT, MAPK, TGF-β, NOTCH, and PI3K/AKT. Therefore, miR-363 can be introduced as a probable therapeutic target as well as a non-invasive diagnostic marker in cancer patients.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hawraa Ibrahim Alshakarchi
- Al-Zahra Center for Medical and Pharmaceutical Research Sciences (ZCMRS), Al-Zahraa University for Women, Karbala, Iraq
| | - Ehsan Saburi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
5
|
The novel histone deacetylase inhibitor pracinostat suppresses the malignant phenotype in human glioma. Mol Biol Rep 2022; 49:7507-7519. [PMID: 35622308 DOI: 10.1007/s11033-022-07559-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/04/2022] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Glioma is the most common malignant brain tumor in adults. The effects of conventional treatment regimens are still limited to prolonging the survival of patients. Histone deacetylases (HDACs) are potential targets for tumor treatment. Pracinostat is a new type of HDAC inhibitor (HDACi) that has a significant antitumor effect on a variety of tumors. Thus, we aim to investigate the role of pracinostat in human glioma and explored its underlying mechanism. METHODS Cell viability, proliferation and apoptosis of human glioma cell lines were measured by Cell Counting kit 8 and flow cytometry. Pathway verification and protein interaction were determined by quantitative real-time polymerase chain reaction, Western blotting and immunofluorescence staining. Transwell technology was used to assess the migration and invasion of cells. Clinical significance of TIMP3, MMP9 and MMP2 in glioma was analyzed through The Cancer Genome Atlas (TCGA) database and the Genotype-Tissue Expression (GTEx) database. RESULTS Functionally, pracinostat not only inhibited proliferation and induced apoptosis but also inhibited migration and invasion in human glioma cell lines. Mechanistically, pracinostat increased the expression of TIMP3 and decreased the expression of MMP2, MMP9 and VEGF in human glioma cells in vitro and in vivo. In addition, pracinostat inhibited both the PI3K/Akt signaling pathway and the STAT3 pathway. CONCLUSIONS Our results strongly support the potential clinical use of pracinostat as a novel therapy for human glioma in the near future.
Collapse
|
6
|
Li Y, Lin M, Wang S, Cao B, Li C, Li G. Novel Angiogenic Regulators and Anti-Angiogenesis Drugs Targeting Angiogenesis Signaling Pathways: Perspectives for Targeting Angiogenesis in Lung Cancer. Front Oncol 2022; 12:842960. [PMID: 35372042 PMCID: PMC8965887 DOI: 10.3389/fonc.2022.842960] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Lung cancer growth is dependent on angiogenesis. In recent years, angiogenesis inhibitors have attracted more and more attention as potential lung cancer treatments. Current anti-angiogenic drugs targeting VEGF or receptor tyrosine kinases mainly inhibit tumor growth by reducing angiogenesis and blocking the energy supply of lung cancer cells. However, these drugs have limited efficiency, raising concerns about limited scope of action and mechanisms of patient resistance to existing drugs. Therefore, current basic research on angiogenic regulators has focused more on screening carcinogenic/anticancer genes, miRNAs, lncRNAs, proteins and other biomolecules capable of regulating the expression of specific targets in angiogenesis signaling pathways. In addition, new uses for existing drugs and new drug delivery systems have received increasing attention. In our article, we analyze the application status and research hotspots of angiogenesis inhibitors in lung cancer treatment as a reference for subsequent mechanistic research and drug development.
Collapse
Affiliation(s)
- Yingying Li
- Pharmacy Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Lin
- Pharmacy Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shiyuan Wang
- Pharmacy Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Cao
- Pharmacy Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyu Li
- Pharmacy Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guohui Li
- Pharmacy Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Sanaei MJ, Razi S, Pourbagheri-Sigaroodi A, Bashash D. The PI3K/Akt/mTOR pathway in lung cancer; oncogenic alterations, therapeutic opportunities, challenges, and a glance at the application of nanoparticles. Transl Oncol 2022; 18:101364. [PMID: 35168143 PMCID: PMC8850794 DOI: 10.1016/j.tranon.2022.101364] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/15/2022] [Accepted: 02/05/2022] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the most common and deadliest human malignancies. The alterations of PI3K/Akt/mTOR pathway are related to lung cancer progression. PI3K axis regulates proliferation, apoptosis, metastasis, and EMT of lung cancer. Agents inhibiting components of PI3K axis diminish lung tumor growth and invasion. Low efficacy and off-target toxicity could be improved by nanoparticle application.
Lung cancer is the leading cause of cancer-related mortality worldwide. Although the PI3K/Akt/mTOR signaling pathway has recently been considered as one of the most altered molecular pathways in this malignancy, few articles reviewed the task. In this review, we aim to summarize the original data obtained from international research laboratories on the oncogenic alterations in each component of the PI3K/Akt/mTOR pathway in lung cancer. This review also responds to questions on how aberrant activation in this axis contributes to uncontrolled growth, drug resistance, sustained angiogenesis, as well as tissue invasion and metastatic spread. Besides, we provide a special focus on pharmacologic inhibitors of the PI3K/Akt/mTOR axis, either as monotherapy or in a combined-modal strategy, in the context of lung cancer. Despite promising outcomes achieved by using these agents, however, the presence of drug resistance as well as treatment-related adverse events is the other side of the coin. The last section allocates a general overview of the challenges associated with the inhibitors of the PI3K pathway in lung cancer patients. Finally, we comment on the future research aspects, especially in which nano-based drug delivery strategies might increase the efficacy of the therapy in this malignancy.
Collapse
|
8
|
Ding Z, Zhang C, Zhang B, Li Q. Unraveling the Proteomic Landscape of Intestinal Epithelial Cell-Derived Exosomes in Mice. Front Physiol 2022; 13:773671. [PMID: 35283765 PMCID: PMC8905357 DOI: 10.3389/fphys.2022.773671] [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: 09/10/2021] [Accepted: 01/14/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose This study aimed to identify the biological functions of small intestine intestinal epithelial cell derived exosomes (IEC-Exos) and further distinguished the difference proteins in IEC-Exos between ileum and jejunum related to function of the digestive system and occurrence of several diseases. Materials and Methods IECs of Male C57BL/6J mice were isolated. IEC-Exos were extracted from jejunum and ileum epithelial cell culture fluid by ultracentrifugation. In addition, isobaric tags for relative and absolute quantitation (iTRAQ) combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) were used to detect IEC-Exo proteins and conduct biological information analysis. Results The results showed that compared with jejunum IEC-Exos from ileum IEC-Exos, there were 393 up-regulated proteins and 346 down-regulated proteins. IECs-Exos, especially derived from jejunum, were rich in angiotensin-converting enzyme 2 (ACE2). The highly expressed proteins from ileum IEC-Exos were mostly enriched in genetic information processing pathways, which mainly mediate the processes of bile acid transport, protein synthesis and processing modification. In contrast, the highly expressed proteins from jejunum IEC-Exos were mainly enriched in metabolic pathways involved in sugar, fatty acid, amino acid, drug, and bone metabolism, etc. The differentially expressed proteins between ileum and jejunum IEC-Exos were not only related to the function of the digestive system but also closely related to the occurrence of infectious diseases, endocrine diseases and osteoarthritis, etc. Conclusion IEC-Exos there were many differentially expressed proteins between ileum and jejunum, which played different roles in regulating intestinal biological functions. ACE2, the main host cell receptor of SARS-CoV-2, was highly expressed in IEC-Exos, which indicated that IEC-Exos may be a potential route of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Zhenyu Ding
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cuiyu Zhang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baokun Zhang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
9
|
Yang D, Ma X, Song P. A prognostic model of non small cell lung cancer based on TCGA and ImmPort databases. Sci Rep 2022; 12:437. [PMID: 35013450 PMCID: PMC8748945 DOI: 10.1038/s41598-021-04268-7] [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: 06/28/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
Bioinformatics methods are used to construct an immune gene prognosis assessment model for patients with non-small cell lung cancer (NSCLC), and to screen biomarkers that affect the occurrence and prognosis of NSCLC. The transcriptomic data and clinicopathological data of NSCLC and cancer-adjacent normal tissues were downloaded from the Cancer Genome Atlas (TCGA) database and the immune-related genes were obtained from the IMMPORT database (http://www.immport.org/); then, the differentially expressed immune genes were screened out. Based on these genes, an immune gene prognosis model was constructed. The Cox proportional hazards regression model was used for univariate and multivariate analyses. Further, the correlations among the risk score, clinicopathological characteristics, tumor microenvironment, and the prognosis of NSCLC were analyzed. A total of 193 differentially expressed immune genes related to NSCLC were screened based on the "wilcox.test" in R language, and Cox single factor analysis showed that 19 differentially expressed immune genes were associated with the prognosis of NSCLC (P < 0.05). After including 19 differentially expressed immune genes with P < 0.05 into the Cox multivariate analysis, an immune gene prognosis model of NSCLC was constructed (it included 13 differentially expressed immune genes). Based on the risk score, the samples were divided into the high-risk and low-risk groups. The Kaplan–Meier survival curve results showed that the 5-year overall survival rate in the high-risk group was 32.4%, and the 5-year overall survival rate in the low-risk group was 53.7%. The receiver operating characteristic model curve confirmed that the prediction model had a certain accuracy (AUC = 0.673). After incorporating multiple variables into the Cox regression analysis, the results showed that the immune gene prognostic risk score was an independent predictor of the prognosis of NSCLC patients. There was a certain correlation between the risk score and degree of neutrophil infiltration in the tumor microenvironment. The NSCLC immune gene prognosis assessment model was constructed based on bioinformatics methods, and it can be used to calculate the prognostic risk score of NSCLC patients. Further, this model is expected to provide help for clinical judgment of the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Dongliang Yang
- Department of General Education, Cangzhou Medical College, Cangzhou, 061001, China
| | - Xiaobin Ma
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 252200, China
| | - Peng Song
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 252200, China.
| |
Collapse
|
10
|
Yang GJ, Wu J, Miao L, Zhu MH, Zhou QJ, Lu XJ, Lu JF, Leung CH, Ma DL, Chen J. Pharmacological inhibition of KDM5A for cancer treatment. Eur J Med Chem 2021; 226:113855. [PMID: 34555614 DOI: 10.1016/j.ejmech.2021.113855] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022]
Abstract
Lysine-specific demethylase 5A (KDM5A, also named RBP2 or JARID1A) is a demethylase that can remove methyl groups from histones H3K4me1/2/3. It is aberrantly expressed in many cancers, where it impedes differentiation and contributes to cancer cell proliferation, cell metastasis and invasiveness, drug resistance, and is associated with poor prognosis. Pharmacological inhibition of KDM5A has been reported to significantly attenuate tumor progression in vitro and in vivo in a range of solid tumors and acute myeloid leukemia. This review will present the structural aspects of KDM5A, its role in carcinogenesis, a comparison of currently available approaches for screening KDM5A inhibitors, a classification of KDM5A inhibitors, and its potential as a drug target in cancer therapy.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, 999078, China
| | - Liang Miao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Ming-Hui Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Qian-Jin Zhou
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Xin-Jiang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, 999078, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, 999077, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
11
|
Shi M, Zhao F, Sun L, Tang F, Gao W, Xie W, Cao X, Zhuang J, Chen X. Bioactive glass activates VEGF paracrine signaling of cardiomyocytes to promote cardiac angiogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112077. [PMID: 33947569 DOI: 10.1016/j.msec.2021.112077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/13/2021] [Accepted: 03/20/2021] [Indexed: 12/28/2022]
Abstract
The heart contains a wide range of cell types, which are not isolated but interact with one another via multifarious paracrine, autocrine and endocrine factors. In terms of cardiac angiogenesis, previous studies have proved that regulating the communication between cardiomyocytes and endothelial cells is efficacious to promote capillary formation. Firstly, this study investigated the effect and underlying mechanism of bioactive glass (BG) acted on vascular endothelial growth factor (VEGF) paracrine signaling in cardiomyocytes. We found that bioactive ions released from BG significantly promoted the VEGF production and secretion of cardiomyocytes. Subsequently, we proved that cardiomyocyte-derived VEGF played an important role in mediating the behavior of endothelial cells. Further research showed that the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/hypoxia-inducible factor 1α (HIF-1α) signaling pathway was upregulated by BG, which was involved in VEGF expression of cardiomyocytes. This study revealed that by means of modulating cellular crosstalk via paracrine signaling of host cells in heart is a new direction for the application of BGs in cardiac angiogenesis.
Collapse
Affiliation(s)
- Miao Shi
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Fujian Zhao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Luyao Sun
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Fengling Tang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Wendong Gao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Weihan Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Xiaodong Cao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China
| | - Jian Zhuang
- School of Medicine, South China University of Technology, Guangzhou 510006, PR China; Guangdong General Hospital, Guangzhou 510080, PR China.
| | - Xiaofeng Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China; School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China; Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, PR China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, PR China; Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
12
|
Yang GJ, Zhu MH, Lu XJ, Liu YJ, Lu JF, Leung CH, Ma DL, Chen J. The emerging role of KDM5A in human cancer. J Hematol Oncol 2021; 14:30. [PMID: 33596982 PMCID: PMC7888121 DOI: 10.1186/s13045-021-01041-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Histone methylation is a key posttranslational modification of chromatin, and its dysregulation affects a wide array of nuclear activities including the maintenance of genome integrity, transcriptional regulation, and epigenetic inheritance. Variations in the pattern of histone methylation influence both physiological and pathological events. Lysine-specific demethylase 5A (KDM5A, also known as JARID1A or RBP2) is a KDM5 Jumonji histone demethylase subfamily member that erases di- and tri-methyl groups from lysine 4 of histone H3. Emerging studies indicate that KDM5A is responsible for driving multiple human diseases, particularly cancers. In this review, we summarize the roles of KDM5A in human cancers, survey the field of KDM5A inhibitors including their anticancer activity and modes of action, and the current challenges and potential opportunities of this field.
Collapse
Affiliation(s)
- Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China.,Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, People's Republic of China
| | - Ming-Hui Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Xin-Jiang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Yan-Jun Liu
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, 210046, People's Republic of China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China.,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China
| | - Chung-Hang Leung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, People's Republic of China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, 999077, People's Republic of China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China. .,Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, People's Republic of China. .,Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China.
| |
Collapse
|
13
|
Metformin Reduces Histone H3K4me3 at the Promoter Regions of Positive Cell Cycle Regulatory Genes in Lung Cancer Cells. Cancers (Basel) 2021; 13:cancers13040739. [PMID: 33578894 PMCID: PMC7916663 DOI: 10.3390/cancers13040739] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary To understand the effect of metformin on epigenetic regulation, we analyzed histone H3 methylation, DNA methylation, and chromatin accessibility in lung cancer cells. Metformin showed little effect on DNA methylation or chromatin accessibility but significantly reduced H3K4me3 levels at the promoters of positive cell cycle regulatory genes. Metformin downregulated H3K4 methyltransferase MLL2 expression and knockdown of MLL2 resulted in suppression of H3K4me3 expression and lung cancer cell proliferation. We further evaluated the clinicopathological significance of MLL2 in tumor and matched normal tissues from 42 non-small cell lung cancer patients. MLL2 overexpression was significantly associated with poor recurrence-free survival in lung adenocarcinoma. Our study facilitates the understanding of the effect of metformin on the regulation of histone H3K4me3 at promoter regions of cell cycle regulatory genes in lung cancer cells, and MLL2 may be a potential therapeutic target for lung cancer therapy. Abstract This study aimed at understanding the effect of metformin on histone H3 methylation, DNA methylation, and chromatin accessibility in lung cancer cells. Metformin significantly reduced H3K4me3 level at the promoters of positive cell cycle regulatory genes such as CCNB2, CDK1, CDK6, and E2F8. Eighty-eight genes involved in cell cycle showed reduced H3K4me3 levels in response to metformin, and 27% of them showed mRNA downregulation. Metformin suppressed the expression of H3K4 methyltransferases MLL1, MLL2, and WDR82. The siRNA-mediated knockdown of MLL2 significantly downregulated global H3K4me3 level and inhibited lung cancer cell proliferation. MLL2 overexpression was found in 14 (33%) of 42 NSCLC patients, and a Cox proportional hazards analysis showed that recurrence-free survival of lung adenocarcinoma patients with MLL2 overexpression was approximately 1.32 (95% CI = 1.08–4.72; p = 0.02) times poorer than in those without it. Metformin showed little effect on DNA methylation and chromatin accessibility at the promoter regions of cell cycle regulatory genes. The present study suggests that metformin reduces H3K4me3 levels at the promoters of positive cell cycle regulatory genes through MLL2 downregulation in lung cancer cells. Additionally, MLL2 may be a potential therapeutic target for reducing the recurrence of lung adenocarcinoma.
Collapse
|
14
|
Ma X, Jin L, Lei X, Tong J, Wang R. MicroRNA‑363‑3p inhibits cell proliferation and induces apoptosis in retinoblastoma cells via the Akt/mTOR signaling pathway by targeting PIK3CA. Oncol Rep 2020; 43:1365-1374. [PMID: 32323827 PMCID: PMC7107813 DOI: 10.3892/or.2020.7544] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/19/2019] [Indexed: 01/03/2023] Open
Abstract
There is extensive evidence suggesting that microRNAs (miRs) can modulate the activity of oncogenes and tumor suppressors, and are associated with the occurrence of cancer. In the present study, the function of miR-363-3p in the progression of retinoblastoma (RB) was investigated. miR-363-3p expression in RB was decreased, and miR-363-3p protein levels were found to be inversely correlated with phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) levels. Overexpression of miR-363-3p in an in vitro model of RB revealed that miR-363-3p had anticancer effects on RB and regulated PIK3CA, pyruvate dehydrogenase kinase 1 (PDK1) and phosphorylated protein kinase B (p-AKT) protein expression. Downregulation of miR-363-3p promoted cell proliferation of RB cells through PIK3CA, PDK1 and p-AKT protein expression. Knockdown of PIK3CA increased the anticancer effects of miR-363-3p in RB cells. Treatment with OSU-03012, a PDK1 inhibitor, accelerated the anticancer effects of miR-363-3p in RB cells. Taken together, the results demonstrate that miR-363-3p functions as a tumor suppressor in RB by targeting PIK3CA.
Collapse
Affiliation(s)
- Xiaojie Ma
- Department of Ophthalmology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710021, P.R. China
| | - Lan Jin
- Department of Ophthalmology, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, Shaanxi 710021, P.R. China
| | - Xiaoqin Lei
- Department of Ophthalmology, Xi'an No. 4 Hospital, Xi'an, Shaanxi 710004, P.R. China
| | - Jingan Tong
- Department of Ophthalmology, The First Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712000, P.R. China
| | - Runsheng Wang
- Department of Ophthalmology, Xi'an No. 4 Hospital, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
15
|
Ruiz CE, Manuguerra S, Curcuraci E, Santulli A, Messina CM. Carbamazepine, cadmium chloride and polybrominated diphenyl ether-47, synergistically modulate the expression of antioxidants and cell cycle biomarkers, in the marine fish cell line SAF-1. MARINE ENVIRONMENTAL RESEARCH 2020; 154:104844. [PMID: 31784109 DOI: 10.1016/j.marenvres.2019.104844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/08/2019] [Accepted: 11/20/2019] [Indexed: 06/10/2023]
Abstract
A wide range of contaminants, industrial by-products, plastics, and pharmaceutics belonging to various categories, have been found in sea water. Although these compounds are detected at concentrations that might be considered as sub-lethal, under certain conditions they could act synergistically producing unexpected effects in term of toxicity or perturbation of biochemical markers leading to standard pathway. In this study, the Sparus aurata fibroblast cell line SAF-1, was exposed to increasing concentrations of carbamazepine (CBZ), polybrominated diphenyl ether 47 (BDE-47) and cadmium chloride (CdCl2) until 72 h, to evaluate the cytotoxicity and the expression of genes related to antioxidant defense, cell cycle and energetic balance. In general, both vitality and gene expression were affected by the exposure to the different toxicants, in terms of antioxidant defense and cell cycle control, showing the most significant effects in cells exposed to the mixture of the three compounds, respect to the single compounds separately. The synergic effect of the compounds on the analyzed biomarkers, underlie the potential negative impact of the contaminants on health of marine organisms.
Collapse
Affiliation(s)
- Cristobal Espinosa Ruiz
- University of Palermo, Dept. of Earth and Sea Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Simona Manuguerra
- University of Palermo, Dept. of Earth and Sea Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Eleonora Curcuraci
- University of Palermo, Dept. of Earth and Sea Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Andrea Santulli
- University of Palermo, Dept. of Earth and Sea Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy; Consorzio Universitario della Provincia di Trapani, Marine Biology Institute, Via Barlotta 4, 91100, Trapani, Italy
| | - Concetta M Messina
- University of Palermo, Dept. of Earth and Sea Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy.
| |
Collapse
|
16
|
Chen Y, Xu Y, Zhu K, Cao Z, Huang Z. Tumor necrosis factor-related apoptosis-inducing ligand modulates angiogenesis and apoptosis to inhibit non-small cell lung carcinoma tumor growth in mice. J Int Med Res 2019; 47:3243-3252. [PMID: 31234689 PMCID: PMC6683941 DOI: 10.1177/0300060519854293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective To investigate the anti-tumor effect and mechanism of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in non-small cell lung carcinoma (NSCLC) in mice. Methods We first established NSCLC animal models using 20 BALB/c nude mice that were randomly divided into two equal groups (n = 10): TRAIL-treated and control untreated groups. We measured expression levels of B cell leukemia/lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), vascular endothelial growth factor (VEGF), and VEGF receptor (VEGFR). We also performed microvessel density, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and immunohistochemical assays to determine the effect of TRAIL on apoptosis and angiogenesis in NSCLC tumors in vitro. Results TRAIL inhibited tumor growth in the NSCLC mouse model, and the TUNEL assay showed that it induced tumor cell apoptosis. Immunohistochemical staining revealed that TRAIL induced Bcl-2 protein downregulation, suggesting that the mitochondrial apoptotic pathway is involved in regulating NSCLC apoptosis. However, TRAIL did not affect Bax protein expression. Immunohistochemical staining also revealed significantly reduced VEGF and VEGFR protein expression in the TRAIL group, indicating that TRAIL limits angiogenesis in NSCLC tumor tissues. Conclusions In conclusion, TRAIL inhibits NSCLC growth both by inducing tumor cell apoptosis and restricting angiogenesis in tumors.
Collapse
Affiliation(s)
- Yuanmei Chen
- 1 Department of Thoracic Surgery, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, P.R. China
| | - Yuanji Xu
- 2 Department of Radiation Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, P.R. China
| | - Kunshou Zhu
- 1 Department of Thoracic Surgery, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, P.R. China
| | - Zhiyun Cao
- 3 Fujian Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, P.R. China
| | - Zhengrong Huang
- 4 Department of Integrative Traditional Chinese and Western Medicine, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian, P.R. China
| |
Collapse
|
17
|
Espinosa Ruiz C, Manuguerra S, Cuesta A, Esteban MA, Santulli A, Messina CM. Sub-lethal doses of polybrominated diphenyl ethers affect some biomarkers involved in energy balance and cell cycle, via oxidative stress in the marine fish cell line SAF-1. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2019; 210:1-10. [PMID: 30797971 DOI: 10.1016/j.aquatox.2019.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are a class of persistent contaminants which are found all over the world in the marine environment. Sparus aurata fibroblast cell line (SAF-1) was exposed to increasing concentrations of PBDEs 47 and 99, until 72 h to evaluate the cytotoxicity, reactive oxygen species (ROS) production and the expression of some selected molecular markers related to cell cycle, cell signaling, energetic balance and oxidative stress (p53, erk-1, hif-1α and nrf-2), by real-time PCR. Furthermore, SAF-1 cells were exposed for 7 and 15 days to sub-lethal concentrations, in order to evaluate the response of some biomarkers by immunoblotting (p53, ERK-1, AMPK, HIF-1α and NRF-2). After 48 and 72 h, the cells showed a significant decrease of cell vitality as well as an increase of intracellular ROS production. Gene expression analysis showed that sub-lethal concentrations of BDE-99 and 47, after 72 h, up-regulated cell cycle and oxidative stress biomarkers, although exposure to 100 μmol L-1 down-regulated the selected markers related to cell cycle, cell signaling, energetic balance. After 7 and 15 days of sub-lethal doses exposure, all the analyzed markers resulted affected by the contaminants. Our results suggest that PBDEs influence the cells homeostasis first of all via oxidative stress, reducing the cell response and defense capacity and affecting its energetic levels. This situation of stress and energy imbalance could represents a condition that, modifying some of the analyzed biochemical pathways, would predispose to cellular transformation.
Collapse
Affiliation(s)
- Cristobal Espinosa Ruiz
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Simona Manuguerra
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy
| | - Alberto Cuesta
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Maria Angeles Esteban
- Fish Innate Immune System Group, Department of Cell Biology and Histology, Faculty of Biology, Campus Regional de Excelencia Internacional "Campus Mare Nostrum", University of Murcia, 30100, Murcia, Spain
| | - Andrea Santulli
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy; Consorzio Universitario della Provincia di Trapani, Marine Biology Institute, Via Barlotta 4, 91100, Trapani, Italy
| | - Concetta M Messina
- University of Palermo, Dept of Earth and Marine Science DISTEM, Laboratory of Marine Biochemistry and Ecotoxicology, Via Barlotta 4, 91100, Trapani, Italy.
| |
Collapse
|
18
|
Manuguerra S, Espinosa Ruiz C, Santulli A, Messina CM. Sub-lethal Doses of Polybrominated Diphenyl Ethers, in Vitro, Promote Oxidative Stress and Modulate Molecular Markers Related to Cell Cycle, Antioxidant Balance and Cellular Energy Management. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16040588. [PMID: 30781636 PMCID: PMC6406823 DOI: 10.3390/ijerph16040588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 01/08/2023]
Abstract
In the present study, we evaluated the effects of different concentrations of the polybrominated diphenyl ethers (PBDEs) BDE-209, BDE-47 and BDE-99, on the vitality and oxidative stress of a HS-68 human cell culture exposed to the compounds for three days. The results showed that for this exposure time, only the highest concentrations produced a significant vitality reduction and oxidative stress induction (p < 0.05), measured as reactive oxygen species (ROS). Subsequently, in order to verify the effects of sub-lethal doses, cells were exposed for a longer time and data collected, after 12 and 20 days, to study ROS production and some molecular markers related to cell cycle and stress (p53, pRB, PARP, c-Jun and c-Fos), antioxidant status and proliferation (ERK, c-Jun and c-Fos), energy balance (NRF2, AMPK, HIF). Most of the biomarkers were influenced by the treatments, indicating that sub-lethal doses of PBDEs, for longer time, can enhance the production of ROS, altering the energetic metabolism, cell cycle and antioxidant balance, determining possible negative effects on the cell proliferation equilibrium.
Collapse
Affiliation(s)
- Simona Manuguerra
- Department of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, University of Palermo, Via Barlotta 4, 91100 Trapani, Italy.
| | - Cristóbal Espinosa Ruiz
- Department of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, University of Palermo, Via Barlotta 4, 91100 Trapani, Italy.
| | - Andrea Santulli
- Department of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, University of Palermo, Via Barlotta 4, 91100 Trapani, Italy.
- Marine Biology Institute, Consorzio Universitario della Provincia di Trapani, Via Barlotta 4, 91100 Trapani, Italy.
| | - Concetta Maria Messina
- Department of Earth and Sea Science, Laboratory of Marine Biochemistry and Ecotoxicology, University of Palermo, Via Barlotta 4, 91100 Trapani, Italy.
| |
Collapse
|
19
|
Hu D, Jablonowski C, Cheng PH, AlTahan A, Li C, Wang Y, Palmer L, Lan C, Sun B, Abu-Zaid A, Fan Y, Brimble M, Gamboa NT, Kumbhar RC, Yanishevski D, Miller KM, Kang G, Zambetti GP, Chen T, Yan Q, Davidoff AM, Yang J. KDM5A Regulates a Translational Program that Controls p53 Protein Expression. iScience 2018; 9:84-100. [PMID: 30388705 PMCID: PMC6214872 DOI: 10.1016/j.isci.2018.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 09/01/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022] Open
Abstract
The p53 tumor suppressor pathway is frequently inactivated in human cancers. However, there are some cancer types without commonly recognized alterations in p53 signaling. Here we report that histone demethylase KDM5A is involved in the regulation of p53 activity. KDM5A is significantly amplified in multiple types of cancers, an event that tends to be mutually exclusive to p53 mutation. We show that KDM5A acts as a negative regulator of p53 signaling through inhibition of p53 translation via suppression of a subgroup of eukaryotic translation initiation genes. Genetic deletion of KDM5A results in upregulation of p53 in multiple lineages of cancer cells and inhibits tumor growth in a p53-dependent manner. In addition, we have identified a regulatory loop between p53, miR-34, and KDM5A, whereby the induction of miR-34 leads to suppression of KDM5A. Thus, our findings reveal a mechanism by which KDM5A inhibits p53 translation to modulate cancer progression.
Collapse
Affiliation(s)
- Dongli Hu
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Carolyn Jablonowski
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Pei-Hsin Cheng
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Alaa AlTahan
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Chunliang Li
- Department of Tumor Cell Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yingdi Wang
- Department of Oncology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Lance Palmer
- Department of Computational Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Cuixia Lan
- Department of Clinical Laboratory, Affiliated Qingdao Hiser Hospital of Qingdao University, Qingdao 266033, China
| | - Bingmei Sun
- Department of Clinical Laboratory, Qingdao Central Hospital, Affiliated Hospital of Qingdao University, Qingdao 266042, China
| | - Ahmed Abu-Zaid
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yiping Fan
- Department of Computational Biology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Mark Brimble
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Nicolas T Gamboa
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Ramhari C Kumbhar
- Department of Molecular Biosciences, University of Texas at Austin, 100 E 24th St NHB 2.606 Stop A5000, Austin, TX 78712, USA
| | - David Yanishevski
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Kyle M Miller
- Department of Molecular Biosciences, University of Texas at Austin, 100 E 24th St NHB 2.606 Stop A5000, Austin, TX 78712, USA
| | - Guolian Kang
- Department of Biostatistics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Gerard P Zambetti
- Department of Pathology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Qin Yan
- Department of Pathology, Yale School of Medicine, 310 Cedar St, New Haven, CT 06520, USA
| | - Andrew M Davidoff
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Jun Yang
- Department of Surgery, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
20
|
Yang P, Xiong J, Zuo L, Liu K, Zhang H. miR‑140‑5p regulates cell migration and invasion of non‑small cell lung cancer cells through targeting VEGFA. Mol Med Rep 2018; 18:2866-2872. [PMID: 30015904 PMCID: PMC6102653 DOI: 10.3892/mmr.2018.9291] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/22/2018] [Indexed: 01/18/2023] Open
Abstract
Lung cancer is the most common type of cancer worldwide, the most prevalent form of which is non-small cell lung cancer (NSCLC). MicroRNAs (miRs) are involved in the progression of NSCLC; however, the specific function of miR-140-5p in NSCLC remains unclear. The present study demonstrated that miR-140-5p was downregulated in the tumor tissues of patients with NSCLC, and it was associated with a poor prognosis. Furthermore, miR-140-5p significantly suppressed cell migration and invasion of the NSCLC cell line A549. In addition, the direct regulatory effect of miR-140-5p on vascular endothelial growth factor-A (VEGFA) was predicted by TargetScan and verified using a luciferase reporter gene assay. The present study also hypothesized that miR-140-5p may inhibit the expression of phosphorylated-protein kinase B by targeting VEGFA. In conclusion, miR-140-5p may be a potential target for the development of anti-neoplastic therapies in lung cancer.
Collapse
Affiliation(s)
- Peixia Yang
- Respiratory Department of Internal Medicine, Linyi Central Hospital, Linyi, Shandong 276000, P.R. China
| | - Jie Xiong
- Respiratory Department of Internal Medicine, Linyi Central Hospital, Linyi, Shandong 276000, P.R. China
| | - Lin Zuo
- Department of Internal Medicine, Health Service Center of Lanshan District, Linyi, Shandong 276000, P.R. China
| | - Kequn Liu
- Department of Hemodialysis, People's Hospital of Yishui County, Linyi, Shandong 276000, P.R. China
| | - Houbin Zhang
- Department of Thoracic Surgery, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
21
|
Chao WH, Lai MY, Pan HT, Shiu HW, Chen MM, Chao HM. Dendrobium nobile Lindley and its bibenzyl component moscatilin are able to protect retinal cells from ischemia/hypoxia by dowregulating placental growth factor and upregulating Norrie disease protein. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:193. [PMID: 29933759 PMCID: PMC6013934 DOI: 10.1186/s12906-018-2256-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/13/2018] [Indexed: 02/04/2023]
Abstract
Background Presumably, progression of developmental retinal vascular disorders is mainly driven by persistent ischemia/hypoxia. An investigation into vision-threatening retinal ischemia remains important. Our aim was to evaluate, in relation to retinal ischemia, protective effects and mechanisms of Dendrobium nobile Lindley (DNL) and its bibenzyl component moscatilin. The therapeutic mechanisms included evaluations of levels of placental growth factor (PLGF) and Norrie disease protein (NDP). Methods An oxygen glucose deprivation (OGD) model involved cells cultured in DMEM containing 1% O2, 94% N2 and 0 g/L glucose. High intraocular pressure (HIOP)-induced retinal ischemia was created by increasing IOP to 120 mmHg for 60 min in Wistar rats. The methods included electroretinogram (ERG), histopathology, MTT assay and biochemistry. Results When compared with cells cultured in DMEM containing DMSO (DMSO+DMEM), cells subjected to OGD and pre-administrated with DMSO (DMSO+OGD) showed a significant reduction in the cell viability and NDP expression. Moreover, cells that received OGD and 1 h pre-administration of 0.1 μM moscatilin (Pre-OGD Mos 0.1 μM) showed a significant counteraction of the OGD-induced decreased cell viability. Furthermore, compared with the DMSO+OGD group (44.54 ± 3.15%), there was significant elevated NDP levels in the Pre-OGD Mos 0.1 μM group (108.38 ± 29.33%). Additionally, there were significant ischemic alterations, namely reduced ERG b-wave, less numerous retinal ganglion cells, decreased inner retinal thickness, and reduced/enhanced amacrine’s ChAT/Müller’s GFAP or vimentin immunolabelings. Moreover, there were significantly increased protein levels of HIF-1α, VEGF, PKM2, RBP2 and, particularly, PLGF (pg/ml; Sham vs. Vehicle: 15.11 ± 1.58 vs. 39.53 ± 5.25). These ischemic effects were significantly altered when 1.0 g/Kg/day DNL (DNL1.0 + I/R or I/R+ DNL1.0) was applied before and/or after ischemia, but not vehicle (Vehicle+I/R). Of novelty and significance, the DNL1.0 action mechanism appears to be similar to that of the anti-PLGF Eylea [PLGF (pg/ml); DNL1.0 vs. Eylea+I/R: 19.93 ± 2.24 vs. 6.44 ± 0.60]. Conclusions DNL and moscatilin are able to protect against retinal ischemic/hypoxic changes respectively by downregulating PLGF and upregulating NDP. Progression of developmental retinal vascular disorders such as Norrie disease due to persistent ischemia/hypoxia might be thus prevented. Electronic supplementary material The online version of this article (10.1186/s12906-018-2256-z) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Zhang YL, Qiao SK, Wang RY, Guo XN. NGAL attenuates renal ischemia/reperfusion injury through autophagy activation and apoptosis inhibition in rats. Chem Biol Interact 2018; 289:40-46. [DOI: 10.1016/j.cbi.2018.04.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 01/10/2023]
|
23
|
Tan SQ, Geng X, Liu JH, Pan WHT, Wang LX, Liu HK, Hu L, Chao HM. Xue-fu-Zhu-Yu decoction protects rats against retinal ischemia by downregulation of HIF-1α and VEGF via inhibition of RBP2 and PKM2. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:365. [PMID: 28709426 PMCID: PMC5513111 DOI: 10.1186/s12906-017-1857-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 06/23/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Retinal ischemia-related eye diseases result in visual dysfunction. This study investigates the protective effects and mechanisms of Xue-Fu-Zhu-Yu decoction (XFZYD) with respect to retinal ischemia. METHODS Retinal ischemia (I) was induced in Wistar rats by a high intraocular pressure (HIOP) of 120 mmHg for 1 h, which was followed by reperfusion of the ischemic eye; the fellow untreated eye acted as a control. Electroretinogram (ERG), biochemistry and histopathology investigations were performed. RESULTS Significant ischemic changes occurred after ischemia including decreased ERG b-wave ratios, less numerous retinal ganglion cells (RGCs), reduced inner retinal thickness, fewer choline acetyltransferase (ChAT) labeled amacrine cell bodies, increased glial fibrillary acidic protein (GFAP) immunoreactivity and increased vimentin Müller immunolabeling. These were accompanied by significant increases in the mRNA/protein concentrations of vascular endothelium growth factor, hypoxia-inducible factor-1α, pyruvate kinase M2 and retinoblastoma-binding protein 2. The ischemic changes were concentration-dependently and significantly altered when XFZYD was given for seven consecutive days before or after retina ischemia, compared to vehicle. These alterations included enhanced ERG b-wave amplitudes, more numerous RGCs, enhanced inner retinal thickness, a greater number of ChAT immunolabeled amacrine cell bodies and decreased GFAP/vimentin immunoreactivity. Furthermore, decreased mRNA levels of VEGF, HIF-1α, PKM2, and RBP2 were also found. Reduced protein concentrations of VEGF, HIF-1α, PKM2, and RBP2 were also demonstrated. Furthermore, there was an inhibition of the ischemia-associated increased ratios (target protein/β-actin) in the protein levels of VEGF, HIF-1α, PKM2, and RBP2, which were induced by Shikonin, JIB-04 or Avastin. CONCLUSION XFZYD would seem to protect against well-known retinal ischemic changes via a synergistic inhibition of RBP2 and PKM2, as well as down-regulation of HIF-1α and a reduction in VEGF secretion.
Collapse
Affiliation(s)
- Shu-Qiu Tan
- Department of Ophthalmology, Affiliated Hospital of Taishan Medical University, Taishan, Shandong China
| | - Xue Geng
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong China
| | - Jorn-Hon Liu
- Department of Ophthalmology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Wynn Hwai-Tzong Pan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Li-Xiang Wang
- Department of Pharmacology, School of Medicine, Shandong University, Jinan, Shandong China
| | - Hui-Kang Liu
- Division of Basic Chinese Medicine, National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Lei Hu
- Department of Ophthalmology, Affiliated Hospital of Taishan Medical University, Taishan, Shandong China
| | - Hsiao-Ming Chao
- Department of Ophthalmology, Cheng Hsin General Hospital, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Chinese Medicine, School of Chinese Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Wang ZY, Yang J, Liu CK, Shen SQ. High Expression of Retinoblastoma-Binding Protein 2 (RBP2) in Patients with Hepatocellular Carcinoma and Its Prognostic Significance. Med Sci Monit 2017; 23:2736-2744. [PMID: 28582381 PMCID: PMC5469320 DOI: 10.12659/msm.905262] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Recently, some studies have found that retinoblastoma-binding protein 2 (RBP2) is involved in the development and progression of many kinds of malignant tumors. This study aimed to explore the expression level of RBP2 in hepatocellular carcinoma (HCC) and its prognostic significance. Material/Methods Immunohistochemical analysis was used to evaluate the RBP2 expression level in 130 HCC patients and adjacent normal tissues. Tumor angiogenesis was marked by CD31 and vascular endothelial growth factor (VEGF) staining. Kaplan-Meier and Cox regression analyses were performed to examine the relationship between RBP2 expression and prognosis of HCC patients. Results RBP2 expression was significantly higher in HCC tissues (positive expression rate: 72.3%, 94/130). Increased RBP2 expression was dramatically associated with AFP level (P=0.016), degree of differentiation (P=0.000), and TNM stage (P=0.035). Moreover, tumors with RBP2-positive expression showed significantly higher intratumoral MVD than those with RBP2-negative expression (P=0.000). Kaplan-Meier analysis revealed RBP2-positive expression was related to decreased disease-free survival (DFS) (P=0.000) and overall survival (OS) (P=0.000). Furthermore, RBP2 was an independent poor prognostic factor of DFS and OS (P=0.029 and 0.010, respectively) as demonstrated by multivariate analysis. Conclusions Increased RBP2 expression, as an independent poor prognostic factor for DFS and OS of HCC patients, is closely related to tumor angiogenesis. RBP2 is expected to become a new potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhen-Yu Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Jie Yang
- Department of Emergency Surgery, The Second People's Hospital of Wuhu, Wuhu, Anhui, China (mainland)
| | - Chang-Kuo Liu
- Department of General Surgery, Second People's Hospital of Wuhu, Wuhu, Anhui, China (mainland)
| | - Shi-Qiang Shen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
25
|
Maggi EC, Crabtree JS. Novel targets in the treatment of neuroendocrine tumors: RBP2. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2017. [DOI: 10.2217/ije-2016-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Retinoblastoma binding protein 2, also known as RBP2, JARID1A or KDM5A, is an H3K4 demethylase implicated in a variety of non-neuroendocrine, and more recently, neuroendocrine tumors (NETs). NETs are tumors that form from neuroendocrine cells in tissues of the GI tract, endocrine pancreas, lung, skin and other tissues. RBP2 is expressed at abnormally high levels in NETs and recent work demonstrates that modulation of RBP2 in vitro and in vivo impacts end points of tumorigenesis. Interestingly, the demethylase activity of RBP2 is not exclusively responsible for these changes, as RBP2's binding partners may mediate its activity in a tissue- or context-dependent manner. Here, we discuss the features of RBP2 and its role in cell cycle regulation, angiogenesis and drug resistance in cancer.
Collapse
Affiliation(s)
- Elaine C Maggi
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Judy S Crabtree
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
26
|
He YH, Li MF, Zhang XY, Meng XM, Huang C, Li J. NLRC5 promotes cell proliferation via regulating the AKT/VEGF-A signaling pathway in hepatocellular carcinoma. Toxicology 2016; 359-360:47-57. [DOI: 10.1016/j.tox.2016.06.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/16/2016] [Accepted: 06/17/2016] [Indexed: 12/22/2022]
|
27
|
Liu X, Zhu M, Yang X, Wang Y, Qin B, Cui C, Chen H, Sang A. Inhibition of RACK1 ameliorates choroidal neovascularization formation in vitro and in vivo. Exp Mol Pathol 2016; 100:451-9. [PMID: 27112838 DOI: 10.1016/j.yexmp.2016.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/18/2016] [Accepted: 04/20/2016] [Indexed: 10/21/2022]
Abstract
Choroidal neovascularization (CNV) occurs as a result of age-related macular degeneration (AMD) and causes severe vision loss among elderly patients. The receptor for activated C-kinase 1 (RACK1) serves as a scaffold protein which is recently found to promote angiogenesis. However, the impact of RACK1 on the vascular endothelial growth factor (VEGF) expression in endothelial cells and subsequent choroidal angiogenesis formation remains to be elucidated. In this study, we found that RACK1 and VEGF expression increased, and reached the peak at 7d in mouse CNV model by laser application. Furthermore, on RPE/choroid cryosections, RACK1 co-localized with CD31, suggesting that RACK1 was expressed in endothelial cells. In vitro, RF/6A cell hypoxia model showed that RACK1 expression was up-regulated in parallel with hypoxia-induced factor 1 (HIF-1α) and VEGF expression, reaching the peak at 6h. Silencing of RACK1 suppressed the invasion and tube formation activity of RF/6A cells in ARPE-19 and RF/6A co-culture system, possibly through VEGF signal pathway. Overexpression of RACK1 showed the opposite effect. Intravitreal injection of anti-RACK1 monoclonal antibody predominantly decreased RACK1 and VEGF expression in mouse laser-induced CNV model. Meanwhile, anti-RACK1 monoclonal antibody intravitreal injection also decreased incidence of CNV and leakage area. These data indicated that RACK1 promoted CNV formation via VEGF pathway. Additionally, anti-RACK1 monoclonal antibody significantly decreased CNV in mouse model and may have therapeutic potential in human CNV.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Manhui Zhu
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaowei Yang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Ying Wang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Bai Qin
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Chen Cui
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China
| | - Hui Chen
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China.
| | - Aimin Sang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China; Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
28
|
Li D, Wei Y, Wang D, Gao H, Liu K. MicroRNA-26b suppresses the metastasis of non-small cell lung cancer by targeting MIEN1 via NF-κB/MMP-9/VEGF pathways. Biochem Biophys Res Commun 2016; 472:465-70. [DOI: 10.1016/j.bbrc.2016.01.163] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 01/25/2016] [Indexed: 12/15/2022]
|
29
|
Wang X, Zhou M, Fu Y, Sun T, Chen J, Qin X, Yu Y, Jia J, Chen C. RBP2 Promotes Adult Acute Lymphoblastic Leukemia by Upregulating BCL2. PLoS One 2016; 11:e0152142. [PMID: 27008505 PMCID: PMC4805198 DOI: 10.1371/journal.pone.0152142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/09/2016] [Indexed: 12/23/2022] Open
Abstract
Despite recent increases in the cure rate of acute lymphoblastic leukemia (ALL), adult ALL remains a high-risk disease that exhibits a high relapse rate. In this study, we found that the histone demethylase retinoblastoma binding protein-2 (RBP2) was overexpressed in both on-going and relapse cases of adult ALL, which revealed that RBP2 overexpression was not only involved in the pathogenesis of ALL but that its overexpression might also be related to relapse of the disease. RBP2 knockdown induced apoptosis and attenuated leukemic cell viability. Our results demonstrated that BCL2 is a novel target of RBP2 and supported the notion of RBP2 being a regulator of BCL2 expression via directly binding to its promoter. As the role of RBP2 in regulating apoptosis was confirmed, RBP2 overexpression and activation of BCL2 might play important roles in ALL development and progression.
Collapse
Affiliation(s)
- Xiaoming Wang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| | - Minran Zhou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| | - Yue Fu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| | - Ting Sun
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| | - Jin Chen
- Anhui Medical College, Hefei, Anhui, P. R. China
| | - Xuemei Qin
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| | - Yuan Yu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| | - Jihui Jia
- Department of Microbiology/Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, Shandong, P. R. China
| | - Chunyan Chen
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong, P. R. China
| |
Collapse
|
30
|
Zhang YL, Zhang J, Cui LY, Yang S. Autophagy activation attenuates renal ischemia-reperfusion injury in rats. Exp Biol Med (Maywood) 2015; 240:1590-8. [PMID: 25898836 DOI: 10.1177/1535370215581306] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/24/2015] [Indexed: 11/16/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury is a leading cause of acute kidney injury (AKI), which is a common clinical complication but lacks effective therapies. This study investigated the role of autophagy in renal I/R injury and explored potential mechanisms in an established rat renal I/R injury model. Forty male Wistar rats were randomly divided into four groups: Sham, I/R, I/R pretreated with 3-methyladenine (3-MA, autophagy inhibitor), or I/R pretreated with rapamycin (autophagy activator). All rats were subjected to clamping of the left renal pedicle for 45 min after right nephrectomy, followed by 24 h of reperfusion. The Sham group underwent the surgical procedure without ischemia. 3-MA and rapamycin were injected 15 min before ischemia. Renal function was indicated by blood urea nitrogen and serum creatinine. Tissue samples from the kidneys were scored histopathologically. Autophagy was indicated by light chain 3 (LC3), Beclin-1, and p62 levels and the number of autophagic vacuoles. Apoptosis was evaluated by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) method and expression of caspase-3. Autophagy was activated after renal I/R injury. Inhibition of autophagy by 3-MA before I/R aggravated renal injury, with worsened renal function, higher renal tissue injury scores, and more tubular apoptosis. In contrast, rapamycin pretreatment ameliorated renal injury, with improved renal function, lower renal tissue injury scores, and inhibited apoptosis based on fewer TUNEL-positive cells and lower caspase-3 expression. Our results demonstrate that autophagy could be activated during I/R injury and play a protective role in renal I/R injury. The mechanisms were involved in the regulation of several autophagy and apoptosis-related genes. Furthermore, autophagy activator may be a promising therapy for I/R injury and AKI in the future.
Collapse
Affiliation(s)
- Ya-Li Zhang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Jie Zhang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Li-Yan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Shuo Yang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
31
|
Liu K, Liu Y, Lau JL, Min J. Epigenetic targets and drug discovery Part 2: Histone demethylation and DNA methylation. Pharmacol Ther 2015; 151:121-40. [PMID: 25857453 DOI: 10.1016/j.pharmthera.2015.04.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/31/2015] [Indexed: 02/06/2023]
Abstract
Chromatin structure is dynamically modulated by various chromatin modifications, such as histone/DNA methylation and demethylation. We have reviewed histone methyltransferases and methyllysine binders in terms of small molecule screening and drug discovery in the first part of this review series. In this part, we will summarize recent progress in chemical probe and drug discovery of histone demethylases and DNA methyltransferases. Histone demethylation and DNA methylation have attracted a lot of attention regarding their biology and disease implications. Correspondingly, many small molecule compounds have been designed to modulate the activity of histone demethylases and DNA methyltransferases, and some of them have been developed into therapeutic drugs or put into clinical trials.
Collapse
Affiliation(s)
- Ke Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Central China Normal University, Wuhan 430079, PR China; Structural Genomics Consortium, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Yanli Liu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Central China Normal University, Wuhan 430079, PR China; Structural Genomics Consortium, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Johnathan L Lau
- Structural Genomics Consortium, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jinrong Min
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, College of Life Science, Central China Normal University, Wuhan 430079, PR China; Structural Genomics Consortium, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
32
|
Geng J, Li X, Zhou Z, Wu CL, Bai X, Dai M. EZH2 promotes tumor progression via regulating VEGF-A/AKT signaling in non-small cell lung cancer. Cancer Lett 2015; 359:275-87. [DOI: 10.1016/j.canlet.2015.01.031] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/31/2022]
|
33
|
Shi X, Liang W, Yang W, Xia R, Song Y. Decorin is responsible for progression of non-small-cell lung cancer by promoting cell proliferation and metastasis. Tumour Biol 2014; 36:3345-54. [PMID: 25524578 DOI: 10.1007/s13277-014-2968-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/09/2014] [Indexed: 12/30/2022] Open
Abstract
Decorin, a member of the small leucine-rich proteoglycans family, exists and plays multifunctional roles in stromal and epithelial cells. Emerging evidences showed that decorin is dysregulated expression in a wide variety of human tumors and affects a broad biology process of cancer cells, including growth, metastasis, and angiogenesis. Recent studies demonstrated that decorin could affect A549 proliferation though decreasing TGF-β1, cycling D1 expression and increasing P53 and P21 expression. However, limited data are available on the effect of decorin on metastasis of non-small-cell lung cancer (NSCLC) cell lines and how decorin impacts metastasis is still unknown. In this study, we identified decorin mRNA expression through Oncomine database and verified the expression of decorin mRNA and protein in 50 patients who underwent primary surgical resection of a NSCLC in the Department of Thoracic Surgery, Jinling Hospital, Nanjing University School of Medicine, China, between September 2013 and March 2014 by quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and Western blot. Also, the correlationship between decorin and the NSCLC patients' clinical characteristics or survival ( www.kmplot.com ) was analyzed. Via ectopic expression analyses and Western blot, the roles of decorin in proliferation, metastasis, and the underline mechanism for decorin expression were further explored. We found that decorin was downregulated in NSCLC tissues compared with the adjacent normal lung tissues or normal tissues. Additionally, the expression of decorin was correlated with tumor size, lymph node metastasis, tumor stage, and prognosis. We also showed that overexpression of decorin could inhibit NSCLC cell lines proliferation and metastasis. Through Western blot analysis, we identified that E-cadherin and vascular endothelial growth factor (VEGF) are two key factors responsible for the growth arrest and metastasis inhibition induced by decorin in NSCLC. Our results indicated that decorin plays crucial roles in NSCLC against carcinogenesis and progression. Decorin might be a predictive factor and an attractive therapeutic target for NSCLC patients.
Collapse
Affiliation(s)
- Xuefei Shi
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China,
| | | | | | | | | |
Collapse
|