1
|
Arakawa H, Komatsu S, Kiuchi J, Imamura T, Nishibeppu K, Kamiya H, Takashima Y, Ishida R, Hamada S, Yamauchi M, Ohashi T, Shimizu H, Arita T, Konishi H, Shiozaki A, Kubota T, Fujiwara H, Tsuda H, Otsuji E. Overexpression of SHANK2 contributes malignant outcomes as a Hippo pathway regulator in gastric cancer. Am J Cancer Res 2025; 15:363-374. [PMID: 39949934 PMCID: PMC11815361 DOI: 10.62347/nzmo2658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/03/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Recent studies identified that SH3 and multiple ankyrin repeat domains 2 (SHANK2) is located in a gene-amplified region 11q13 of various human cancers, and has oncogenic functions as a Hippo pathway suppressor in hepatocellular carcinoma. In this study, we tested whether SHANK2 acts as a cancer-promoting gene through its activation or overexpression in gastric cancer (GC). MATERIALS AND METHODS We analyzed 5 GC cell lines and 172 primary tumor samples of GC, which were curatively resected in our hospital. RESULTS Overexpression of SHANK2 protein was frequently detected in GC cell lines (4/5 cell lines, 80%). Knockdown of SHANK2 inhibited cell proliferation, migration and invasion of GC cells in a TP53 mutation-independent manner, and induced the overexpression of the Hippo pathway genes. Fluorescent immunostaining showed that overexpression of SHANK2 in cytoplasm was inversely correlated with Yes1-associated transcriptional regulator (YAP) expression, suggesting that SHANK2 may play a role in suppressing the Hippo pathway in GC. In primary GC samples, both overexpression of SHANK2 in cytoplasm and low expression of SHANK2 in nucleus, which are defined as high SHANK2 index, correlated with more aggressive venous invasion, advanced tumor and nodal stage. Patients with high SHANK2 index tumors had a lower overall survival rate than those with non-expressing tumors. Multivariate analysis demonstrated that high SHANK2 index was independently associated with poor outcomes. CONCLUSIONS These findings suggest that SHANK2 plays a crucial role in tumor malignant potential through the Hippo pathway suppression and highlight its usefulness as a prognostic factor and potential therapeutic target in GC.
Collapse
Affiliation(s)
- Hiroshi Arakawa
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Hajime Kamiya
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Yusuke Takashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Ryo Ishida
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Satoshi Hamada
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Masateru Yamauchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Defense Medical College HospitalSaitama, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKyoto, Japan
| |
Collapse
|
2
|
Cecchini K, Ajaykumar N, Bagci A, Zamore PD, Gainetdinov I. Mouse Pachytene piRNAs Cleave Hundreds of Transcripts, But Alter the Steady-State Abundance of Only a Minority of Targets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.02.621675. [PMID: 39554027 PMCID: PMC11566022 DOI: 10.1101/2024.11.02.621675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
In animals, 18-35-nt piRNAs guide PIWI proteins to regulate complementary RNAs. During male meiosis, mammals produce an exceptionally abundant class of piRNAs called pachytene piRNAs. Pachytene piRNAs are required for spermatogenesis and have been proposed to control gene expression by various mechanisms. Here, we show that pachytene piRNAs regulate targets predominantly, if not exclusively, by endonucleolytic cleavage. Remarkably, pachytene piRNAs slice hundreds of RNAs, yet a change in steady-state level is detectable for a small fraction of transcripts. Our data suggest that cleavage of the few targets whose abundance is reduced significantly by piRNAs is essential for male fertility. Other pachytene piRNA targets are enriched for highly transcribed genes, which may explain why piRNA cleavage is often inconsequential for the steady-state abundance of targets. We propose that the retention of pachytene piRNAs throughout mammalian evolution is driven by the selective advantage conferred by a tiny minority of piRNAs.
Collapse
Affiliation(s)
- Katharine Cecchini
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | | - Ayca Bagci
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Phillip D Zamore
- RNA Therapeutics Institute and Howard Hughes Medical Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|
3
|
Liu X, Xie B, Wang S, Wu Y, Zhang Y, Ruan L. Alteration of RNA m6A methylation mediates aberrant RNA binding protein expression and alternative splicing in condyloma acuminatum. PeerJ 2024; 12:e17376. [PMID: 38784389 PMCID: PMC11114121 DOI: 10.7717/peerj.17376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Background Condyloma acuminatum (CA) is caused by low-risk human papillomavirus, and is characterized by high recurrence after treatment. The RNA modification N6-methyladenosine (m6A) plays an important role during diverse viral infections, including high-risk HPV infection in cervical cancer. However, it is unclear whether low-risk HPV infection changes the RNA m6A methylation in CA. Methods High-throughputm6A-sequencing was performed to profile the transcriptome-wide mRNA modifications of CA tissues infected by LR-HPVs and the paired normal tissues from CA patients. We further investigated the regulation of alternative splicing by RNA binding proteins (RBPs) with altered m6A modification and constructed a regulatory network among these RBPs, regulated alternative splicing events (RASEs) and regulated alternative splicing genes (RASGs) in CA. Results The results show that the m6A level in CA tissues differed from that in the paired controls. Furthermore, cell cycle- and cell adhesion- associated genes with m6A modification were differentially expressed in CA tissues compared to the paired controls. In particular, seven RNA binding protein genes with specific m6A methylated sites, showed a higher or lower expression at the mRNA level in CA tissues than in the paired normal tissues. In addition, these differentially expressed RNA binding protein genes would regulate the alternative splicing pattern of apoptotic process genes in CA tissue. Conclusions Our study reveals a sophisticated m6A modification profile in CA tissue that affects the response of host cells to HPV infection, and provides cues for the further exploration of the roles of m6A and the development of a novel treatment strategy for CA.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Dermatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Xie
- Department of Urology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Su Wang
- Department of Dermatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yinhua Wu
- Department of Dermatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Dermatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Liming Ruan
- Department of Dermatology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Lughmani H, Patel H, Chakravarti R. Structural Features and Physiological Associations of Human 14-3-3ζ Pseudogenes. Genes (Basel) 2024; 15:399. [PMID: 38674334 PMCID: PMC11049341 DOI: 10.3390/genes15040399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
There are about 14,000 pseudogenes that are mutated or truncated sequences resembling functional parent genes. About two-thirds of pseudogenes are processed, while others are duplicated. Although initially thought dead, emerging studies indicate they have functional and regulatory roles. We study 14-3-3ζ, an adaptor protein that regulates cytokine signaling and inflammatory diseases, including rheumatoid arthritis, cancer, and neurological disorders. To understand how 14-3-3ζ (gene symbol YWHAZ) performs diverse functions, we examined the human genome and identified nine YWHAZ pseudogenes spread across many chromosomes. Unlike the 32 kb exon-to-exon sequence in YWHAZ, all pseudogenes are much shorter and lack introns. Out of six, four YWHAZ exons are highly conserved, but the untranslated region (UTR) shows significant diversity. The putative amino acid sequence of pseudogenes is 78-97% homologous, resulting in striking structural similarities with the parent protein. The OMIM and Decipher database searches revealed chromosomal loci containing pseudogenes are associated with human diseases that overlap with the parent gene. To the best of our knowledge, this is the first report on pseudogenes of the 14-3-3 family protein and their implications for human health. This bioinformatics-based study introduces a new insight into the complexity of 14-3-3ζ's functions in biology.
Collapse
Affiliation(s)
| | | | - Ritu Chakravarti
- Department of Physiology and Pharmacology, The University of Toledo, Toledo, OH 43614, USA; (H.L.); (H.P.)
| |
Collapse
|
5
|
Dolatshahi M, Bahrami AR, Sheikh QI, Ghanbari M, Matin MM. Gastric cancer and mesenchymal stem cell-derived exosomes: from pro-tumorigenic effects to anti-cancer vehicles. Arch Pharm Res 2024; 47:1-19. [PMID: 38151649 DOI: 10.1007/s12272-023-01477-8] [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: 06/27/2023] [Accepted: 12/15/2023] [Indexed: 12/29/2023]
Abstract
Gastric cancer (GC) is one of the most prevalent malignancies in the world, with a high mortality rate in both women and men. Conventional treatments, like chemotherapy, radiotherapy and surgery, are facing some drawbacks like acquired drug resistance and various side effects, leading to cancer recurrence and increased morbidity; thus, development of novel approaches in targeted therapy would be very beneficial. Exosomes, extracellular vesicles with a size distribution of sub-150 nm, interplay in physiological and pathophysiological cell-cell communications and can pave the way for targeted cancer therapy. Accumulating pieces of evidence have indicated that exosomes derived from mesenchymal stem cells (MSC-EXs) can act as a double-edged sword in some cancers. The purpose of this review is to assess the differences between stem cell therapy and exosome therapy. Moreover, our aim is to demonstrate how naïve MSCs transform into GC-MSCs in the tumor microenvironment. Additionally, the tumorigenic and anti-proliferation effects of MSC-EXs derived from different origins were investigated. Finally, we suggest potential modifications and combination options that involve utilizing MSC-EXs from the foreskin and umbilical cord as promising sources to enhance the efficacy of gastric cancer treatment. This approach is presented in contrast to bone marrow cells, which are more heterogeneous, age-related, and are also easily affected by the patient's circulation system.
Collapse
Affiliation(s)
- Maryam Dolatshahi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Qaiser Iftikhar Sheikh
- School of Biosciences, Western Bank, Firth Court, University of Sheffield, Sheffield, S10 2TN, England, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
6
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [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: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
7
|
Shang J, Nie X, Qi Y, Zhou J, Qi Y. Short-term smoking cessation leads to a universal decrease in whole blood genomic DNA methylation in patients with a smoking history. World J Surg Oncol 2023; 21:227. [PMID: 37496025 PMCID: PMC10369823 DOI: 10.1186/s12957-023-03099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND Epigenetics is involved in various human diseases. Smoking is one of the most common environmental factors causing epigenetic changes. The DNA methylation changes and mechanisms after quitting smoking have yet to be defined. The present study examined the changes in DNA methylation levels before and after short-term smoking cessation and explored the potential mechanism. METHODS Whole blood and clinical data were collected from 8 patients before and after short-term smoking cessation, DNA methylation was assessed, and differentially methylated sites were analyzed, followed by a comprehensive analysis of the differentially methylated sites with clinical data. GO/KEGG enrichment and protein-protein interaction (PPI) network analyses identified the hub genes. The differentially methylated sites between former and current smokers in GSE50660 from the GEO database were detected by GEO2R. Then, a Venn analysis was carried out using the differentially methylated sites. GO/KEGG enrichment analysis was performed on the genes corresponding to the common DNA methylation sites, the PPI network was constructed, and hub genes were predicted. The enriched genes associated with the cell cycle were selected, and the pan-cancer gene expression and clinical significance in lung cancer were analyzed based on the TCGA database. RESULTS Most genes showed decreased DNA methylation levels after short-term smoking cessation; 694 upregulated methylation CpG sites and 3184 downregulated methylation CpG sites were identified. The DNA methylation levels were altered according to the clinical data (body weight, expiratory, and tobacco dependence score). Enrichment analysis, construction of the PPI network, and pan-cancer analysis suggested that smoking cessation may affect various biological processes. CONCLUSIONS Smoking cessation leads to epigenetic changes, mainly decreased in the decline of most DNA methylation levels. Bioinformatics further identified the biologically relevant changes after short-term smoking cessation.
Collapse
Affiliation(s)
- Junyi Shang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University; People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Xinran Nie
- Academy of Medical Science, Zhengzhou University, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Yanan Qi
- Department of Respiratory and Critical Care Medicine, Central China Fuwai Hospital; Central China Fuwai Hospital of Zhengzhou University; People's Hospital of Zhengzhou University; Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Jing Zhou
- Department of Health Management, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, 450003, China
| | - Yong Qi
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University; People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
8
|
Cao F, Jiang Y, Chang L, Du H, Chang D, Pan C, Huang X, Yu D, Zhang M, Fan Y, Bian X, Li K. High-throughput functional screen identifies YWHAZ as a key regulator of pancreatic cancer metastasis. Cell Death Dis 2023; 14:431. [PMID: 37452033 PMCID: PMC10349114 DOI: 10.1038/s41419-023-05951-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Pancreatic cancer is a leading cause of cancer death due to its early metastasis and limited response to the current therapies. Metastasis is a complicated multistep process, which is determined by complex genetic alterations. Despite the identification of many metastasis-related genes, distinguishing the drivers from numerous passengers and establishing the causality in cancer pathophysiology remains challenging. Here, we established a high-throughput and piggyBac transposon-based genetic screening platform, which enables either reduced or increased expression of chromosomal genes near the incorporation site of the gene search vector cassette that contains a doxycycline-regulated promoter. Using this strategy, we identified YWHAZ as a key regulator of pancreatic cancer metastasis. We demonstrated that functional activation of Ywhaz by the gene search vector led to enhanced metastatic capability in mouse pancreatic cancer cells. The metastasis-promoting role of YWHAZ was further validated in human pancreatic cancer cells. Overexpression of YWHAZ resulted in more aggressive metastatic phenotypes in vitro and a shorter survival rate in vivo by modulating epithelial-to-mesenchymal transition. Hence, our study established a high-throughput screening method to investigate the functional relevance of novel genes and validated YWHAZ as a key regulator of pancreatic cancer metastasis.
Collapse
Affiliation(s)
- Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yunpeng Jiang
- Department of Biochemistry and Biophysics, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lin Chang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Endoscopy Center, Peking University Cancer Hospital & Institute, Beijing, China
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China
| | - Hongzhen Du
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China
| | - De Chang
- Department of Pulmonary and Critical Care Medicine, 7th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chunxiao Pan
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China
| | - Xiaozheng Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Donglin Yu
- Department of Biochemistry and Biophysics, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Mi Zhang
- Department of Pulmonary and Critical Care Medicine, 7th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yongna Fan
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China
| | - Xiaocui Bian
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China.
| | - Kailong Li
- Department of Biochemistry and Biophysics, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
9
|
Evidence and Metabolic Implications for a New Non-Canonical Role of Cu-Zn Superoxide Dismutase. Int J Mol Sci 2023; 24:ijms24043230. [PMID: 36834640 PMCID: PMC9966940 DOI: 10.3390/ijms24043230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Copper-zinc superoxide dismutase 1 (SOD1) has long been recognized as a major redox enzyme in scavenging superoxide radicals. However, there is little information on its non-canonical role and metabolic implications. Using a protein complementation assay (PCA) and pull-down assay, we revealed novel protein-protein interactions (PPIs) between SOD1 and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ) or epsilon (YWHAE) in this research. Through site-directed mutagenesis of SOD1, we studied the binding conditions of the two PPIs. Forming the SOD1 and YWHAE or YWHAZ protein complex enhanced enzyme activity of purified SOD1 in vitro by 40% (p < 0.05) and protein stability of over-expressed intracellular YWHAE (18%, p < 0.01) and YWHAZ (14%, p < 0.05). Functionally, these PPIs were associated with lipolysis, cell growth, and cell survival in HEK293T or HepG2 cells. In conclusion, our findings reveal two new PPIs between SOD1 and YWHAE or YWHAZ and their structural dependences, responses to redox status, mutual impacts on the enzyme function and protein degradation, and metabolic implications. Overall, our finding revealed a new unorthodox role of SOD1 and will provide novel perspectives and insights for diagnosing and treating diseases related to the protein.
Collapse
|
10
|
Pu Z, Zhao Q, Chen J, Xie Y, Mou L, Zha X. Single-cell RNA analysis to identify five cytokines signaling in immune-related genes for melanoma survival prognosis. Front Immunol 2023; 14:1148130. [PMID: 37026000 PMCID: PMC10070796 DOI: 10.3389/fimmu.2023.1148130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Melanoma is one of the deadliest skin cancers. Recently, developed single-cell sequencing has revealed fresh insights into melanoma. Cytokine signaling in the immune system is crucial for tumor development in melanoma. To evaluate melanoma patient diagnosis and treatment, the prediction value of cytokine signaling in immune-related genes (CSIRGs) is needed. In this study, the machine learning method of least absolute selection and shrinkage operator (LASSO) regression was used to establish a CSIRG prognostic signature of melanoma at the single-cell level. We discovered a 5-CSIRG signature that was substantially related to the overall survival of melanoma patients. We also constructed a nomogram that combined CSIRGs and clinical features. Overall survival of melanoma patients can be consistently predicted with good performance as well as accuracy by both the 5-CSIRG signature and nomograms. We compared the melanoma patients in the CSIRG high- and low-risk groups in terms of tumor mutation burden, infiltration of the immune system, and gene enrichment. High CSIRG-risk patients had a lower tumor mutational burden than low CSIRG-risk patients. The CSIRG high-risk patients had a higher infiltration of monocytes. Signaling pathways including oxidative phosphorylation, DNA replication, and aminoacyl tRNA biosynthesis were enriched in the high-risk group. For the first time, we constructed and validated a machine-learning model by single-cell RNA-sequencing datasets that have the potential to be a novel treatment target and might serve as a prognostic biomarker panel for melanoma. The 5-CSIRG signature may assist in predicting melanoma patient prognosis, biological characteristics, and appropriate therapy.
Collapse
Affiliation(s)
- Zuhui Pu
- Imaging Department, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Qing Zhao
- Department of Dermatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Dermatology, Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Jiaqun Chen
- Department of Dermatology, Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Yubin Xie
- Department of Dermatology, Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Lisha Mou
- Imaging Department, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- MetaLife Center, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
- *Correspondence: Lisha Mou, ; Xushan Zha,
| | - Xushan Zha
- Department of Dermatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- *Correspondence: Lisha Mou, ; Xushan Zha,
| |
Collapse
|
11
|
Feng S, Xu Z, Zhang Z, Mo Y, Deng Y, Li L, Fei S, Wu J, Wang K, Zhang Q, Song J, Zhou R. RNA-Seq approach to investigate the effects of melatonin on bone marrow-derived dendritic cells from dextran sodium sulfate-induced colitis mice. Toxicology 2022; 481:153354. [DOI: 10.1016/j.tox.2022.153354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/30/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022]
|
12
|
Interactions between 14-3-3 Proteins and Actin Cytoskeleton and Its Regulation by microRNAs and Long Non-Coding RNAs in Cancer. ENDOCRINES 2022. [DOI: 10.3390/endocrines3040057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
14-3-3s are a family of structurally similar proteins that bind to phosphoserine or phosphothreonine residues, forming the central signaling hub that coordinates or integrates various cellular functions, thereby controlling many pathways important in cancer, cell motility, cell death, cytoskeletal remodeling, neuro-degenerative disorders and many more. Their targets are present in all cellular compartments, and when they bind to proteins they alter their subcellular localization, stability, and molecular interactions with other proteins. Changes in environmental conditions that result in altered homeostasis trigger the interaction between 14-3-3 and other proteins to retrieve or rescue homeostasis. In circumstances where these regulatory proteins are dysregulated, it leads to pathological conditions. Therefore, deeper understanding is needed on how 14-3-3 proteins bind, and how these proteins are regulated or modified. This will help to detect disease in early stages or design inhibitors to block certain pathways. Recently, more research has been devoted to identifying the role of MicroRNAs, and long non-coding RNAs, which play an important role in regulating gene expression. Although there are many reviews on the role of 14-3-3 proteins in cancer, they do not provide a holistic view of the changes in the cell, which is the focus of this review. The unique feature of the review is that it not only focuses on how the 14-3-3 subunits associate and dissociate with their binding and regulatory proteins, but also includes the role of micro-RNAs and long non-coding RNAs and how they regulate 14-3-3 isoforms. The highlight of the review is that it focuses on the role of 14-3-3, actin, actin binding proteins and Rho GTPases in cancer, and how this complex is important for cell migration and invasion. Finally, the reader is provided with super-resolution high-clarity images of each subunit of the 14-3-3 protein family, further depicting their distribution in HeLa cells to illustrate their interactions in a cancer cell.
Collapse
|
13
|
Su L, Liu G, Guo Y, Zhang X, Zhu X, Wang J. Integration of Protein-Protein Interaction Networks and Gene Expression Profiles Helps Detect Pancreatic Adenocarcinoma Candidate Genes. Front Genet 2022; 13:854661. [PMID: 35711911 PMCID: PMC9197464 DOI: 10.3389/fgene.2022.854661] [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: 01/24/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
More and more cancer-associated genes (CAGs) are being identified with the development of biological mechanism research. Integrative analysis of protein-protein interaction (PPI) networks and co-expression patterns of these genes can help identify new disease-associated genes and clarify their importance in specific diseases. This study proposed a PPI network and co-expression integration analysis model (PRNet) to integrate PPI networks and gene co-expression patterns to identify potential risk causative genes for pancreatic adenocarcinoma (PAAD). We scored the importance of the candidate genes by constructing a high-confidence co-expression-based edge-weighted PPI network, extracting protein regulatory sub-networks by random walk algorithm, constructing disease-specific networks based on known CAGs, and scoring the genes of the sub-networks with the PageRank algorithm. The results showed that our screened top-ranked genes were more critical in tumours relative to the known CAGs list and significantly differentiated the overall survival of PAAD patients. These results suggest that the PRNet method of ranking cancer-associated genes can identify new disease-associated genes and is more informative than the original CAGs list, which can help investigators to screen potential biomarkers for validation and molecular mechanism exploration.
Collapse
Affiliation(s)
- Lili Su
- College of Electronics and Information Engineering, School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Guang Liu
- College of Electronics and Information Engineering, School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ying Guo
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xuanping Zhang
- College of Electronics and Information Engineering, School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Zhu
- College of Electronics and Information Engineering, School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jiayin Wang
- College of Electronics and Information Engineering, School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Mogal MR, Junayed A, Mahmod MR, Sompa SA, Lima SA, Kar N, TasminaTarin, Khatun M, Zubair MA, Sikder MA. A Computational Approach to Justifying Stratifin as a Candidate Diagnostic and Prognostic Biomarker for Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1617989. [PMID: 35547358 PMCID: PMC9085308 DOI: 10.1155/2022/1617989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer (PC) is considered a silent killer because it does not show specific symptoms at an early stage. Thus, identifying suitable biomarkers is important to avoid the burden of PC. Stratifin (SFN) encodes the 14-3-3σ protein, which is expressed in a tissue-dependent manner and plays a vital role in cell cycle regulation. Thus, SFN could be a promising therapeutic target for several types of cancer. This study was aimed at investigating, using online bioinformatics tools, whether SFN could be used as a diagnostic and prognostic biomarker in PC. SFN expression was explored by utilizing the ONCOMINE, UALCAN, GEPIA2, and GENT2 tools, which revealed that SFN expression is higher in PC than in normal tissues. The clinicopathological analysis using the ULCAN tool showed that the intensity of SFN expression is commensurate with cancer progression. GEPIA2, R2, and OncoLnc revealed a negative correlation between SFN expression and survival probability in PC patients. The ONCOMINE, UCSC Xena, and GEPIA2 tools showed that cofilin 1 is strongly coexpressed with SFN. Moreover, enrichment and network analyses of SFN were performed using the Enrichr and NetworkAnalyst platforms, respectively. Receiver operating characteristic (ROC) curves revealed that tissue-dependent expression of the SFN gene could serve as a diagnostic and prognostic biomarker. However, further wet laboratory studies are necessary to determine the relevance of SFN expression as a biomarker.
Collapse
Affiliation(s)
- Md Roman Mogal
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Asadullah Junayed
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Rashel Mahmod
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Sagarika Adhikary Sompa
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Suzana Afrin Lima
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Newton Kar
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - TasminaTarin
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Marina Khatun
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Abu Zubair
- Department of Food Technology and Nutritional Science, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Asaduzzaman Sikder
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| |
Collapse
|
15
|
Takashima Y, Komatsu S, Ohashi T, Kiuchi J, Kamiya H, Shimizu H, Arita T, Konishi H, Shiozaki A, Kubota T, Okamoto K, Fujiwara H, Tsuda H, Otsuji E. Overexpression of Tetraspanin31 contributes to malignant potential and poor outcomes in gastric cancer. Cancer Sci 2022; 113:1984-1998. [PMID: 35307915 PMCID: PMC9207375 DOI: 10.1111/cas.15342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 12/24/2022] Open
Abstract
Tetraspanin has important functions in many cancers by aggregating with various proteins that interact with intracellular signaling proteins. The molecular function of Tetraspanin31 (TSPAN31), located in the 12q14 amplified region in various cancers, remains unclear in gastric cancer (GC). We tested whether TSPAN31 acts as a cancer‐promoting gene through its activation or overexpression in GC. We analyzed seven GC cell lines and 189 primary tumors, which were curatively resected in our hospital between 2011 and 2013. Overexpression of the TSPAN31 protein was frequently detected in three GC cell lines (42.9%) and 62 primary GC specimens (32.8%). Overexpression of TSPAN31 was significantly correlated with lymphatic invasion, venous invasion, more advanced pT and pN stages, and a higher recurrence rate. Moreover, TSPAN31 positivity was an independent factor predicting worse patient outcomes (p = 0.0283, hazard ratio 3.97). Ectopic overexpression of TSPAN31 facilitated cell proliferation of GC cells, and knockdown of TSPAN31 inhibited cell proliferation, migration, invasion, and epithelial–mesenchymal transition of GC cells through the PI3K‐Akt pathway and increased cell apoptosis in a TP53 mutation‐independent manner. In vivo analysis also revealed knockdown of TSPAN31 suppressed tumor progression. In addition, knockdown of TSPAN31 improved chemosensitivity to cisplatin through the suppression of ABCC2. These findings suggest that TSPAN31 plays a crucial role in tumor‐malignant potential through overexpression, highlighting its utility as a prognostic factor and a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Yusuke Takashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hajime Kamiya
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan.,Department of Basic Pathology, National Defense Medical College, Tokorozawa, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
16
|
Liu Z, Chen L, Gao X, Zou R, Meng Q, Fu Q, Xie Y, Miao Q, Chen L, Tang X, Zhang S, Zhang H, Schroyen M. Quantitative proteomics reveals tissue-specific toxic mechanisms for acute hydrogen sulfide-induced injury of diverse organs in pig. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150365. [PMID: 34555611 DOI: 10.1016/j.scitotenv.2021.150365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
Hydrogen sulfide (H2S) is a highly toxic gas in many environmental and occupational places. It can induce multiple organ injuries particularly in lung, trachea and liver, but the relevant mechanisms remain poorly understood. In this study, we used a TMT-based discovery proteomics to identify key proteins and correlated molecular pathways involved in the pathogenesis of acute H2S-induced toxicity in porcine lung, trachea and liver tissues. Pigs were subjected to acute inhalation exposure of up to 250 ppm of H2S for 5 h for the first time. Changes in hematology and biochemical indexes, serum inflammatory cytokines and histopathology demonstrated that acute H2S exposure induced organs inflammatory injury and dysfunction in the porcine lung, trachea and liver. The proteomic data showed 51, 99 and 84 proteins that were significantly altered in lung, trachea and liver, respectively. Gene ontology (GO) annotation, KEGG pathway and protein-protein interaction (PPI) network analysis revealed that acute H2S exposure affected the three organs via different mechanisms that were relatively similar between lung and trachea. Further analysis showed that acute H2S exposure caused inflammatory damages in the porcine lung and trachea through activating complement and coagulation cascades, and regulating the hyaluronan metabolic process. Whereas antigen presentation was found in the lung but oxidative stress and cell apoptosis was observed exclusively in the trachea. In the liver, an induced dysfunction was associated with protein processing in the endoplasmic reticulum and lipid metabolism. Further validation of some H2S responsive proteins using western blotting indicated that our proteomics data were highly reliable. Collectively, these findings provide insight into toxic molecular mechanisms that could potentially be targeted for therapeutic intervention for acute H2S intoxication.
Collapse
Affiliation(s)
- Zhen Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, University of Liège, Passage des Déportés 2, Gembloux 5030, Belgium
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xin Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Ruixia Zou
- Graduate School, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qingshi Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qin Fu
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Yanjiao Xie
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qixiang Miao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lei Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, University of Liège, Passage des Déportés 2, Gembloux 5030, Belgium
| |
Collapse
|
17
|
Narykov O, Johnson NT, Korkin D. Predicting protein interaction network perturbation by alternative splicing with semi-supervised learning. Cell Rep 2021; 37:110045. [PMID: 34818539 DOI: 10.1016/j.celrep.2021.110045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022] Open
Abstract
Alternative splicing introduces an additional layer of protein diversity and complexity in regulating cellular functions that can be specific to the tissue and cell type, physiological state of a cell, or disease phenotype. Recent high-throughput experimental studies have illuminated the functional role of splicing events through rewiring protein-protein interactions; however, the extent to which the macromolecular interactions are affected by alternative splicing has yet to be fully understood. In silico methods provide a fast and cheap alternative to interrogating functional characteristics of thousands of alternatively spliced isoforms. Here, we develop an accurate feature-based machine learning approach that predicts whether a protein-protein interaction carried out by a reference isoform is perturbed by an alternatively spliced isoform. Our method, called the alternatively spliced interactions prediction (ALT-IN) tool, is compared with the state-of-the-art PPI prediction tools and shows superior performance, achieving 0.92 in precision and recall values.
Collapse
Affiliation(s)
- Oleksandr Narykov
- Department of Computer Science, and Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Nathan T Johnson
- Department of Computer Science, and Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA; Harvard Program in Therapeutic Sciences, Harvard Medical School, and Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dmitry Korkin
- Department of Computer Science, and Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA.
| |
Collapse
|
18
|
Liu W, Hu W, Hou K, Zhu S. Circular RNA Paired-Related Homeobox 1 Promotes Gastric Carcinoma Cell Progression via Regulating MicroRNA-665/YWHAZ Axis. Dig Dis Sci 2021; 66:3842-3853. [PMID: 33201331 DOI: 10.1007/s10620-020-06705-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gastric carcinoma (GC) is a ubiquitous malignant tumor worldwide. Circular RNA paired-related homeobox 1 (circ-PRRX1), one kind of non-coding RNAs, has been reported to act as a promoter in tumor growth. This study aims to explore the effects of circ-PRRX1 on proliferation, apoptosis, and metastasis in GC and the underlying regulatory mechanisms. METHODS The expression of circ-PRRX1, miR-665, and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta (YWHAZ) mRNA was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot was used to analyze YWHAZ protein expression. 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-Htetrazolium bromide (MTT), flow cytometry, and transwell assay were carried out to assess the viability, apoptosis, migration, and invasion in GC cells. The interaction between miR-665 and circ-PRRX1 or YWHAZ was predicted by StarBase v2.0 and identified by dual-luciferase reporter system. Xenograft mouse model was employed to determine the effects of circ-PRRX1 knockdown on GC growth in vivo. RESULTS Compared with normal tissues and cells, circ-PRRX1 and YWHAZ levels were upregulated, and miR-665 was downregulated in GC tissues and cells. Functionally, circ-PRRX1 knockdown inhibited the viability, migration, and invasion and promoted apoptosis in GC cells, whereas anti-miR-665 abolished these effects. Mechanistically, circ-PRRX1 was confirmed as a sponge of miR-665 to regulate YWHAZ expression. Xenograft mouse model suggested that circ-PRRX1 knockdown reduced GC cells growth in vivo. CONCLUSION Circ-PRRX1 knockdown suppressed GC development by targeting miR-665 to inhibit YWHAZ expression, and the potential molecular mechanism may provide a theoretical basis for GC therapy.
Collapse
Affiliation(s)
- Wei Liu
- Department of General Surgery, Shidong Hospital, No. 999, Shiguang Road, Shanghai, 200438, China
| | - Weigao Hu
- Department of General Surgery, Shidong Hospital, No. 999, Shiguang Road, Shanghai, 200438, China
| | - Kezhu Hou
- Department of General Surgery, Shidong Hospital, No. 999, Shiguang Road, Shanghai, 200438, China
| | - Song Zhu
- Department of General Surgery, Shidong Hospital, No. 999, Shiguang Road, Shanghai, 200438, China.
| |
Collapse
|
19
|
Ribeiro IP, Esteves L, Santos A, Barroso L, Marques F, Caramelo F, Melo JB, Carreira IM. A seven-gene signature to predict the prognosis of oral squamous cell carcinoma. Oncogene 2021; 40:3859-3869. [PMID: 33972685 DOI: 10.1038/s41388-021-01806-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 11/09/2022]
Abstract
The prognosis of oral squamous cell carcinoma (OSCC) patients remains poor without implemented biomarkers in the clinical routine practice to help in the patient's management. With this study we aimed to identify specific prognostic biomarkers for OSCC using a whole genome technology as well as to verify the clinical utility of a head and neck cancer-specific multiplex ligation-dependent probe amplification (MLPA) panel. A genomic characterization of tumor samples from 62 OSCC patients was performed using array comparative genomic hybridization (aCGH) and a more straightforward and cost-effective molecular technology, MLPA. The identification of a genomic signature and prognosis biomarkers was carried out by applying several statistical methods. With aCGH we observed that the chromosomes most commonly altered were 3p, 3q, 5q, 6p, 7q, 8p, 8q, 11q, 15q, 17q, and 18q. The MLPA results showed that the chromosomes with a higher frequency of alterations were 3p, 3q, 8p, 8q, and 11q. We identified a genomic signature with seven genes OCLN (3p21.31), CLDN16 (3q29), SCRIB (3q29), IKBKB (3q22.3), PAK2 (8q22.3), PIK3CB (3q28), and YWHAZ (8q24.3) that together allow to differentiate the patients that developed metastases or relapses after primary tumor treatment, with an overall accuracy of 79%. Amplification of PIK3CB as a predictor of metastases or relapses development was validated using TCGA data. This amplified gene showed a reduction in more than 5 years in the median survival of the patients. The identified biomarkers might have a significant impact in the patients' management and could leverage the OSCC precision medicine.
Collapse
Affiliation(s)
- Ilda Patrícia Ribeiro
- University of Coimbra, Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Luísa Esteves
- University of Coimbra, Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, Coimbra, Portugal
| | - Ana Santos
- University of Coimbra, Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, Coimbra, Portugal
| | - Leonor Barroso
- Maxillofacial Surgery Department, Coimbra Hospital and University Centre (CHUC), EPE, Coimbra, Portugal
| | - Francisco Marques
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Department of Dentistry, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Stomatology Unit, Coimbra Hospital and University Centre (CHUC), EPE, Coimbra, Portugal
| | - Francisco Caramelo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Laboratory of Biostatistics and Medical Informatics, iCBR-Faculty of Medicine, Coimbra, Portugal
| | - Joana Barbosa Melo
- University of Coimbra, Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Isabel Marques Carreira
- University of Coimbra, Cytogenetics and Genomics Laboratory, Institute of Cellular and Molecular Biology, Faculty of Medicine, Coimbra, Portugal. .,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, Coimbra, Portugal. .,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal. .,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
20
|
Hu W, Tao Z, Zhou Q, Zhao D, Gu L, Zhu S, Chen J. Effects of S100 calcium-binding protein A8 (S100A8) and S100 calcium-binding protein A9 (S100A9) on matrix metalloproteinase (MMP) expression in nasopharyngeal carcinoma CNE-2 cells. Transl Cancer Res 2021; 10:1874-1884. [PMID: 35116509 PMCID: PMC8798214 DOI: 10.21037/tcr-21-441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/15/2021] [Indexed: 11/17/2022]
Abstract
Background Studies have shown that S100A8 and S100A9 are highly expressed in a variety of tumors, including NPC, and are associated with tumor invasion and migration. MMPs are associated with the invasion and migration of tumor cells. To further investigate the mechanism by which S100A8 and S100A9 affect the invasion and migration of NPC cells, the present study examined the effects of S100A8 and S100A9 on MMPs in NPC CNE-2 cells. Methods Recombinant pEGFP-N1-S100A8 and recombinant pEGFP-N1-S100A9 overexpression vectors and S100A8 and S100A9 RNA interference (RNAi) vectors were constructed and transfected into NPC CNE-2 cells. The transfection efficiency in each group of cells was assessed, and the gene and protein expression of MMP7, MMP9 and MMP12 were determined. Results The transfection efficiency was approximately 60–70%. Compared with those in the control group, the expression levels of MMP7, MMP9 and MMP12 in the S100A8 and S100A9 overexpression groups was significantly higher (P<0.05), and the expression levels of MMP7, MMP9 and MMP12 in the S100A8-RNAi and S100A9-RNAi groups were significantly lower (P<0.05). The number of cells in S100A8 overexpression group and S100A9 overexpression group at 24, 48 and 72 h was higher than that in RNAi group, RNAi control group, overexpression control group and normal control group, with statistical significance; The cell doubling time in S100A8 and S100A9 overexpression group was significantly shorter than that in RNAi control group, overexpression control group and normal control group, with statistical significance. Conclusions High S100A8 and S100A9 expression may promote the expression of MMP7, MMP9 and MMP12, which are related to the invasion and metastasis of NPC cells.
Collapse
Affiliation(s)
- Wenbing Hu
- Department of Oncology, Huangshi Central Hospital of the Edong Healthcare Group, Huangshi, China
| | - Zezhang Tao
- Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiang Zhou
- Department of Oncology, Huangshi Central Hospital of the Edong Healthcare Group, Huangshi, China
| | - Deqing Zhao
- Department of Oncology, Huangshi Central Hospital of the Edong Healthcare Group, Huangshi, China
| | - Lei Gu
- Department of Oncology, Huangshi Central Hospital of the Edong Healthcare Group, Huangshi, China
| | - Sujin Zhu
- Department of Oncology, Huangshi Central Hospital of the Edong Healthcare Group, Huangshi, China
| | - Jun Chen
- Department of Oncology, Huangshi Central Hospital of the Edong Healthcare Group, Huangshi, China
| |
Collapse
|
21
|
Gogl G, Tugaeva KV, Eberling P, Kostmann C, Trave G, Sluchanko NN. Hierarchized phosphotarget binding by the seven human 14-3-3 isoforms. Nat Commun 2021; 12:1677. [PMID: 33723253 PMCID: PMC7961048 DOI: 10.1038/s41467-021-21908-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
The seven 14-3-3 isoforms are highly abundant human proteins encoded by similar yet distinct genes. 14-3-3 proteins recognize phosphorylated motifs within numerous human and viral proteins. Here, we analyze by X-ray crystallography, fluorescence polarization, mutagenesis and fusicoccin-mediated modulation the structural basis and druggability of 14-3-3 binding to four E6 oncoproteins of tumorigenic human papillomaviruses. 14-3-3 isoforms bind variant and mutated phospho-motifs of E6 and unrelated protein RSK1 with different affinities, albeit following an ordered affinity ranking with conserved relative KD ratios. Remarkably, 14-3-3 isoforms obey the same hierarchy when binding to most of their established targets, as supported by literature and a recent human complexome map. This knowledge allows predicting proportions of 14-3-3 isoforms engaged with phosphoproteins in various tissues. Notwithstanding their individual functions, cellular concentrations of 14-3-3 may be collectively adjusted to buffer the strongest phosphorylation outbursts, explaining their expression variations in different tissues and tumors.
Collapse
Affiliation(s)
- Gergo Gogl
- Equipe Labellisee Ligue 2015, Department of Integrated Structural Biology, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France.
| | - Kristina V Tugaeva
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia
| | - Pascal Eberling
- Equipe Labellisee Ligue 2015, Department of Integrated Structural Biology, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France
| | - Camille Kostmann
- Equipe Labellisee Ligue 2015, Department of Integrated Structural Biology, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France
| | - Gilles Trave
- Equipe Labellisee Ligue 2015, Department of Integrated Structural Biology, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Universite de Strasbourg, Illkirch, France.
| | - Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
22
|
Salazar Y, Zheng X, Brunn D, Raifer H, Picard F, Zhang Y, Winter H, Guenther S, Weigert A, Weigmann B, Dumoutier L, Renauld JC, Waisman A, Schmall A, Tufman A, Fink L, Brüne B, Bopp T, Grimminger F, Seeger W, Pullamsetti SS, Huber M, Savai R. Microenvironmental Th9 and Th17 lymphocytes induce metastatic spreading in lung cancer. J Clin Invest 2021; 130:3560-3575. [PMID: 32229721 DOI: 10.1172/jci124037] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/24/2020] [Indexed: 01/10/2023] Open
Abstract
Immune microenvironment plays a critical role in lung cancer control versus progression and metastasis. In this investigation, we explored the effect of tumor-infiltrating lymphocyte subpopulations on lung cancer biology by studying in vitro cocultures, in vivo mouse models, and human lung cancer tissue. Lymphocyte conditioned media (CM) induced epithelial-mesenchymal transition (EMT) and migration in both primary human lung cancer cells and cell lines. Correspondingly, major accumulation of Th9 and Th17 cells was detected in human lung cancer tissue and correlated with poor survival. Coculturing lung cancer cells with Th9/Th17 cells or exposing them to the respective CM induced EMT in cancer cells and modulated the expression profile of genes implicated in EMT and metastasis. These features were reproduced by the signatory cytokines IL-9 and IL-17, with gene regulatory profiles evoked by these cytokines partly overlapping and partly complementary. Coinjection of Th9/Th17 cells with tumor cells in WT, Rag1-/-, Il9r-/-, and Il17ra-/- mice altered tumor growth and metastasis. Accordingly, inhibition of IL-9 or IL-17 cytokines by neutralizing antibodies decreased EMT and slowed lung cancer progression and metastasis. In conclusion, Th9 and Th17 lymphocytes induce lung cancer cell EMT, thereby promoting migration and metastatic spreading and offering potentially novel therapeutic strategies.
Collapse
Affiliation(s)
- Ylia Salazar
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Xiang Zheng
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Hartmann Raifer
- Institute for Medical Microbiology and.,CoreFacility Flow Cytometry, University of Marburg, Marburg, Germany
| | | | | | - Hauke Winter
- Translational Research Unit, Thoraxklinik at Heidelberg University, member of the DZL, Heidelberg, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Laure Dumoutier
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Anja Schmall
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Amanda Tufman
- Respiratory Medicine and Thoracic Oncology, Internal Medicine V, Ludwig-Maximilians-University of Munich and Thoracic Oncology Centre, member of the DZL, Munich, Germany
| | - Ludger Fink
- Institute of Pathology and Cytology, Wetzlar, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany. Research Center for Immunotherapy and University Medical Center, Johannes Gutenberg-University, Mainz, Germany. German Cancer Consortium, Heidelberg, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany.,Institute or Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany
| | | | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany.,Institute or Lung Health (ILH), Justus Liebig University, Giessen, Germany
| |
Collapse
|
23
|
Lage-Vickers S, Bizzotto J, Valacco MP, Sanchis P, Nemirovsky S, Labanca E, Scorticati C, Mazza O, Mitrofanova A, Navone N, Vazquez E, Cotignola J, Gueron G. The expression of YWHAZ and NDRG1 predicts aggressive outcome in human prostate cancer. Commun Biol 2021; 4:103. [PMID: 33483585 PMCID: PMC7822895 DOI: 10.1038/s42003-020-01645-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/16/2020] [Indexed: 01/27/2023] Open
Abstract
Some prostate cancers (PCas) are histo-pathologically grouped within the same Gleason Grade (GG), but can differ significantly in outcome. Herein, we aimed at identifying molecular biomarkers that could improve risk prediction in PCa. LC ESI-MS/MS was performed on human PCa and benign prostatic hyperplasia (BPH) tissues and peptide data was integrated with omic analyses. We identified high YWHAZ and NDRG1 expression to be associated with poor PCa prognosis considering all Gleason scores (GS). YWHAZ and NDRG1 defined two subpopulations of PCa patients with high and intermediate risk of death. Multivariable analyses confirmed their independence from GS. ROC analysis unveiled that YWHAZ outperformed GS beyond 60 months post-diagnosis. The genomic analysis of PCa patients with YWHAZ amplification, or increased mRNA or protein levels, revealed significant alterations in key DNA repair genes. We hereby state the relevance of YWHAZ in PCa, showcasing its role as an independent strong predictor of aggressiveness.
Collapse
Affiliation(s)
- Sofia Lage-Vickers
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Juan Bizzotto
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Maria Pia Valacco
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Pablo Sanchis
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Sergio Nemirovsky
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carlos Scorticati
- Cátedra de Urología, Hospital de Clínicas, Buenos Aires, C1120AAR, Argentina
| | - Osvaldo Mazza
- Cátedra de Urología, Hospital de Clínicas, Buenos Aires, C1120AAR, Argentina
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, Rutgers School of Health Professions, Rutgers Cancer Institute of New Jersey, New Jersey, NJ, 07101, USA
| | - Nora Navone
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Elba Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina
| | - Javier Cotignola
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina.
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, C1428EGA, Argentina.
| |
Collapse
|
24
|
Gong Y, Wei Z, Liu J. MiRNA-1225 Inhibits Osteosarcoma Tumor Growth and Progression by Targeting YWHAZ. Onco Targets Ther 2021; 14:15-27. [PMID: 33442263 PMCID: PMC7797335 DOI: 10.2147/ott.s282485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/04/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction Osteosarcoma is the most common bone tumor and is characterized by the presence of malignant mesenchymal cells produced in the bone stroma. MiRNAs are known to function as post-transcriptional negative regulators of gene expression. Emerging evidence showed that miR-1225-5P functions as a tumor suppressor in several types of cancers. The detailed mechanisms of which miR-1225-5P suppresses tumor growth are not fully understood. The objective of the present study was to test the hypothesis that miR-1225-5P inhibits osteosarcoma cell growth in vitro and tumor growth in vivo by targeting YWHAZ expression. Methods Real-time PCR and Western blot were carried out to test the expression of miR-1225-5P and YWHAZ in osteosarcoma cell lines. Luciferase assay was used to demonstrate whether miR-1225-5P targets YWHAZ 3ʹ UTR. To assess the function of miR-1225-5P in human osteosarcoma cell lines, gain-of-function and loss-of-function of miR-1225-5P were performed by transfecting miR-1225-5P mimic or miR-1225-5P inhibitor into osteosarcoma cell lines. Furthermore, cell cycle analysis was performed to elucidate the possible mechanisms of the action of miR-1225-5P and YWHAZ in human osteosarcoma cells. The potential therapeutic effect of miR-1225-5p was tested in human osteosarcoma xenograft mouse model, by intravenous injection of miR-1225-5P into nude mice. Tumor sizes were measured and lung metastasis was counted after the mice were sacrificed. Results The expression of miR-1225-5P was inversely correlated with the expression of YWHAZ in human osteosarcoma cell lines. Database search revealed that miR-1225-5P targeted YWHAZ 3ʹ UTR. Transfection of miR-1225-5P mimic downregulated YWHAZ expression, which was demonstrated by real-time PCR, Western blot and luciferase assay. Over-expression of miR-1225-5P reduced human osteosarcoma cell growth, migration and invasion by downregulating YWHAZ expression. Cell growth, migration and invasion were increased by inhibiting miR-1225-5P in human osteosarcoma cells. The inhibition of cell growth, migration and invasion was rescued by over-expression of YWHAZ in osteosarcoma cells. Cell cycle analysis revealed that miR-1225-5P inhibited G1/G0 phase exit. In vivo xenograft model demonstrated that miR-1225-5P inhibited in vivo osteosarcoma tumor growth and lung metastasis. Conclusion Our findings suggested that miR-1225-5P inhibits osteosarcoma cell growth in vitro and tumor growth in vivo by targeting YWHAZ. This study suggested that miR-1225-5P can serve as a potential therapeutic method for treating osteosarcoma.
Collapse
Affiliation(s)
- Yubao Gong
- Department of Orthopedics, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Zhengren Wei
- Department of Pharmacology, Basic Medical School, Jilin University, Changchun 130021, People's Republic of China
| | - Jianguo Liu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| |
Collapse
|
25
|
Nishibeppu K, Komatsu S, Kiuchi J, Kishimoto T, Takashima Y, Shoda K, Arita T, Kosuga T, Konishi H, Shiozaki A, Kubota T, Okamoto K, Fujiwara H, Tsuda H, Otsuji E. TRIM37 contributes to malignant outcomes and CDDP resistance in gastric cancer. J Cancer 2021; 12:316-325. [PMID: 33391428 PMCID: PMC7739001 DOI: 10.7150/jca.47577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022] Open
Abstract
Background: TRIM37 (Tripartite Motif Containing 37) is an E3 ubiquitin ligase for histone H2A and inhibits transcription in several genes. However, it is not known whether it plays a role in gastric cancer (GC). In this study, we tested whether TRIM37 acts as a cancer-promoting factor by being overexpressed in GC. Methods: We analyzed GC cell lines and 124 primary tumors, which were curatively resected in our hospital between 2001 and 2003. Results: Overexpression of the TRIM37 protein was detected in almost all GC cell lines and GC samples (76 out of 124 cases) and was significantly correlated with lymphatic and venous invasion, advanced T-Stage, N-Stage, histology and high recurrence rate. Patients with TRIM37 overexpressing tumors had a worse survival rate than those with non-expressing tumors (P=0.0057). Moreover, TRIM37 positivity was identified as an independent factor predicting worse outcomes (P=0.018, Hazard ratio 3.41). The apoptotic cell analysis showed that the knockdown of TRIM37 increased apoptosis in comparison with the control. In TRIM37 overexpressing GC cells, knockdown of TRIM37 suppressed the migration and invasion. Conclusions: TRIM37 plays a crucial role in tumor malignant potential through its overexpression and highlight its usefulness as a prognostic factor and potential therapeutic target in GC.
Collapse
Affiliation(s)
- Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Takuma Kishimoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yusuke Takashima
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Katsutoshi Shoda
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hitoshi Tsuda
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan.,Department of Basic Pathology, National Defense Medical College, Tokorozawa, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
26
|
Go YY, Kim SR, Kim DY, Chae SW, Song JJ. Cannabidiol enhances cytotoxicity of anti-cancer drugs in human head and neck squamous cell carcinoma. Sci Rep 2020; 10:20622. [PMID: 33244087 PMCID: PMC7692486 DOI: 10.1038/s41598-020-77674-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022] Open
Abstract
Cannabidiol (CBD) has anti-tumorigenic activity. However, the anti-cancer effect of CBD on head and neck squamous cell carcinoma (HNSCC) remains unclear. The cytotoxicity of CBD on HNSCC was analyzed using cell survival and colony-forming assays in vitro. RNA-seq was used for determining the mechanism underlying CBD-induced cell death. Xenograft mouse models were used to determine CBD’s effects in vivo. CBD treatment significantly reduced migration/invasion and viability of HNSCC cells in a dose- and time-dependent manner. HNSCC mouse xenograft models revealed anti-tumor effects of CBD. Furthermore, combinational treatment with CBD enhanced the efficacy of chemotherapy drugs. Apoptosis and autophagy processes were involved in CBD-induced cytotoxicity of HNSCCs. RNA-seq identified decreased expression of genes associated with DNA repair, cell division, and cell proliferation, which were involved in CBD-mediated cytotoxicity toward HNSCCs. We identified CBD as a new potential anti-cancer compound for single or combination therapy of HNSCC.
Collapse
Affiliation(s)
- Yoon Young Go
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, South Korea.,Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Su Ra Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, South Korea
| | - Do Yeon Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, South Korea
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, South Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-dong, Guro-gu, Seoul, 08308, South Korea. .,Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Parajón E, Surcel A, Robinson DN. The mechanobiome: a goldmine for cancer therapeutics. Am J Physiol Cell Physiol 2020; 320:C306-C323. [PMID: 33175572 DOI: 10.1152/ajpcell.00409.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cancer progression is dependent on heightened mechanical adaptation, both for the cells' ability to change shape and to interact with varying mechanical environments. This type of adaptation is dependent on mechanoresponsive proteins that sense and respond to mechanical stress, as well as their regulators. Mechanoresponsive proteins are part of the mechanobiome, which is the larger network that constitutes the cell's mechanical systems that are also highly integrated with many other cellular systems, such as gene expression, metabolism, and signaling. Despite the altered expression patterns of key mechanobiome proteins across many different cancer types, pharmaceutical targeting of these proteins has been overlooked. Here, we review the biochemistry of key mechanoresponsive proteins, specifically nonmuscle myosin II, α-actinins, and filamins, as well as the partnering proteins 14-3-3 and CLP36. We also examined a wide range of data sets to assess how gene and protein expression levels of these proteins are altered across many different cancer types. Finally, we determined the potential of targeting these proteins to mitigate invasion or metastasis and suggest that the mechanobiome is a goldmine of opportunity for anticancer drug discovery and development.
Collapse
Affiliation(s)
- Eleana Parajón
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexandra Surcel
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
28
|
G3BP1 interacts with YWHAZ to regulate chemoresistance and predict adjuvant chemotherapy benefit in gastric cancer. Br J Cancer 2020; 124:425-436. [PMID: 32989225 PMCID: PMC7852868 DOI: 10.1038/s41416-020-01067-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND A large proportion of gastric cancer patients are susceptible to chemoresistance, while the underlying mechanism remains obscure. Stress granules (SGs) play a self-defence role for tumour cells in inhibiting chemotherapy-induced apoptosis. As an SG assembly effector, G3BP1 (Ras-GTPase-activating protein SH3 domain-binding protein) has been reported to be overexpressed in gastric cancer; thus, here we aim to explore its potent roles in gastric cancer chemoresistance. METHODS Kaplan–Meier analysis was used to compare survival rates in gastric cancer patients with different G3BP1 expression. The influence of G3BP1 on gastric cancer cell chemoresistance and apoptosis were evaluated by in vitro and in vivo approaches. The interaction between G3BP1 and YWHAZ was assessed by immunohistochemistry, immunoprecipitation and immunofluorescence. RESULTS G3BP1 was associated with the poor outcome of gastric cancer patients who received adjuvant chemotherapy. G3BP1 knockdown significantly increased the sensitivity of gastric cancer cells to chemotherapy drugs. Mechanically, cell apoptosis and pro-apoptotic-associated molecules were significantly elevated upon G3BP1 depletion. Gene co-expression network analyses identified YWHAZ as the critical interlayer of G3BP1; as a result, G3BP1 interacted with YWHAZ to sequester Bax into the cytoplasm. Clinically, G3BP1highYWHAZhigh gastric cancer patients displayed the worst outcome compared with other patients after chemotherapy. CONCLUSIONS The expression of G3BP1 and YWHAZ could predict the adjuvant chemotherapy benefit in gastric cancer patients.
Collapse
|
29
|
Liu Y, Guo J, Shen K, Wang R, Chen C, Liao Z, Zhou J. Paclitaxel Suppresses Hepatocellular Carcinoma Tumorigenesis Through Regulating Circ-BIRC6/miR-877-5p/ YWHAZ Axis. Onco Targets Ther 2020; 13:9377-9388. [PMID: 33061425 PMCID: PMC7519811 DOI: 10.2147/ott.s261700] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
Background Paclitaxel is an effective chemotherapeutic agent for the treatment of cancer patients. Accumulating evidence suggests that circular RNAs (circRNAs) play critical roles in the occurrence and development of human cancers. However, there are few studies on interactions between paclitaxel and circRNAs in hepatocellular carcinoma (HCC). Materials and Methods Cell counting kit-8 (CCK-8) assay and colony formation assay were conducted to determine cell proliferation. Cell apoptosis was assessed by flow cytometry. The expression levels of circRNA baculoviral IAP repeat-containing 6 (circ-BIRC6), microRNA-877-5p (miR-877-5p), and tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta (YWHAZ) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The mice xenograft model was established to investigate the roles of circ-BIRC6 and paclitaxel in vivo. The interaction between miR-877-5p and circ-BIRC6 or YWHAZ was predicted by bioinformatics analysis and verified by dual-luciferase reporter assay. Western blot assay was applied for measuring the protein expression of YWHAZ. Results Paclitaxel suppressed HCC tumorigenesis through decreasing cell proliferation and accelerating apoptosis. Circ-BIRC6 and YWHAZ were upregulated, and miR-877-5p was downregulated in HCC tissues and cells. Paclitaxel treatment inhibited the expression of circ-BIRC6 and YWHAZ while promoted the expression of miR-877-5p. Circ-BIRC6 overexpression or miR-877-5p interference reversed the inhibitory effect of paclitaxel on HCC tumorigenesis. Moreover, miR-877-5p could specially bind to YWHAZ, and its knockdown abated the suppressive effect of circ-BIRC6 depletion on HCC tumorigenesis. Additionally, YWHAZ was identified as a direct target of miR-877-5p. Besides, circ-BIRC6 functioned as a molecular sponge of miR-877-5p to regulate YWHAZ expression. Conclusion Paclitaxel limited HCC tumorigenesis via modulating circ-BIRC6/miR-877-5p/YWHAZ axis, providing a novel therapeutic approach for the treatment of HCC.
Collapse
Affiliation(s)
- Yi Liu
- Department of Traditional Chinese Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| | - Jianchao Guo
- Department of Gastroenterology, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| | - Ka Shen
- Department of General Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| | - Renlong Wang
- Department of General Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| | - Cheng Chen
- Department of General Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| | - Zhiyuan Liao
- Department of General Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| | - Jianbo Zhou
- Department of General Surgery, Suizhou Hospital, Hubei University of Medicine, Suizhou 441300, People's Republic of China
| |
Collapse
|
30
|
Zhang W, Liu X, Piao L. Chlorogenic acid-enriched extract of Ilex kudingcha C.J. Tseng tea inhibits neutrophil recruitment in injured zebrafish by promoting reverse migration via the focal adhesion pathway. J Food Biochem 2020; 44:e13279. [PMID: 32686853 DOI: 10.1111/jfbc.13279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
Abstract
Neutrophil-regulated inflammation plays crucial roles in tissue damage and repair. Dysregulation of the neutrophil response system can contribute to diseases such as cancer. Clearance of excessive neutrophils at the site of inflammation by reverse migration provides a promising strategy to mitigate the negative effects. Chlorogenic acid treatment of injured zebrafish embryos showed low-developmental toxicity. Using a transgenic zebrafish Tg (mpx: egfp) model, chlorogenic acid-enriched kudingcha extract promoted neutrophil reverse migration via phosphorylation of ERK and AKT. Using i-TRAQ analysis, differentially expressed proteins involved in focal adhesion were identified, such as: Cdc42, SRC, MLC, ITGA, and Calpain. In support of this, ERK and AKT proteins are involved in the focal adhesion pathway. Real time qPCR determined that CGA downregulates genes associated with cancer metastasis, such as: HSPA5, YWHAZ, RP17, and ITGAV. Together, these results suggest that CGA-enriched Kudingcha extract may have potential as an anticancer or anti-inflammatory therapeutic agent. PRACTICAL APPLICATIONS: Ilex kudingcha C.J Tseng, commonly referred to as the large-leaved kudingcha, is a tea variety naturally rich in chlorogenic acid. Chlorogenic acid, the ester of caffeic and quinic acids, has antioxidant, antibacterial, anticancer, and anti-inflammatory, activities. Kudingcha has several known biological functions, including: anticancer, anti-inflammatory, antidiabetic, and hypolipidemic effects. Treatment with kudingcha extract reduces the recruitment of neutrophils, potentially by inhibiting focal adhesion, and activation of cancer metastasis-related genes. Importantly, kudingcha extract could be used as an alternative nutritional supplement for anticancer or anti-inflammation via its ability to suppress neutrophil recruitment.
Collapse
Affiliation(s)
- Weihao Zhang
- School of Life and Pharmaceutical Science, Hainan University, Haikou, China
| | - Xiande Liu
- School of Life and Pharmaceutical Science, Hainan University, Haikou, China
| | - Linghua Piao
- Department of Physiology, Hainan Medical University, Haikou, China
| |
Collapse
|
31
|
Shen Z, Chai T, Luo F, Liu Z, Xu H, Zhang P, Kang M, Chen S. Loss of miR-204-5p Promotes Tumor Proliferation, Migration, and Invasion Through Targeting YWHAZ/PI3K/AKT Pathway in Esophageal Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:4679-4690. [PMID: 32547097 PMCID: PMC7263804 DOI: 10.2147/ott.s243215] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/28/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE MicroRNAs dysregulation has been confirmed in multiple malignancies. This paper reported the molecular mechanism of miR-204-5p in esophageal squamous cell carcinoma (ESCC). METHODS miR-204-5p expression in 30 ESCC tumor tissues and 10 normal tissues was downloaded from RNA-seq data. ESCC tissues/normal tissues of 97 ESCC patients were collected. TE-1 and KYSE510 cells were transfected by miR-204-5p mimic, inhibitor, siYWHAZ or their corresponding controls. The phenotype of cells was detected by CCK-8 assay, transwell experiment, and flow cytometry. Luciferase reporter gene assay and RNA-binding protein immunoprecipitation (RIP) were performed to verify the targeting relationship between miR-204-5p and YWHAZ. miR-204-5p and YWHAZ expression in tissues/cells was detected by qRT-PCR and Western blot. Xenograft tumor experiment was performed. RESULTS miR-204-5p expression was declined in ESCC patients and cells, which was indicated the poor outcome of patients. Compared with siNC group, TE-1 cells in miR-204-5p inhibitor group had higher OD450 value, less cell percentage in G1 phase, and more cell percentage in S phase, lower apoptosis percentage, and higher migration and invasion cell numbers. Moreover, KYSE510 cells of miR-204-5p mimic group showed lower OD450 value, more cell percentage in G1 phase and less cell percentage in S phase, higher apoptosis percentage, and lower migration and invasion cell numbers than control. YWHAZ was directly inhibited by miR-204-5p. Relative to siNC group, TE-1 cells of miR-inhibitor group exhibited higher YWHAZ protein expression, higher OD450 value, less cell percentage in G1 phase and more cell percentage in S phase, lower apoptosis percentage, higher migration and invasion cell numbers, and higher p-PI3K/PI3K and p-AKT/AKT protein expression, while siYWHAZ rescued the effects of miR-inhibitor. miR-204-5p up-regulation inhibited ESCC growth in vivo. CONCLUSION miR-204-5p inhibits ESCC progression by targeted inhibition of YWHAZ/PI3K/AKT.
Collapse
Affiliation(s)
- Zhimin Shen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| | - Tianci Chai
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| | - Fei Luo
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| | - Zhun Liu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| | - Hui Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou350001, People’s Republic of China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou350122, People’s Republic of China
| | - Sui Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou350001, People’s Republic of China
| |
Collapse
|
32
|
Xiao J, Zhang R, Peng J, Yang Z. BAP1 maintains chromosome stability by stabilizing DIDO1 in renal cell carcinoma. Am J Cancer Res 2020; 10:1455-1466. [PMID: 32509391 PMCID: PMC7269789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/04/2020] [Indexed: 06/11/2023] Open
Abstract
BRCA1-associated protein 1 (BAP1) is a member of the ubiquitin C-terminal hydrolase family of deubiquitinating enzymes and is implicated in transcriptional regulation. The BAP1 gene is mutated in 5%-15% of patients with clear cell renal cell carcinoma (ccRCC), the most common form of renal cancer, which suggests that BAP1 is a tumor suppressor. However, whether BAP1 influences the progression of ccRCC tumors expressing wildtype (WT) BAP1 is unclear. Here, we identified DIDO1 as a bona fide substrate for BAP1. DIDO1 is a component of the centrosome proteins and plays an essential role in spindle assembly. BAP1 binds to DIDO1 and stabilizes DIDO1 through de-ubiquitination. BAP1 contributes to chromosome stability partially via DIDO1. A positive correlation was identified between BAP1 and DIDO1 expression in ccRCC tissues. Downregulation of both BAP1-loss and DIDO1 protein expression in ccRCC was associated with adverse clinicopathological features. This study revealed a novel mechanism involving BAP1 in the regulation of DIDO1 stability, and the results also provide insight into the relationship between BAP1 mutations and chromosome instability in ccRCC.
Collapse
Affiliation(s)
- Jiantao Xiao
- Department of Urology, Zhongnan Hospital of Wuhan UniversityWuhan, China
| | - Ruhan Zhang
- Department of Nursing, Zhongnan Hospital of Wuhan UniversityWuhan, China
| | - Jingtao Peng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Zhonghua Yang
- Department of Urology, Zhongnan Hospital of Wuhan UniversityWuhan, China
| |
Collapse
|
33
|
Kumari M, Mohan A, Ecelbarger CM, Gupta A, Prasad N, Tiwari S. miR-451 Loaded Exosomes Are Released by the Renal Cells in Response to Injury and Associated With Reduced Kidney Function in Human. Front Physiol 2020; 11:234. [PMID: 32322216 PMCID: PMC7158952 DOI: 10.3389/fphys.2020.00234] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 03/02/2020] [Indexed: 12/19/2022] Open
Abstract
Micro-RNAs (miRs) encapsulated inside urinary exosomes (uEs) have the potential as early biomarkers. Previously, we reported that a rise in uE miR-451 predicted albuminuria in diabetic rats; however, whether the rise was protective or detrimental, and occurred in response to injury or general hyperglycemia, was unknown. To address this, we studied both human and rat models of renal disease. In humans, uE miR-451 was approximately twofold higher in subjects with early-stage chronic kidney disease (CKD; serum creatinine < 2.0 mg/dl; n = 28), as compared to age-matched healthy controls (n = 23), and had a significant negative correlation with estimated glomerular filtration rate (eGFR) (r2 = −0.10, p = 0.01). Subgroup analysis of CKD subjects showed that those without diabetes had slightly (∼30%) but significantly higher uE miR-451 as compared to those with diabetes, with no differences in albumin excretion, eGFR, serum sodium, and potassium. Using human proximal tubule (hPT) cells, we found that locked nucleic acid (LNA) inhibition of miR-451 resulted in a significant increase in the messenger RNA (mRNA) expression of kidney-injury-associated miR-451 targets, e.g., CAB39, TBX1, and YWHAZ, as compared to treatment with a control LNA. Moreover, hPT cells and their secreted exosomes showed an increase in miR-451 in response to mechanical injury but not high glucose (20 versus 5 mM). For further proof of concept, in diabetic rats, we showed that atorvastatin (AT), a treatment proven to attenuate renal injury without affecting systemic glucose levels, reduced uE miR-451 with the concomitant restoration of renal miR-451. These data elucidate the stimuli for renal miR-451 expression and exosomal release and support its role as a therapeutic target and early biomarker for renal injury in humans.
Collapse
Affiliation(s)
- Manju Kumari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Aradhana Mohan
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | | | - Amit Gupta
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Narayan Prasad
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
34
|
Ashrafizadeh M, Rafiei H, Mohammadinejad R, Farkhondeh T, Samarghandian S. Wnt-regulating microRNAs role in gastric cancer malignancy. Life Sci 2020; 250:117547. [PMID: 32173311 DOI: 10.1016/j.lfs.2020.117547] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is responsible for high morbidity and mortality worldwide. This cancer claims fifth place among other cancers. There are a number of factors associated with GC development such as alcohol consumption and tobacco smoking. It seems that genetic factors play significant role in GC malignancy and progression. MicroRNAs (miRs) are short non-coding RNA molecules with negative impact on the expression of target genes. A variety of studies have elucidated the potential role of miRs in GC growth. Investigation of molecular pathways has revealed that miRs function as upstream modulators of Wnt signaling pathway. This signaling pathway involves in important biological processes such as cell proliferation and differentiation, and its dysregulation is associated with GC invasion. At the present review, we demonstrate that how miRs regulate Wnt signaling pathway in GC malignancy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Hossein Rafiei
- Department of Biology, Faculty of Sciences, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
35
|
Tang C, Zhao R, Ni H, Zhao K, He Y, Fang S, Chen Q. Molecule mechanisms of Ganoderma lucidum treated hepatocellular carcinoma based on the transcriptional profiles and miRNA-target network. Biomed Pharmacother 2020; 125:110028. [PMID: 32106374 DOI: 10.1016/j.biopha.2020.110028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/13/2020] [Accepted: 02/17/2020] [Indexed: 12/19/2022] Open
Abstract
Ganoderma lucidum has salutary effects on tumor treatment, including pancreatic cancer and hepatocellular carcinoma. However, the molecular mechanisms underlying Ganoderma lucidum therapy is obscure. In this study, the Hepa1-6-bearing C57 BL/6 mouse model was utilized to explore the therapeutic efficacy of Ganoderma lucidum extract (GLE), documenting that it could effectively inhibit tumor growth. The microRNA (miRNA) profiles of GLE-treated and untreated mice were detected, and 25 differentially expressed (DE) miRNAs were determined, including 24 up-expressed and one down-expressed miRNAs. Using the ClusterOne algorithm, 8 hub miRNAs were isolated from the established miRNA-target network. The qRT-PCR assay demonstrated that these 8 miRNAs were up-expressed in the GLE treated tumor mice. Furthermore, the mRNA profiles showed that there are 76 DE mRNAs between GLE treated and model groups. The protein-protein interaction (PPI) network shows that Cntn1, Irs1, Nfkbia, Rybp and Ywhaz playing important roles, and qRT-PCR further revealed they were down-expressed in GLE treated Hepa1-6-bearing C57 BL/6 mice. The rebuilt miRNA-target network was shown that these 5 mRNAs were regulated by mmu-mir-23a-5p, -3102-3p, -337-3p, and -467a-3p, respectively. This study suggested that these 4 interesting miRNAs were potential biomarkers for evaluation of GLE efficacy, which may down-regulate the expression of Cntn1, Irs1, Nfkbia, Rybp and Ywhaz, and mediate many signaling pathways occurring in tumor treatment.
Collapse
Affiliation(s)
- Chenchen Tang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruolin Zhao
- Department of Basic Medicine, Naval Medical University, Shanghai 200433, China
| | - Hongmei Ni
- School of Basic Medicine College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kunpeng Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - YuMin He
- School of Basic Medicine College, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shengquan Fang
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Qilong Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
36
|
Gan Y, Ye F, He XX. The role of YWHAZ in cancer: A maze of opportunities and challenges. J Cancer 2020; 11:2252-2264. [PMID: 32127952 PMCID: PMC7052942 DOI: 10.7150/jca.41316] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 11/30/2019] [Indexed: 12/21/2022] Open
Abstract
YWHAZ (also named 14-3-3ζ) is a central hub protein for many signal transduction pathways and plays a significant role in tumor progression. Accumulating evidences have demonstrated that YWHAZ is frequently up-regulated in multiple types of cancers and acts as an oncogene in a wide range of cell activities including cell growth, cell cycle, apoptosis, migration, and invasion. Moreover, YWHAZ was reported to be regulated by microRNAs (miRNAs) or long non-coding RNAs and exerted its malignant functions by targeting downstream molecules like protein kinase, apoptosis proteins, and metastasis-related molecules. Additionally, YWHAZ may be a potential biomarker of diagnosis, prognosis and chemoresistance in several cancers. Targeting YWHAZ by siRNA, shRNA or miRNA was reported to have great help in suppressing malignant properties of cancer cells. In this review, we perform literature and bioinformatics analysis to reveal the oncogenic role and molecular mechanism of YWHAZ in cancer, and discuss the potential clinical applications of YWHAZ concerning diagnosis, prognosis, and therapy in malignant tumors.
Collapse
Affiliation(s)
- Yun Gan
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Ye
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-Xing He
- Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Wu X, Zhou X, Ding X, Chu M, Liang C, Pei J, Xiong L, Bao P, Guo X, Yan P. The Selection of Reference Genes for Quantitative Real-Time PCR in the Ashidan Yak Mammary Gland During Lactation and Dry Period. Animals (Basel) 2019; 9:ani9110943. [PMID: 31717620 PMCID: PMC6912359 DOI: 10.3390/ani9110943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/02/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The Ashidan yak is a new cultivated breed which has polled characteristics and a mild temperament. Improving milk yield is an important aspect of a breeding program for this breed. The mammary gland undergoes dramatic physiological and metabolic changes during the transition from lactation to dry periods, which involves the expression and regulation of a great number of genes. Quantification of gene expression levels by real-time quantitative polymerase chain reaction (RT-qPCR) is important to reveal the molecular mechanisms of mammary gland development and lactation. The accuracy of RT-qPCR is strongly influenced by the expression stability of reference genes, however, a systematic approach for selecting reference genes used for analyzing gene expression of the Ashidan yak has not been developed. In this study, we selected reference genes and analyzed their expression stability at different physiological stages (lactation and dry period). We found the hydroxymethylbilane synthase gene (HMBS) and the tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide gene (YWHAZ) were the most stable genes of the mammary gland of the Ashidan yak. These results help to improve the accuracy of gene expression analysis and provide a basis for future functional studies of target gene expression in the mammary gland of the Ashidan yak. Abstract Investigating the critical genes related to milk synthesis is essential for the improvement of the milk yield of the yak. Real-time quantitative polymerase chain reaction (RT-qPCR) is a reliable and widely used method to measure and evaluate gene expression levels. Selection of suitable reference genes is mandatory to acquire accurate normalization of gene expression results from RT-qPCR. To select the most stable reference genes for reliable normalization of mRNA expression by RT-qPCR in the mammary gland of the Ashidan yak, we selected 16 candidate reference genes and analyzed their expression stability at different physiological stages (lactation and dry period). The expression stability of the candidate reference genes was assessed using geNorm, NormFinder, BestKeeper, Delta Ct, and RefFinder methods. The results showed that the hydroxymethylbilane synthase gene (HMBS) and the tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide gene (YWHAZ) were the most stable genes across all treatment samples. The reliability of selected reference genes was validated by normalizing relative expression of the lactation-related 60S ribosomal protein L35 gene (RPL35). The relative expression of RPL35 varied considerably according to the different reference genes. This work provides valuable information to further promote research in the molecular mechanisms involved in lactation and mammary gland development and provides a foundation for the improvement of the milk yield and quality of the Ashidan yak.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xian Guo
- Correspondence: (X.G.); (P.Y.); Tel.: +86-0931-2115257 (X.G.); +86-0931-2115288 (P.Y.)
| | - Ping Yan
- Correspondence: (X.G.); (P.Y.); Tel.: +86-0931-2115257 (X.G.); +86-0931-2115288 (P.Y.)
| |
Collapse
|
38
|
Yang B, Sun L, Liang L. MiRNA-802 suppresses proliferation and migration of epithelial ovarian cancer cells by targeting YWHAZ. J Ovarian Res 2019; 12:100. [PMID: 31640760 PMCID: PMC6806521 DOI: 10.1186/s13048-019-0576-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 09/27/2019] [Indexed: 12/18/2022] Open
Abstract
Background The imbalance of expression of microRNA-802 may have a significant place in tumor progression. However, the bio-function of epithelial ovarian cancer cells remains unclear. Therefore, we setup this study to explore the pathogenesis of epithelial ovarian cancer based on microRNA-802. Methods RT-qPCR analysis was used to measure the expression level of microRNA802 and YWHAZ in epithelial ovarian cancer. CCK-8, colony formation, flow cytometry and transwell assay were used to detect the effects of microRNA-802 on cell proliferation, apoptosis, invasion and migration. Target gene prediction and screening, luciferase reporting experiments were applied to validate the downstream target genes of microRNA-802. The effects of microRNA-802 on the expression of YWHAZ and its biological effects were measured by Western blotting and RT-qPCR. Results Compared with normal cell lines and tissues, the expression level of microRNA-802 was obviously down-regulated in cancer related cell lines and tissues. Overexpression of microRNA-802 could obviously inhibit the invasion and proliferation and induce apoptosis. In addition, YWHAZ was the binding target protein of miR-802 for epithelial ovarian cancer cells. YWHAZ was obviously up-regulated in human epithelial ovarian cancer cells, and YWHAZ was negatively correlated with the expression of miR-802. YWHAZ can partly eliminate the inhibitory effect caused by overexpression of miR-802 on growth and metastasis of epithelial ovarian cancer cells. Conclusion miR-802 can regulate the occurrence and development of epithelial ovarian cancer by targeting YWHAZ.
Collapse
Affiliation(s)
- Bo Yang
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital, 398 Zhongshan West Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Li Sun
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital, 398 Zhongshan West Road, Shijiazhuang, 050000, Hebei Province, China
| | - Lei Liang
- Department of Obstetrics and Gynecology, Bethune International Peace Hospital, 398 Zhongshan West Road, Shijiazhuang, 050000, Hebei Province, China
| |
Collapse
|
39
|
Conforte AJ, Tuszynski JA, da Silva FAB, Carels N. Signaling Complexity Measured by Shannon Entropy and Its Application in Personalized Medicine. Front Genet 2019; 10:930. [PMID: 31695721 PMCID: PMC6816034 DOI: 10.3389/fgene.2019.00930] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/05/2019] [Indexed: 12/28/2022] Open
Abstract
Traditional approaches to cancer therapy seek common molecular targets in tumors from different patients. However, molecular profiles differ between patients, and most tumors exhibit inherent heterogeneity. Hence, imprecise targeting commonly results in side effects, reduced efficacy, and drug resistance. By contrast, personalized medicine aims to establish a molecular diagnosis specific to each patient, which is currently feasible due to the progress achieved with high-throughput technologies. In this report, we explored data from human RNA-seq and protein–protein interaction (PPI) networks using bioinformatics to investigate the relationship between tumor entropy and aggressiveness. To compare PPI subnetworks of different sizes, we calculated the Shannon entropy associated with vertex connections of differentially expressed genes comparing tumor samples with their paired control tissues. We found that the inhibition of up-regulated connectivity hubs led to a higher reduction of subnetwork entropy compared to that obtained with the inhibition of targets selected at random. Furthermore, these hubs were described to be participating in tumor processes. We also found a significant negative correlation between subnetwork entropies of tumors and the respective 5-year survival rates of the corresponding cancer types. This correlation was also observed considering patients with lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) based on the clinical data from The Cancer Genome Atlas database (TCGA). Thus, network entropy increases in parallel with tumor aggressiveness but does not correlate with PPI subnetwork size. This correlation is consistent with previous reports and allowed us to assess the number of hubs to be inhibited for therapy to be effective, in the context of precision medicine, by reference to the 100% patient survival rate 5 years after diagnosis. Large standard deviations of subnetwork entropies and variations in target numbers per patient among tumor types characterize tumor heterogeneity.
Collapse
Affiliation(s)
- Alessandra J Conforte
- Laboratory of Biological Systems Modeling, Center of Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Laboratory of Computational Modeling of Biological Systems, Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jack Adam Tuszynski
- Department of Oncology, University of Alberta, Edmonton, AB, Canada.,Department of Physics, University of Alberta, Edmonton, AB, Canada.,DIMEAS, Politecnico di Torino, Turin, Italy
| | - Fabricio Alves Barbosa da Silva
- Laboratory of Computational Modeling of Biological Systems, Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Nicolas Carels
- Laboratory of Biological Systems Modeling, Center of Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
Hashemi M, Zali A, Hashemi J, Oraee-Yazdani S, Akbari A. Down-regulation of 14-3-3 zeta sensitizes human glioblastoma cells to apoptosis induction. Apoptosis 2019; 23:616-625. [PMID: 30101359 DOI: 10.1007/s10495-018-1476-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Strong 14-3-3 zeta protein expression plays an important role in tumorigenesis, including in the maintenance of cell growth, resistance increase, and the prevention of apoptosis. In this study, we focus on two targets: (1) the expression of 14-3-3 zeta in the different grades of human astrocytoma (II-IV), (2) suppression of 14-3-3 zeta protein expression in glioblastoma derived astrocytes by 14-3-3 zeta shRNA lentiviral particles. The tissues of human astrocytoma were provided from 30 patients (ten of each grade of astrocytoma). Control tissues were obtained from the peritumoral brain zone of those patients with glioblastoma. The protein and mRNA expression levels of each astrocytoma grade were assessed via western blotting and RT-PCR, respectively. Results indicated that 14-3-3 zeta was significantly expressed in glioblastoma multiforme (grade IV) and 14-3-3 zeta expression levels enhanced according to the increase of astrocytoma malignancy. In the cellular study for knock down of the 14-3-3 zeta protein, surgical biopsy of glioblastoma was used to isolate primary astrocyte. Astrocytes were transduced with 14-3-3 zeta shRNA or non-targeted shRNA lentiviral particles. Furthermore, reduction of the 14-3-3 zeta protein expression in the astrocytes evaluated through qRT-PCR and western blot after transduction of 14-3-3 zeta shRNA lentiviral particles. Moreover, apoptosis properties, including DNA fragmentation and ratio increase of Bax/Bcl-2 were observed in astrocytes following reduction of 14-3-3 zeta protein expression. Further observation indicated that the mitochondrial pathway through release of cytochorome c and caspase-3 activity was involved in the apoptosis induction. Hence, this study demonstrates a key role of the 14-3-3 zeta protein in tumorigenesis but also indicates that 14-3-3 zeta can be considered as a target for the astrocytoma treatment specially glioblastoma.
Collapse
Affiliation(s)
- Mansoureh Hashemi
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Shohada Tajrish Hospital, Functional Neurosurgery Research Center, Shahrdari St, Tajrish Square, Tehran, 1989934148, Iran.
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Hashemi
- Department of Chemical Engineering, University of Louisville, Louisville, KY40292, USA
| | - Saeed Oraee-Yazdani
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akhtar Akbari
- Functional Neurosurgery Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Quantitative proteomics analysis of differentially expressed proteins in activated B-cell-like diffuse large B-cell lymphoma using quantitative proteomics. Pathol Res Pract 2019; 215:152528. [DOI: 10.1016/j.prp.2019.152528] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/10/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022]
|
42
|
Chi J, Liu T, Shi C, Luo H, Wu Z, Xiong B, Liu S, Zeng Y. Long non-coding RNA LUCAT1 promotes proliferation and invasion in gastric cancer by regulating miR-134-5p/YWHAZ axis. Biomed Pharmacother 2019; 118:109201. [PMID: 31545227 DOI: 10.1016/j.biopha.2019.109201] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/25/2019] [Accepted: 07/02/2019] [Indexed: 01/28/2023] Open
Abstract
PURPOSE The aim of this study was to research the function of lncRNA LUCAT1 in gastric cancer. METHODS Human gastric cancer tissues and paracancer tissues were obtained from 98 patients undergoing surgical resection in our hospital. The human gastric cancer cell lines (HGC27, BGC823, MGC803, SGC7901 and AGS), and normal gastric mucosal cell line GSE1 were used to research the role of lncRNA LUCAT1. ShRNAs specifically targeting lncRNA LUCAT1, miR-134-5p mimic, miR-134-5p inhibitor and their related controls were transfected into cells. Quantitative real-time PCR was used to detect the expression of lncRNA LUCAT1, miR-134-5p and YWHAZ. The cell proliferation of SGC7901 cells was determined by CCK8 kit. Colony formation assay was undertaken. Cell apoptosis assay was processed using the Annexin V-FITC / propidium iodide (annxinV/PI) apoptosis detection kit. Migration and invasion were detected by transwell assay. Tumor xenograft model was conducted to calculate the size and weight of the tumors. Luciferase reporter assay was used to confirm the interactions among lncRNA LUCAT1, miR-134-5p and YWHAZ. RESULTS LncRNA LUCAT1 was confirmed to be highly expressed in gastric cancer. Patients with high LUCAT1 level displayed short overall survival and disease-free survival periods. LUCAT1 knockdown or miR-134-5p overexpression decreased the proliferation, colony formation, migration and invasion of SGC7901 cells. CONCLUSIONS LncRNA LUCAT1 could promote proliferation and invasion of gastric cancer by regulating miR-134-5p/YWHAZ axis.
Collapse
Affiliation(s)
- Junlin Chi
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Tonglei Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Chengmin Shi
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Huayou Luo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Zhizhong Wu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Binghong Xiong
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Shuang Liu
- Department of Ultrasound, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Yujian Zeng
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
43
|
Hernández-Camarero P, López-Ruiz E, Griñán-Lisón C, García MÁ, Chocarro-Wrona C, Marchal JA, Kenyon J, Perán M. Pancreatic (pro)enzymes treatment suppresses BXPC-3 pancreatic Cancer Stem Cell subpopulation and impairs tumour engrafting. Sci Rep 2019; 9:11359. [PMID: 31388092 PMCID: PMC6684636 DOI: 10.1038/s41598-019-47837-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Cancer stem cells (CSCs) subpopulation within the tumour is responsible for metastasis and cancer relapse. Here we investigate in vitro and in vivo the effects of a pancreatic (pro)enzyme mixture composed of Chymotrypsinogen and Trypsinogen (PRP) on CSCs derived from a human pancreatic cell line, BxPC3. Exposure of pancreatic CSCs spheres to PRP resulted in a significant decrease of ALDEFLUOR and specific pancreatic CSC markers (CD 326, CD 44 and CxCR4) signal tested by flow cytometry, further CSCs markers expression was also analyzed by western and immunofluorescence assays. PRP also inhibits primary and secondary sphere formation. Three RT2 Profiler PCR Arrays were used to study gene expression regulation after PRP treatment and resulted in, (i) epithelial-mesenchymal transition (EMT) inhibition; (ii) CSCs related genes suppression; (iii) enhanced expression of tumour suppressor genes; (iv) downregulation of migration and metastasis genes and (v) regulation of MAP Kinase Signalling Pathway. Finally, in vivo anti-tumor xenograft studies demonstrated high anti-tumour efficacy of PRP against tumours induced by BxPC3 human pancreatic CSCs. PRP impaired engrafting of pancreatic CSC’s tumours in nude mice and displayed an antigrowth effect toward initiated xenografts. We concluded that (pro)enzymes treatment is a valuable strategy to suppress the CSC population in solid pancreatic tumours.
Collapse
Affiliation(s)
- Pablo Hernández-Camarero
- Department of Health Sciences, University of Jaén, Jaén, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Elena López-Ruiz
- Department of Health Sciences, University of Jaén, Jaén, Spain.,Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - María Ángel García
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Biochemistry and Molecular Biology 3 and Immunology, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Carlos Chocarro-Wrona
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Julian Kenyon
- The Dove Clinic for Integrated Medicine, Twyford, SO21 1RG, UK.
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain. .,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain.
| |
Collapse
|
44
|
Wen M, Qu X, Zhu Y, Tian L, Shen Z, Yang X, Shi X. Exploration of Novel Biomarkers in Vasculitis by Integrated Bioinfomatic Approaches. J Comput Biol 2019; 26:1448-1457. [PMID: 31356108 DOI: 10.1089/cmb.2019.0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Angiitis, also known as vasculitis, is a chronic inflammatory disease characterized by the infiltration of inflammatory cells in surroundings of blood vessels, accompanied by vascular damage including fibrin deposition, collagen fiber degeneration, myocyte, and endotheliocyte necrosis. This work aimed to perform an integrated bioinformatic analysis of three data sets concerning vasculitis to explore and examine the potential diagnostic and therapeutic makers contributing to illuminating the pathomechanisms of vasculitis. We collected three sets of gene expression data designed by dual-channel method from Gene Expression Omnibus, which were based on the same platform (Agilent-014850 Whole Human Genome Microarray 4x44K G4112F). The meta-analysis was used to analyze the gene expression profiles and screen the differentially expressed genes followed by functional features identification. Subsequently, a protein-protein interaction and transcriptional regulation network were conducted for further investigation of expression mechanisms of vasculitis. Totally, 73 consistently upregulated genes, 49 consistently downregulated genes, and 26 genes with different expression directions were identified. Functional enrichment and transcription regulation analysis suggested upregulated genes (PPBP, PLAU, and HIST1H2BH) and downregulated genes such as IL23A gene were predominately associated with immune responses and cytokine receptors function. In addition, specific cancer-related genes such as MRVI1 was also extracted and considered as promising biomarkers of the development and progression of vasculitis. This study established an integrated meta-analysis approach and identified novel biomarkers involved in vasculitis, which further facilitate to explore and unravel the etiopathogenesis of vasculitis.
Collapse
Affiliation(s)
- Mingxiang Wen
- Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xiang Qu
- Department of Emergency and Guizhou Provincial People's Hospital, Guiyang, China
| | - Yanyan Zhu
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Lingfang Tian
- Department of Endocrine, The First People's Hospital of Guiyang, Guiyang, China
| | - Zhongqin Shen
- Department of Endocrine, The First People's Hospital of Guiyang, Guiyang, China
| | - Xiulin Yang
- Department of Emergency and Guizhou Provincial People's Hospital, Guiyang, China
| | - Xianqing Shi
- Intensive Care Unit, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
45
|
Fan X, Cui L, Zeng Y, Song W, Gaur U, Yang M. 14-3-3 Proteins Are on the Crossroads of Cancer, Aging, and Age-Related Neurodegenerative Disease. Int J Mol Sci 2019; 20:ijms20143518. [PMID: 31323761 PMCID: PMC6678932 DOI: 10.3390/ijms20143518] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
14-3-3 proteins are a family of conserved regulatory adaptor molecules which are expressed in all eukaryotic cells. These proteins participate in a variety of intracellular processes by recognizing specific phosphorylation motifs and interacting with hundreds of target proteins. Also, 14-3-3 proteins act as molecular chaperones, preventing the aggregation of unfolded proteins under conditions of cellular stress. Furthermore, 14-3-3 proteins have been shown to have similar expression patterns in tumors, aging, and neurodegenerative diseases. Therefore, we put forward the idea that the adaptor activity and chaperone-like activity of 14-3-3 proteins might play a substantial role in the above-mentioned conditions. Interestingly, 14-3-3 proteins are considered to be standing at the crossroads of cancer, aging, and age-related neurodegenerative diseases. There are great possibilities to improve the above-mentioned diseases and conditions through intervention in the activity of the 14-3-3 protein family.
Collapse
Affiliation(s)
- Xiaolan Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Lang Cui
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Yao Zeng
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Wenhao Song
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Uma Gaur
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
46
|
Enguita FJ. New promising circulating RNA biomarkers for early diagnosis of lung adenocarcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S130. [PMID: 31576337 PMCID: PMC6685864 DOI: 10.21037/atm.2019.05.70] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/27/2019] [Indexed: 01/22/2023]
Affiliation(s)
- Francisco J. Enguita
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Cardiomics Unit, Centro de Cardiologia da Universidade de Lisboa (CCUL), Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
47
|
Liu J, Song S, Lin S, Zhang M, Du Y, Zhang D, Xu W, Wang H. Circ-SERPINE2 promotes the development of gastric carcinoma by sponging miR-375 and modulating YWHAZ. Cell Prolif 2019; 52:e12648. [PMID: 31199037 PMCID: PMC6668981 DOI: 10.1111/cpr.12648] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/30/2019] [Accepted: 05/13/2019] [Indexed: 12/30/2022] Open
Abstract
Objectives Circular RNAs (circRNAs) exist extensively in the eukaryotic genome. The study aimed to identify the role of hsa_circ_0008365 (Circ‐SERPINE2) in gastric carcinoma (GC) cells and its downstream mechanisms. Materials and methods Gene Expression Omnibus (GEO) database was applied to screen differentially expressed circRNAs. CircInteractome, TargetScan and miRecords websites were used to predict target relationships. qRT‐PCR and RNase R treatment were utilised to detect molecule expression and confirm the existence of circ‐SERPINE2. RNA pull‐down assay and dual‐luciferase reporter assay were performed for interaction between circRNA and miRNA or mRNA. EdU assay, colony formation assay, and flow cytometry for apoptosis and cell cycle detections were utilised to assess cell function. Western blot and immunohistochemistry (IHC) assays were applied for detection of proteins in tissues or cells. Results Circ‐SERPINE2 and YWHAZ were upregulated, and miR‐375 was downregulated in GC tissues and cells. Circ‐SERPINE2 and YWHAZ targetedly bound to miR‐375. Circ‐SERPINE2 promoted cell proliferation and cell cycle progress and inhibited cell apoptosis by sponging miR‐375 and regulating YWHAZ expression in vitro. Circ‐SERPINE2 repressed solid tumour growth through enhancing miR‐375 expression and reducing YWHAZ expression in vivo. Conclusions Circ‐SERPINE2 is a novel proliferative promoter through the regulation of miR‐375/YWHAZ. Circ‐SERPINE2/miR‐375/YWHAZ axis might provide a novel therapeutic target of GC.
Collapse
Affiliation(s)
- Jianing Liu
- Department of Thyroid and Pancreatic Disease, The Second Hospital of Shandong University, Jinan, China
| | - Suzhen Song
- Department of Internal Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Sen Lin
- Department of Digestive Disease, The Second Hospital of Shandong University, Jinan, China
| | - Mingbao Zhang
- Department of Digestive Disease, The Second Hospital of Shandong University, Jinan, China
| | - Yating Du
- Department of Digestive Disease, The Second Hospital of Shandong University, Jinan, China
| | - Dongdong Zhang
- Department of Digestive Disease, The Second Hospital of Shandong University, Jinan, China
| | - Weihua Xu
- Department of Digestive Disease, The Second Hospital of Shandong University, Jinan, China
| | - Hongbo Wang
- Department of Digestive Disease, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
48
|
Analysis of the Antiproliferative Effect of Ankaferd Hemostat on Caco-2 Colon Cancer Cells via LC/MS Shotgun Proteomics Approach. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5268031. [PMID: 31240215 PMCID: PMC6556321 DOI: 10.1155/2019/5268031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/20/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022]
Abstract
Ankaferd hemostat (ABS), a traditional herbal extract, is a hemostatic agent used for wound healing and bleeding treatment. A standardized form of plants contains many biomolecules. In recent years, previous studies have demonstrated the antineoplastic effect of ABS. In the present work, we focused on the mechanism of its antineoplastic effect over Caco-2 colon cancer cells. The LC/MS-based proteomics method was used to understand the effect of ABS at the protein level. The results were evaluated with gene ontology, protein interaction, and pathway analysis. As shown by our results, ABS altered glucose, fatty acids, and protein metabolism. Moreover, ABS affects the cell cycle machinery. Moreover, we found that ABS induced critical cancer target and suppressor proteins such as carboxyl-terminal hydrolase 1, 60S ribosomal protein L5, Tumor protein D52-like2, karyopherin alpha 2, and protein deglycase DJ-1. In conclusion, the proteomics results indicated that ABS affects various cancer targets and suppressor proteins. Moreover ABS has systematical effect on cell metabolism and cell cycle in Caco-2 cells, suggesting that it could be used as an antineoplastic agent.
Collapse
|
49
|
Down-regulation of 14-3-3zeta reduces proliferation and
increases apoptosis in human glioblastoma. Cancer Gene Ther 2019; 27:399-411. [DOI: 10.1038/s41417-019-0097-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/08/2019] [Accepted: 04/13/2019] [Indexed: 11/08/2022]
|
50
|
Qin J, Wang S, Wang P, Wang X, Ye H, Song C, Dai L, Wang K, Jiang B, Zhang J. Autoantibody against 14-3-3 zeta: a serological marker in detection of gastric cancer. J Cancer Res Clin Oncol 2019; 145:1253-1262. [PMID: 30887154 DOI: 10.1007/s00432-019-02884-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Autoantibody to 14-3-3 zeta was identified in gastric cancer (GC) by serological proteome analysis (SERPA) in our previous study. We comprehensively evaluated its ability to detect GC, determined its association with clinical characteristics, and explored its temporal change in GC patients before and after gastrectomy resection in this study. METHODS Anti-14-3-3 zeta antibody was examined by immunoassay in sera from 465 GC patients and 465 normal individuals, and also in 69 serial sera from 26 GC patients before and after resection. RESULTS The frequency of anti-14-3-3 zeta were significantly higher in GC group than in control group, with AUC of 0.627. The appearance of anti-14-3-3 zeta showed no difference in different tumor stage, tumor size, tumor differentiation, and lymphatic metastasis, but was higher in GC patients with family tumor history than without family tumor history. When anti-14-3-3 zeta was combined with clinical markers (CEA, CA199 and CA724), the sensitivity increased to 52.7%. In the follow-up analysis, the titer of anti-14-3-3 zeta was higher in post-resection sera than pre-resection sera, and no difference was observed in CEA, CA199 and CA724. Anti-14-3-3 zeta showed an increase from negative status to positive status in six patients after resection, while other three clinical markers presented different change in GC patients after resection. CONCLUSIONS Autoantibody against 14-3-3 zeta could be a potential diagnostic biomarker and improve the sensitivity of CEA, CA199 and CA724 in diagnosis of GC. Further largescale studies will be needed to validate its performance in GC patients after resection.
Collapse
Affiliation(s)
- Jiejie Qin
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | - Shuaibing Wang
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
- Third Affiliated Hospital of Zhengzhou University, 23 Kangfu Road, Zhengzhou, 450052, China
| | - Peng Wang
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | - Xiao Wang
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, 40 N Daxue Road, Zhengzhou, Henan, China
| | - Hua Ye
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | - Chunhua Song
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | - Liping Dai
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, 40 N Daxue Road, Zhengzhou, Henan, China
| | - Kaijuan Wang
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China
| | - Binghua Jiang
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, 40 N Daxue Road, Zhengzhou, Henan, China
| | - Jianying Zhang
- Department of Epidemiology and Health statistics, Henan Key Laboratory for Tumor Epidemiology, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, China.
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, 40 N Daxue Road, Zhengzhou, Henan, China.
| |
Collapse
|