1
|
Zhao W, Qi C, Mao Y, Ye F, Xia T, Zhao M, Min P, Zhang Y, Du J. High MICAL-L2 promotes cancer progression and drug resistance in renal clear cell carcinoma cells through stabilization of ACTN4 following vimentin expression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167628. [PMID: 39689763 DOI: 10.1016/j.bbadis.2024.167628] [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: 08/27/2024] [Revised: 11/18/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
Targeted therapies persist as the conventional method of treatment of kidney clear cell carcinoma (KIRC). However, resistance to these drugs emerges as a significant impediment to the management of renal cancer. MICAL-L2 plays a pivotal role in cytoskeleton rearrangement. This study sought to elucidate the clinical relevance of MICAL-L2 in KIRC and its regulatory mechanism driving cancer progression and resistance to therapy. TCGA data mining was utilized to assess the expression of MICAL-L2 in samples from patients with KIRC. Kaplan-Meier analysis and immunohistochemistry were employed to explore the clinical significance of MICAL-L2. In vitro experiments, including assays for wound healing and Transwell migration, CCK-8, EDU staining, RT-PCR, flow cytometry, and co-immunoprecipitation analysis were conducted to investigate the effects of MICAL-L2 on the drug sensitivity of KIRC cells and to elucidate the molecular mechanisms involved. The results showed that MICAL-L2 was overexpressed in KIRC tissues. High levels of MICAL-L2 were associated with poor survival and a poor response to drug therapy among patients with KIRC. Overexpression of MICAL-L2 stimulated cell migration, proliferation, and rendered KIRC cells insensitive to sunitinib and everolimus, two traditional therapies for KIRC. Furthermore, MICAL-L2 overexpression accelerated cancer progression and resistance to therapy in KIRC cells by interacting with its downstream regulator α-actinin-4 (ACTN4) in a Rab13-dependent manner, which reduced the degradation of ACTN4, leading to increased Vimentin expression. All these findings indicate that MICAL-L2 plays a crucial role in the progression of KIRC and suggest that MICAL-L2 may serve as a potential therapeutic target for KIRC treatment.
Collapse
Affiliation(s)
- Weizhen Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chenxiang Qi
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yixin Mao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Fengwen Ye
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Tianxiang Xia
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Mingyu Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Pengxiang Min
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
2
|
Wen P, Li J, Wen Z, Guo X, Ma G, Hu S, Xu J, Zhao H, Li R, Liu Y, Wang Y, Gao J. MICAL-L2, as an estrogen-responsive gene, is involved in ER-positive breast cancer cell progression and tamoxifen sensitivity via the AKT/mTOR pathway. Biochem Pharmacol 2024; 225:116256. [PMID: 38729448 DOI: 10.1016/j.bcp.2024.116256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/25/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Endocrine treatment, particularly tamoxifen, has shown significant improvement in the prognosis of patients with estrogen receptor-positive (ER-positive) breast cancer. However, the clinical utility of this treatment is often hindered by the development of endocrine resistance. Therefore, a comprehensive understanding of the underlying mechanisms driving ER-positive breast cancer carcinogenesis and endocrine resistance is crucial to overcome this clinical challenge. In this study, we investigated the expression of MICAL-L2 in ER-positive breast cancer and its impact on patient prognosis. We observed a significant upregulation of MICAL-L2 expression in ER-positive breast cancer, which correlated with a poorer prognosis in these patients. Furthermore, we found that estrogen-ERβ signaling promoted the expression of MICAL-L2. Functionally, our study demonstrated that MICAL-L2 not only played an oncogenic role in ER-positive breast cancer tumorigenesis but also influenced the sensitivity of ER-positive breast cancer cells to tamoxifen. Mechanistically, as an estrogen-responsive gene, MICAL-L2 facilitated the activation of the AKT/mTOR signaling pathway in ER-positive breast cancer cells. Collectively, our findings suggest that MICAL-L2 could serve as a potential prognostic marker for ER-positive breast cancer and represent a promising molecular target for improving endocrine treatment and developing therapeutic approaches for this subtype of breast cancer.
Collapse
Affiliation(s)
- Pushuai Wen
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China; Biological Anthropology Institute, Jinzhou Medical University, Jinzhou 121001, China.
| | - Jing Li
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, China
| | - Zihao Wen
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Xiaoyan Guo
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, China
| | - Guoqun Ma
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Shuzhen Hu
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Jiamei Xu
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Hongli Zhao
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Ruixin Li
- Department of Pathophysiology, Jinzhou Medical University, Jinzhou 121001, China
| | - Ying Liu
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, China.
| | - Yu Wang
- Liaoning Technology and Engineering Center for Tumor Immunology and Molecular Theranotics, Collaborative Innovation Center for Age-related Disease, Life Science Institute of Jinzhou Medical University, Jinzhou 121001, China.
| | - Jing Gao
- Department of Ultrasonography, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121001, China.
| |
Collapse
|
3
|
Min P, Li Y, Wang C, Fan J, Liu S, Chen X, Tang Y, Han F, Zhang A, Feng L. Cyclopeptide moroidin inhibits vasculogenic mimicry formed by glioblastoma cells via regulating β-catenin activation and EMT pathways. J Biomed Res 2024; 38:322-333. [PMID: 38807414 PMCID: PMC11300521 DOI: 10.7555/jbr.38.20240015] [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: 01/19/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024] Open
Abstract
Glioblastoma (GBM) is a highly vascularized malignant brain tumor with poor clinical outcomes. Vasculogenic mimicry (VM) formed by aggressive GBM cells is an alternative approach for tumor blood supply and contributes to the failure of anti-angiogenic therapy. To date, there is still a lack of effective drugs that target VM formation in GBM. In the present study, we evaluated the effects of the plant cyclopeptide moroidin on VM formed by GBM cells and investigated its underlying molecular mechanisms. Moroidin significantly suppressed cell migration, tube formation, and the expression levels of α-smooth muscle actin and matrix metalloproteinase-9 in human GBM cell lines at sublethal concentrations. The RNA sequencing data suggested the involvement of the epithelial-mesenchymal transition (EMT) pathway in the mechanism of moroidin. Exposure to moroidin led to a concentration-dependent decrease in the expression levels of the EMT markers N-cadherin and vimentin in GBM cells. Moreover, moroidin significantly reduced the level of phosphorylated extracellular signal-regulated protein kinase (p-ERK) and inhibited the activation of β-catenin. Finally, we demonstrated that the plant cyclopeptide moroidin inhibited VM formation by GBM cells through inhibiting the ERK/β-catenin-mediated EMT. Therefore, our study indicates a potential application of moroidin as an anti-VM agent in the treatment of GBM.
Collapse
Affiliation(s)
- Pengxiang Min
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingying Li
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Cuirong Wang
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Junting Fan
- Department of Pharmaceutical Analysis, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shangming Liu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiang Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yamin Tang
- Department of Analysis and Testing Center, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Aixia Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lili Feng
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
4
|
Wu X, Cao C, Li Z, Xie Y, Zhang S, Sun W, Guo J. Circular RNA CircSLC22A23 Promotes Gastric Cancer Progression by Activating HNRNPU Expression. Dig Dis Sci 2024; 69:1200-1213. [PMID: 38400886 DOI: 10.1007/s10620-024-08291-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/09/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND Circular RNAs (CircRNAs) play essential roles in cancer occurrence as regulatory RNAs. However, circRNA-mediated regulation of gastric cancer (GC) remains poorly understood. AIM The purpose of this study was to investigate the molecular mechanism of circSLC22A23 (hsa_circ_0075504) underlying GC occurrence. METHODS CircSLC22A23 levels were first quantified by quantitative real-time reverse transcription-polymerase chain reaction in GC cell lines, 80 paired GC tissues and adjacent normal tissues, and 27 pairs of plasma samples from preoperative and postoperative patients with GC. Then circSLC22A23 was knocked-down with short hairpin RNA to analyze its oncogenic effects on the proliferation, migration, and invasion of GC cells. Finally, circRNA-binding proteins and their downstream target genes were identified by RNA pulldown, mass spectrometry, RNA immunoprecipitation, quantitative real-time reverse transcription-polymerase chain reaction, and Western blot assays. RESULTS CircSLC22A23 was found to be highly expressed in GC cells, GC tissues, and plasma from GC patients. Knockdown of circSLC22A23 inhibited GC cell proliferation, migration and invasion. RNA pulldown and RNA immunoprecipitation assays verified the interaction between circSLC22A23 and heterogeneous nuclear ribonucleoprotein U (HNRNPU). Knockdown of circSLC22A23 decreased HNRNPU protein levels. Moreover, rescue assays showed that the tumor suppressive effect of circSLC22A23 knockdown was reversed by HNRNPU overexpression. Finally, epidermal growth factor receptor (EGFR) was found to be one of the downstream target genes of HNRNPU that was up regulated by circSLC22A23. CONCLUSION CircSLC22A23 regulated the transcription of EGFR through activation of HNRNPU in GC cells, suggesting that circSLC22A23 may serve as a potential therapeutic target for the treatment of GC.
Collapse
Affiliation(s)
- Xinxin Wu
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The Affiliated No. 1 Hospital, Ningbo University, Ningbo, 315211, China
| | - Chunli Cao
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated People's Hospital, Ningbo University, Ningbo, 315040, China
| | - Zhe Li
- Department of Gastroenterology, The Affiliated No. 1 Hospital, Ningbo University, Ningbo, 315211, China
| | - Yaoyao Xie
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The Affiliated No. 1 Hospital, Ningbo University, Ningbo, 315211, China
| | - Shuangshuang Zhang
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Department of Gastroenterology, The Affiliated No. 1 Hospital, Ningbo University, Ningbo, 315211, China
| | - Weiliang Sun
- The Affiliated People's Hospital, Ningbo University, Ningbo, 315040, China
| | - Junming Guo
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China.
- Department of Gastroenterology, The Affiliated No. 1 Hospital, Ningbo University, Ningbo, 315211, China.
- Institute of Digestive Diseases of Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
5
|
Ye F, Xia T, Zhao M, Zhao W, Min P, Wang Y, Wang Q, Zhang Y, Du J. PlexinA1 promotes gastric cancer migration through preventing MICAL1 protein ubiquitin/proteasome-mediated degradation in a Rac1-dependent manner. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167124. [PMID: 38508474 DOI: 10.1016/j.bbadis.2024.167124] [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: 12/23/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024]
Abstract
Metastasis promotes the development of tumors and is a significant cause of gastric cancer death. For metastasis to proceed, tumor cells must become mobile by modulating their cytoskeleton. MICAL1 (Molecule Interacting with CasL1) is known as an actin cytoskeleton regulator, but the mechanisms by which it drives gastric cancer cell migration are still unclear. Analysis of gastric cancer tissues revealed that MICAL1 expression is dramatically upregulated in stomach adenocarcinoma (STAD) samples as compared to noncancerous stomach tissues. Patients with high MICAL1 expression had shorter overall survival (OS), post-progression survival (PPS) and first-progression survival (FPS) compared with patients with low MICAL1 expression. RNAi-mediated silencing of MICAL1 inhibited the expression of Vimentin, a protein involved in epithelial-mesenchymal transition. This effect correlates with a significant reduction in gastric cancer cell migration. MICAL1 overexpression reversed these preventive effects. Immunoprecipitation experiments and immunofluorescence assays revealed that PlexinA1 forms a complex with MICAL1. Importantly, specific inhibition of PlexinA1 blocked the Rac1 activation and ROS production, which, in turn, impaired MICAL1 protein stability by accelerating MICAL1 ubiquitin/proteasome-dependent degradation. Overexpression of PlexinA1 enhanced Rac1 activation, ROS production, MICAL1 and Vimentin expressions, and favored cell migration. In conclusion, this study identified MICAL1 as an important facilitator of gastric cancer cell migration, at least in part, by affecting Vimentin expression and PlexinA1 promotes gastric cancer cell migration by binding to and suppressing MICAL1 degradation in a Rac1/ROS-dependent manner.
Collapse
Affiliation(s)
- Fengwen Ye
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Tianxiang Xia
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - MingYu Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Weizhen Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Pengxiang Min
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yueyuan Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qianwen Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
6
|
Rasouli M, Safari F, Sobhani N, Alavi M, Roudi R. Regulation of Cellular-Signaling Pathways by Mammalian Proteins Containing Bacterial EPIYA or EPIYA-Like Motifs Predicted to be Phosphorylated. DNA Cell Biol 2024; 43:74-84. [PMID: 38153368 DOI: 10.1089/dna.2023.0350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023] Open
Abstract
The effector proteins of several pathogenic bacteria contain the Glu-Pro-Ile-Tyr-Ala (EPIYA) motif or other similar motifs. The EPIYA motif is delivered into the host cells by type III and IV secretion systems, through which its tyrosine residue undergoes phosphorylation by host kinases. These motifs atypically interact with a wide range of Src homology 2 (SH2) domain-containing mammalian proteins through tyrosine phosphorylation, which leads to the perturbation of multiple signaling cascades, the spread of infection, and improved bacterial colonization. Interestingly, it has been reported that EPIYA (or EPIYA-like) motifs exist in mammalian proteomes and regulate mammalian cellular-signaling pathways, leading to homeostasis and disease pathophysiology. It is possible that pathogenic bacteria have exploited EPIYA (or EPIYA-like) motifs from mammalian proteins and that the mammalian EPIYA (or EPIYA-like) motifs have evolved to have highly specific interactions with SH2 domain-containing proteins. In this review, we focus on the regulation of mammalian cellular-signaling pathways by mammalian proteins containing these motifs.
Collapse
Affiliation(s)
- Mohammad Rasouli
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mana Alavi
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, California, USA
| |
Collapse
|
7
|
Xia T, Ye F, Zhao W, Min P, Qi C, Wang Q, Zhao M, Zhang Y, Du J. Comprehensive Analysis of MICALL2 Reveals Its Potential Roles in EGFR Stabilization and Ovarian Cancer Cell Invasion. Int J Mol Sci 2023; 25:518. [PMID: 38203692 PMCID: PMC10778810 DOI: 10.3390/ijms25010518] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Molecules interacting with CasL (MICALs) are critical mediators of cell motility that act by cytoskeleton rearrangement. However, the molecular mechanisms underlying the regulation of cancer cell invasion remain elusive. The aim of this study was to investigate the potential role of one member of MICALs, i.e., MICALL2, in the invasion and function of ovarian cancer cells. We showed by bioinformatics analysis that MICALL2 expression was significantly higher in tissues of advanced-stage ovarian cancer and associated with poor overall survival of patients. MICALL2 was strongly correlated with the infiltration of multiple types of immune cells and T-cell exhaustion markers. Moreover, enrichment analyses showed that MICALL2 was involved in the tumor-related matrix degradation pathway. Mechanistically, MMP9 was identified as the target gene of MICALL2 for the regulation of invadopodium formation and SKOV3, HO-8910PM cell invasion. In addition, EGFR-AKT-mTOR signaling was identified as the downstream pathway of MICALL2 in the regulation of MMP9 expression. Furthermore, MICALL2 silencing promoted EGFR degradation; however, this effect was abrogated by treatment with the autophagy inhibitors acadesine and chloroquine diphosphate. Silencing of MICALL2 resulted in a suppressive activity of Rac1 while suppressing Rac1 activation attenuated the pro-EGFR, pro-MMP9, and proinvasive effects induced by the overexpression of MICALL2. Collectively, our results indicated that MICALL2 participated in the process of immune infiltration and invasion by ovarian cancer cells. Moreover, MICALL2 prevented EGFR degradation in a Rac1-dependent manner, consequently leading to EGFR-AKT-mTOR-MMP9 signaling activation and invadopodia-mediated matrix degradation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China; (T.X.); (F.Y.); (W.Z.); (P.M.); (C.Q.); (Q.W.); (M.Z.); (Y.Z.)
| |
Collapse
|
8
|
Chen Y, Song W, Zhang H, Ji X. MICALL2 participates in the regulation of epithelial-mesenchymal transition in alveolar epithelial cells - Potential roles in pulmonary fibrosis. Arch Biochem Biophys 2023; 747:109730. [PMID: 37690696 DOI: 10.1016/j.abb.2023.109730] [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: 06/13/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023]
Abstract
Epithelial-mesenchymal transition (EMT) of alveolar epithelial cells is a vital process in idiopathic pulmonary fibrosis (IPF), which results in the accumulation of fibroblasts and myofibroblasts and excessive extracellular matrix deposition. Based on RNA sequencing analysis and GEO dataset reanalysis, we screened out MICALL2, a gene upregulated in the lungs of IPF mice and alveolar epithelial type II (ATII) cells from IPF patients, and aimed to explore its role in IPF. We validated the expression of MICALL2 in bleomycin (BLM)-induced IPF mice and TGF-β1-stimulated ATII cells (primary murine ATII cells and A549 cells), and explored the role of MICALL2 in IPF by knockdown of MICALL2 in BLM-induced mice and TGF-β1-stimulated ATII cells. We found that MICALL2 was upregulated in the lungs of BLM-induced mice and TGF-β1-stimulated ATII cells. MICALL2-deficient mice had reduced fibrogenesis and restrained EMT upon BLM challenge. Knockdown of MICALL2 restrained the EMT process, in vitro, through impeding β-catenin nuclear translocation. Mechanistically, we demonstrated that NPAS2 is directly bound to the promoter of MICALL2. Altogether, our data revealed transactivation of MICALL2 induced by NPAS2, contributing to activation of the Wnt/β-catenin pathway in ATII cells, thus leading to the EMT process and subsequent pulmonary fibrosis. Interfering with MICALL2 may represent an innovative therapeutic target to mitigate the extent of IPF.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Wei Song
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - He Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Xinping Ji
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
9
|
Schiavoni V, Campagna R, Pozzi V, Cecati M, Milanese G, Sartini D, Salvolini E, Galosi AB, Emanuelli M. Recent Advances in the Management of Clear Cell Renal Cell Carcinoma: Novel Biomarkers and Targeted Therapies. Cancers (Basel) 2023; 15:3207. [PMID: 37370817 PMCID: PMC10296504 DOI: 10.3390/cancers15123207] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Renal cell carcinoma (RCC) belongs to a heterogenous cancer group arising from renal tubular epithelial cells. Among RCC subtypes, clear cell renal cell carcinoma (ccRCC) is the most common variant, characterized by high aggressiveness, invasiveness and metastatic potential, features that lead to poor prognosis and high mortality rate. In addition, diagnosis of kidney cancer is incidental in the majority of cases, and this results in a late diagnosis, when the stage of the disease is advanced and the tumor has already metastasized. Furthermore, ccRCC treatment is complicated by its strong resistance to chemo- and radiotherapy. Therefore, there is active ongoing research focused on identifying novel biomarkers which could be useful for assessing a better prognosis, as well as new molecules which could be used for targeted therapy. In this light, several novel targeted therapies have been shown to be effective in prolonging the overall survival of ccRCC patients. Thus, the aim of this review is to analyze the actual state-of-the-art on ccRCC diagnosis, prognosis and therapeutic options, while also reporting the recent advances in novel biomarker discoveries, which could be exploited for a better prognosis or for targeted therapy.
Collapse
Affiliation(s)
- Valentina Schiavoni
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Monia Cecati
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Giulio Milanese
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Andrea Benedetto Galosi
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60020 Ancona, Italy; (V.S.); (R.C.); (V.P.); (M.C.); (G.M.); (A.B.G.); (M.E.)
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, 60131 Ancona, Italy
| |
Collapse
|
10
|
Yang Y, Zhao W, Wang Y, Du J. Prognostic impact of MICALL1 and associates with immune infiltration in liver hepatocellular carcinoma patients. Cancer Biomark 2023:CBM220370. [PMID: 37248888 DOI: 10.3233/cbm-220370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
BACKGROUND Liver hepatocellular carcinoma (LIHC) is one of the most malignancy over the world. Previous studies have proven that Molecules Interacting with CasL-Like 1 (MICALL1) participated in cellular trafficking cascades, while there has no study to explore the function and carcinogenic mechanism MICALL1 in LIHC. METHODS We aimed to investigate the relationship between MICALL1 mRNA expression and LIHC using TCGA database. The expression of MICALL1 protein in clinic samples were examined by UALCAN database. Kaplan-Meier method was used for survival analysis. Logistic regression and Cox regression were performed to evaluate the prognostic significance of MICALL1. The MICALL1-binding protein were built by the STRING tool. Enrichment analysis by GO, KEGG and GSEA was used to explore possible function of MICALL1. The ssGSEA method was used to investigate the association between MICALL1 expression and the immune infiltration level in LIHC. RESULTS The expression and prognostic value of different MICAL family members in LIHC were evaluated. The expression of MICALL1 was significantly increased at both the transcript and protein levels in LIHC tissues. Further, the LIHC patients with high MICALL1 levels showed a worse OS, DSS and PFI. Some clinicopathologic features were identified to be related to MICALL1 expression in LIHC included clinical T stage, pathologic stage, histologic grade and AFP concentration. Univariate and multivariate survival analysis showed that MICALL1 was an independent prognostic marker for OS and DSS. Further enrichment analysis revealed that the K-RAS, TNFα/NF-κB and inflammatory response were significantly enriched in the high MICALL1 expression group. Immune infiltration analysis showed that high MICALL1 expression was correlated with infiltration level of macrophage cells, Th2 cells and some other immune cell types, including TFH. CONCLUSIONS MICALL1 expression was significantly associated with immune cell infiltration and may regarded as a promising prognostic biomarker for LIHC patients.
Collapse
Affiliation(s)
- Yixing Yang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weizhen Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yueyuan Wang
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Zeng X, Wang H, Yang J, Hu J. Micall2 Is Responsible for the Malignancy of Clear Cell Renal Cell Carcinoma. Yonago Acta Med 2023; 66:171-179. [PMID: 36811029 PMCID: PMC9937966 DOI: 10.33160/yam.2023.02.021] [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: 05/17/2022] [Accepted: 09/05/2022] [Indexed: 02/21/2023]
Abstract
Background There lacks a sufficient research on the tumorigenesis of clear cell renal cell carcinoma (ccRCC), causing that the prognosis of ccRCC was not effectively improved. Micall2 contributes to the malignancy of cancer. Moreover, Micall2 is considered a typical cell mobility-promoting factor. However, the relationship between Micall2 and ccRCC malignancy is unknown. Methods In this study, we first investigated the expression patterns of Micall2 in ccRCC tissues and cell lines. Next, we explored the in vitro and in vivo roles of Micall2 in ccRCC tumorigenesis based on ccRCC cell lines with different Micall2 expression and gene manipulation assays. Results Our study showed that ccRCC tissues and cell lines expressed higher level of Micall2 than paracancerous tissues and normal renal tubular epithelial cell, and Micall2 expression was overexpressed on cancerous tissue with significant metastasis and enlargement. Among three ccRCC cell lines, the expression of Micall2 was the highest in 786-O cells and the lowest in CAKI-1 cells. Moreover, 786-O cells showed the highest malignancy in vitro and in vivo (including proliferation, migration, invasion, reduced E-cadherin expression and tumorigenicity of nude mice in vivo), while CAKI-1 cells showed the contrary results. Furthermore, the upregulated Micall2 by Gene overexpression promoted the proliferation, migration and invasion of ccRCC cells while the downregulated Micall2 by Gene silencing showed the opposite effect. Conclusion Micall2, as a pro-tumorigenic gene marker of ccRCC, contributes the malignancy of ccRCC.
Collapse
Affiliation(s)
- Xianyou Zeng
- Department of Urology, The Affiliated Hospital of Jinggangshan University, Jian, Jiangxi, China
| | - Hongquan Wang
- Department of Urology, Chongqing Fengdu people’s Hospital, Chongqing, China
| | - Jia Yang
- Department of Urology, Chongqing Fengdu people’s Hospital, Chongqing, China
| | - Jing Hu
- Department of Stomatology, The Affiliated Hospital of Jinggangshan University, Jian, Jiangxi, China
| |
Collapse
|
12
|
Yang Y, Ye F, Xia T, Wang Q, Du J. High MICAL1 expression correlates with cancer progression and immune infiltration in renal clear cell carcinoma. BMC Cancer 2022; 22:1355. [PMID: 36575439 PMCID: PMC9793553 DOI: 10.1186/s12885-022-10462-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Molecule interacting with CasL 1 (MICAL1), a multidomain flavoprotein monooxygenase, is strongly involved in the biological processes related to cancer cell proliferation and metastasis. However, there were few reports on the clinical significance of MICAL1 in renal clear cell carcinoma. METHODS The expression and prognostic value of MICAL1 in renal clear cell carcinoma were explored using immunohistochemical assays, public TCGA-KIRC databases and multiple analysis methods, including survival analysis, univariate and multivariate analyses, KEGG and GSEA. Wound healing and Transwell assays were performed to check the 786-O cell and Caki-1 cell migration abilities after knockdown of MICAL1. Western blotting was used to assess the regulatory effect of MICAL1 on the Rac1 activation. Additionally, the function of MICAL1 and the correlations between MICAL1 and immune infiltration levels in KIRC were investigated using TIMER and TISIDB. RESULTS MICAL1 expression was significantly higher in carcinoma tissue compared with non-cancerous tissue. A survival analysis revealed that patients with high MICAL1 expression had shorter overall survival (OS) and disease-specific survival (DSS) compared with patients with low MICAL1 expression. ROC analysis also confirmed that MICAL1 has a high diagnostic value in KIRC. Importantly, the univariate and multivariate Cox analysis further confirmed that high MICAL1 expression was an independent risk factor for OS in patients with KIRC. In accordance with this, knockdown of MICAL1 expression decreased Rac1 activation and cell migration. KEGG and GSEA analysis revealed that the immune infiltration and Ras signaling pathways were significantly upregulated in the high MICAL1 expression group. In terms of immune infiltrating levels, MICAL1 expression was positively associated with CD8+/Treg cell infiltration levels. Specifically, bioinformatic analysis showed that MICAL1 expression had strong relationships with various T cell exhaustion markers. CONCLUSIONS MICAL1 expression may act as a prognostic biomarker for determining the prognosis in renal clear cell carcinoma and plays an important role in regulating tumor immune microenvironment and cell migratory capacity.
Collapse
Affiliation(s)
- Yixing Yang
- grid.89957.3a0000 0000 9255 8984The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166 China
| | - Fengwen Ye
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Tianxiang Xia
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Qianwen Wang
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| | - Jun Du
- grid.89957.3a0000 0000 9255 8984Department of Physiology, Nanjing Medical University, Nanjing, 211166 China
| |
Collapse
|
13
|
Wen P, Wang H, Li Y, Sui X, Hou Z, Guo X, Xue W, Liu D, Wang Y, Gao J. MICALL2 as a substrate of ubiquitinase TRIM21 regulates tumorigenesis of colorectal cancer. Cell Commun Signal 2022; 20:170. [DOI: 10.1186/s12964-022-00984-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/01/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Molecule interacting with CasL-like protein 2 (MICALL2) is believed to regulate cytoskeleton dynamics, tight junction formation, and neurite outgrowth. However, its biological role and the underlying mechanism in colorectal cancer (CRC) remain largely elusive.
Methods
qRT-PCR, Western blotting and immunohistochemistry assays were used to detect the expression levels of different genes. Next, mass spectrometry, co-immunoprecipitation and immunofluorescence staining were used to detect the interactions of proteins. Furthermore, MTT assay, colony formation assay, wound-healing assays and xenograft tumor models were performed to demonstrate the functions of MICALL2 in CRC. In addition, transcriptome sequencing and Western blotting were conducted to verify the mechanism of MICALL2 in CRC.
Results
We found that both mRNA and protein levels of MICALL2 are up-regulated in colorectal cancer tissues compared with non-tumor tissues and that its overexpression is closely correlated with poor prognosis. Ubiquitin E3 ligase Tripartite motif-containing protein 21 (TRIM21) mediated MICALL2 ubiquitination and proteasome-dependent degradation, negatively correlated with MICALL2 levels, and reversely regulated the tumorigenic activity of MICALL2 in CRC. Functional studies confirmed that MICALL2 promoted colorectal cancer cell growth and migration via the Wnt/β-catenin signaling pathway.
Conclusions
As a substrate of ubiquitinase TRIM21, MICALL2 enhances the growth and migration of colorectal cancer cells and activates the Wnt/β-catenin signaling pathway.
Collapse
|
14
|
Wang Q, Qi C, Min P, Wang Y, Ye F, Xia T, Zhang Y, Du J. MICAL2 contributes to gastric cancer cell migration via Cdc42-dependent activation of E-cadherin/β-catenin signaling pathway. Cell Commun Signal 2022; 20:136. [PMID: 36064550 PMCID: PMC9442994 DOI: 10.1186/s12964-022-00952-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/06/2022] [Indexed: 11/10/2022] Open
Abstract
Background Gastric cancer is a common and lethal human malignancy worldwide and cancer cell metastasis is the leading cause of cancer-related mortality. MICAL2, a flavoprotein monooxygenase, is an important regulator of epithelial-to-mesenchymal transition. The aim of this study was to explore the effects of MICAL2 on gastric cancer cell migration and determine the underlying molecular mechanisms. Methods Cell migration was examined by wound healing and transwell assays. Changes in E-cadherin/β-catenin signaling were determined by qPCR and analysis of cytoplasmic and nuclear protein fractions. E-cadherin/β-catenin binding was determined by co-immunoprecipitation assays. Cdc42 activity was examined by pulldown assay. Results MICAL2 was highly expressed in gastric cancer tissues. The knockdown of MICAL2 significantly attenuated migratory ability and β-catenin nuclear translocation in gastric cancer cells while LiCl treatment, an inhibitor of GSK3β, reversed these MICAL2 knockdown-induced effects. Meanwhile, E-cadherin expression was markedly enhanced in MICAL2-depleted cells. MICAL2 knockdown led to a significant attenuation of E-cadherin ubiquitination and degradation in a Cdc42-dependent manner, then enhanced E-cadherin/β-catenin binding, and reduced β-catenin nuclear translocation. Conclusions Together, our results indicated that MICAL2 promotes E-cadherin ubiquitination and degradation, leading to enhanced β-catenin signaling via the disruption of the E-cadherin/β-catenin complex and, consequently, the promotion of gastric cell migration. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00952-x.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Chenxiang Qi
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Pengxiang Min
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yueyuan Wang
- Experimental Teaching Center of Basic Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Fengwen Ye
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Tianxiang Xia
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
15
|
Alharbi KS, Javed Shaikh MA, Afzal O, Alfawaz Altamimi AS, Almalki WH, Alzarea SI, Kazmi I, Al-Abbasi FA, Singh SK, Dua K, Gupta G. An overview of epithelial growth factor receptor (EGFR) inhibitors in cancer therapy. Chem Biol Interact 2022; 366:110108. [PMID: 36027944 DOI: 10.1016/j.cbi.2022.110108] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 12/28/2022]
Abstract
Epithelial growth factor receptor (EGFR), a transmembrane receptor on the cell surface, carries extracellular messages into the cell and alters the activity of the nucleus through tyrosine signalling. EGFR-targeted treatments have influenced the new era of precision oncology throughout the last few decades. Despite significant progress, long-term remission from solid tumours is still a distant goal for many oncologists. There are several methods by which tumour cells alter the activity of this protein in solid tumours. EGFR-related oncogenic pathways, resistance mechanisms, and novel avenues to suppress tumour development and metastatic spread were discovered in clinical specimens using preclinical models (cell cultures, xenografts, mouse models), which were then validated in those specimens. EGFR has been implicated in the onset and advancement of a variety of cancers, according to research. An overview of EGFR's structural anatomy and physiology, its role in cancers, and clinical studies that target EGFR in various tumours are included in this review.
Collapse
Affiliation(s)
- Khalid Saad Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | | | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | | | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, 2007, Australia.
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| |
Collapse
|
16
|
Yang Y, Ye F, Xia T, Wang Q, Zhang Y, Du J. High MICAL-L2 expression and its role in the prognosis of colon adenocarcinoma. BMC Cancer 2022; 22:487. [PMID: 35501725 PMCID: PMC9063352 DOI: 10.1186/s12885-022-09614-0] [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: 02/21/2022] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND MICAL-like protein 2 (MICAL-L2), a member of the molecules interacting with CasL (MICAL) family of proteins, is strongly associated with the malignancy of multiple types of cancer. However, the role of MICAL-L2 in colon adenocarcinoma (COAD) has not been well characterized. METHODS In this study, we analyzed the role of MICAL-L2 in COAD using datasets available from public databases. The mRNA and protein expression of MICAL-L2 was investigated using TCGA, UALCAN, and independent immunohistochemical assays. Overall survival (OS) and disease-specific survival (DSS) of COAD patients were assessed based on the MICAL-L2 expression level using the Kaplan-Meier method. Univariate and multivariate analysis was employed to determine whether MICAL-L2 could serve as an independent prognostic indicator of OS. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) were further utilized to explore the possible cellular mechanism underlying the role of MICAL-L2 in COAD. In addition, the correlation between MICAL-L2 expression and immune cell infiltration levels was investigated via single-sample gene set enrichment analysis (ssGSEA). RESULTS Data from TCGA, HPA, and UALCAN datasets indicated that MICAL-L2 expression was significantly higher in COAD tissue than in adjacent normal tissues, and this was confirmed by immunohistochemical assays. Kaplan-Meier survival analysis revealed that patients with MICAL-L2 had shorter OS and DSS. Furthermore, multivariate Cox analysis indicated that MICAL-L2 was an independent risk factor for OS in COAD patients. ROC analysis confirmed the diagnostic value of MICAL-L2, and a prognostic nomogram involving age, M stage, and MICAL-L2 expression was constructed for OS. Functional enrichment analyses revealed that transport-related activity was closely associated with the role of MICAL-L2 in COAD. Regarding immune infiltration levels, MICAL-L2 was found to be positively associated with CD56bright NK cells. CONCLUSIONS Our results suggested that MICAL-L2 is a promising biomarker for determining prognosis and correlated with immune infiltration levels in COAD.
Collapse
Affiliation(s)
- Yixing Yang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Fengwen Ye
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Tianxiang Xia
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Qianwen Wang
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China.
| | - Jun Du
- Department of Physiology, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, China.
| |
Collapse
|
17
|
Gao L, Zhao R, Liu J, Zhang W, Sun F, Yin Q, Wang X, Wang M, Feng T, Qin Y, Cai W, Li Q, Dong H, Chen X, Xiong X, Liu H, Hu J, Chen W, Han B. KIF15 Promotes Progression of Castration Resistant Prostate Cancer by Activating EGFR Signaling Pathway. Front Oncol 2021; 11:679173. [PMID: 34804913 PMCID: PMC8599584 DOI: 10.3389/fonc.2021.679173] [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: 03/11/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) continues to be a major clinical problem and its underlying mechanisms are still not fully understood. The epidermal growth factor receptor (EGFR) activation is an important event that regulates mitogenic signaling. EGFR signaling plays an important role in the transition from androgen dependence to castration-resistant state in prostate cancer (PCa). Kinesin family member 15 (KIF15) has been suggested to be overexpressed in multiple malignancies. Here, we demonstrate that KIF15 expression is elevated in CRPC. We show that KIF15 contributes to CRPC progression by enhancing the EGFR signaling pathway, which includes complex network intermediates such as mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. In CRPC tumors, increased expression of KIF15 is positively correlated with EGFR protein level. KIF15 binds to EGFR, and prevents EGFR proteins from degradation in a Cdc42-dependent manner. These findings highlight the key role of KIF15 in the development of CRPC and rationalize KIF15 as a potential therapeutic target.
Collapse
Affiliation(s)
- Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ru Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Junmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenbo Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feifei Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianshuo Yin
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yiming Qin
- College of Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Wenjie Cai
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianni Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanchen Dong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueqing Chen
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueting Xiong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hui Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
18
|
Qi C, Min P, Wang Q, Wang Y, Song Y, Zhang Y, Bibi M, Du J. MICAL2 Contributes to Gastric Cancer Cell Proliferation by Promoting YAP Dephosphorylation and Nuclear Translocation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9955717. [PMID: 34650666 PMCID: PMC8510804 DOI: 10.1155/2021/9955717] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 08/12/2021] [Accepted: 09/16/2021] [Indexed: 01/19/2023]
Abstract
Dynamic cytoskeletal rearrangements underlie the changes that occur during cell division in proliferating cells. MICAL2 has been reported to possess reactive oxygen species- (ROS-) generating properties and act as an important regulator of cytoskeletal dynamics. However, whether it plays a role in gastric cancer cell proliferation is not known. In the present study, we found that MICAL2 was highly expressed in gastric cancer tissues, and this high expression level was associated with carcinogenesis and poor overall survival in gastric cancer patients. The knockdown of MICAL2 led to cell cycle arrest in the S phase and attenuated cell proliferation. Concomitant with S-phase arrest, a decrease in CDK6 and cyclin D protein levels was observed. Furthermore, MICAL2 knockdown attenuated intracellular ROS generation, while MICAL2 overexpression led to a decrease in the p-YAP/YAP ratio and promoted YAP nuclear localization and cell proliferation, effects that were reversed by pretreatment with the ROS scavenger N-acetyl-L-cysteine (NAC) and SOD-mimetic drug tempol. We further found that MICAL2 induced Cdc42 activation, and activated Cdc42 mediated the effect of MICAL2 on YAP dephosphorylation and nuclear translocation. Collectively, our results showed that MICAL2 has a promotive effect on gastric cancer cell proliferation through ROS generation and Cdc42 activation, both of which independently contribute to YAP dephosphorylation and its nuclear translocation.
Collapse
Affiliation(s)
- Chenxiang Qi
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Pengxiang Min
- Key Laboratory of Cardio Vascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qianwen Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yueyuan Wang
- The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yixuan Song
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Maria Bibi
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
19
|
Chen Z, Xu C, Pan X, Cheng G, Liu M, Li J, Mei Y. lncRNA DSCR8 mediates miR-137/Cdc42 to regulate gastric cancer cell proliferation, invasion, and cell cycle as a competitive endogenous RNA. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:468-482. [PMID: 34553033 PMCID: PMC8430047 DOI: 10.1016/j.omto.2021.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
lncRNA DSCR8 (Down syndrome critical region 8) is involved in progression of many cancers, but its specific role in gastric cancer (GC) is still unclear. Here, qRT-PCR detected upregulated expression of DSCR8 and Cdc42 and downregulated expression of miR-137 in GC. The protein expression level of Cdc42 in GC was upregulated as tested by western blot. Statistical analysis showed that DSCR8 was closely associated with some malignant clinicopathological features (such as tumor size, metastasis, and stage) in GC patients. Fluorescence in situ hybridization showed that DSCR8 was localized in the nucleus and cytoplasm. Dual-luciferase reporter gene, RNA immunoprecipitation, and biotin pull-down assays showed that DSCR8 could bind to miR-137 could bind to Cdc42. In vitro and in vivo assays showed that DSCR8 could promote proliferation, invasion, and the cycle of GC cells and inhibit cell apoptosis. In addition, a rescue experiment showed that DSCR8 regulated progression of GC cells via miR-137. Furthermore, DSCR8 regulated Cdc42 in GC cells by inhibiting miR-137. Taken together, these data indicated that DSCR8 could adsorb miR-137 to reduce its inhibitory effect on Cdc42 expression, thereby promoting the progression of GC cells and regulating the cell cycle. These results provide a novel direction for DSCR8 as a target of GC.
Collapse
Affiliation(s)
- Zhengwei Chen
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| | - Chaobo Xu
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| | - Xiaoming Pan
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| | - Guoxiong Cheng
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| | - Ming Liu
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| | - Jiaxin Li
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| | - Yijun Mei
- Department of Gastrointestinal Surgery, Lishui People's Hospital of Zhejiang Province, 15 Dazhong Street, Liandu District, Lishui City, Zhejiang 323000, China
| |
Collapse
|
20
|
Zhang L, Yao L, Zhou W, Tian J, Ruan B, Lu Z, Deng Y, Li Q, Zeng Z, Yang D, Shang R, Xu M, Zhang M, Cheng D, Yang Y, Ding Q, Yu H. miR-497 defect contributes to gastric cancer tumorigenesis and progression via regulating CDC42/ITGB1/FAK/PXN/AKT signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:567-577. [PMID: 34589278 PMCID: PMC8463315 DOI: 10.1016/j.omtn.2021.07.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 07/27/2021] [Indexed: 01/20/2023]
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related death worldwide. MicroRNAs (miRNAs) are known to be important regulators of GC. This study aims to investigate the role of miRNA (miR)-497 in GC. We demonstrated that the expression of miR-497 was downregulated in human GC tissues. After N-methyl-N-nitrosourea treatment, the incidence of GC in miR-497 knockout mice was significantly higher than that in wild-type mice. miR-497 overexpression suppressed GC cell proliferation, cell-cycle progression, colony formation, anti-apoptosis ability, and cell migration and invasion capacity. Additionally, miR-497 overexpression decreased the expression levels of cell division cycle 42 (CDC42) and integrin β1 (ITGB1) and inhibited the phosphorylation of focal adhesion kinase (FAK), paxillin (PXN), and serine-threonine protein kinase (AKT). Furthermore, overexpression of miR-497 inhibited the metastasis of GC cells in vivo, which could be counteracted by CDC42 restoration. Furthermore, the focal adhesion of GC cells was found to be regulated by miR-497/CDC42 axis via ITGB1/FAK/PXN/AKT signaling. Collectively, it is concluded that miR-497 plays an important role in the repression of GC tumorigenesis and progression, partly via the CDC42/ITGB1/FAK/PXN/AKT pathway.
Collapse
Affiliation(s)
- Lihui Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Liwen Yao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Wei Zhou
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Jinping Tian
- Medical Research Center, Xi’an No. 3 Hospital, the Affiliated Hospital of Northwest University, Weiyang District, Xi’an, Shaanxi 710016, PR China
| | - Banlai Ruan
- Medical Research Center, Xi’an No. 3 Hospital, the Affiliated Hospital of Northwest University, Weiyang District, Xi’an, Shaanxi 710016, PR China
| | - Zihua Lu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Yunchao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Qing Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Dongmei Yang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Renduo Shang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Ming Xu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Mengjiao Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Du Cheng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | - Yanning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
| | - Qianshan Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Medical Research Center, Xi’an No. 3 Hospital, the Affiliated Hospital of Northwest University, Weiyang District, Xi’an, Shaanxi 710016, PR China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, Hubei 430060, PR China
- Hubei Key Laboratory of Digestive System, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive Incision, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| |
Collapse
|
21
|
Chen N, Han X, Yin B, Bai X, Wang Y. FGD5 facilitates tumor growth by regulating EGFR ubiquitination in gastric cancer. Biochem Biophys Res Commun 2021; 562:43-49. [PMID: 34034092 DOI: 10.1016/j.bbrc.2021.04.106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 01/19/2023]
Abstract
FGD5 (faciogenital dysplasia-5), a Rho-family guanine nucleotide exchange factor, has been identified as a key regulator of endothelial cells angiogenesis and apoptosis. However, the expression and role of FGD5 in the pathogenesis of gastric cancer remain unknown. In the present study, we first detected FGD5 expression in tumor tissues of different stages by immunofluorescence and confirmed that FGD5 expression was associated with stages in human gastric cancer. Knockdown FGD5 by shRNA in 7901 and BGC823 human gastric cells lines inhibited tumorigenesis and migration in vivo and in vitro. Mechanistically, co-immunoprecipitation (Co-IP) assay showed that FGD5 interacted with EGFR and decreased EGFR ubiquitination. Additionally, FGD5 sustained the activation of EGFR downstream signaling molecules, including STAT3 and pSTAT3. Furthermore, we showed that FGD5 could induce resistance to chemotherapy 5Fu/CIS, however, a well-known STAT3 inhibitor FLL32 could reverse FGD5-induced chemotherapy resistance in vivo. In conclusion, we are the first to demonstrate that FGD5 expression was associated with tumor stage and proliferation in gastric cancer and targeting FGD5 might be a potential therapeutic target for EGFR-STAT3 resistance gastric cancer.
Collapse
Affiliation(s)
- Na Chen
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Xiangdong Han
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Bo Yin
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Xue Bai
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China
| | - Yubin Wang
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, China.
| |
Collapse
|
22
|
Min P, Zhang L, Wang Y, Qi C, Song Y, Bibi M, Zhang Y, Ma Y, Zhao X, Yu M, Du J. MICAL-L2 Is Essential for c-Myc Deubiquitination and Stability in Non-small Cell Lung Cancer Cells. Front Cell Dev Biol 2021; 8:575903. [PMID: 33520979 PMCID: PMC7841116 DOI: 10.3389/fcell.2020.575903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/16/2020] [Indexed: 01/01/2023] Open
Abstract
Objectives: MICAL-L2, a member of the molecules interacting with the CasL (MICAL) family, was reported to be highly expressed in several types of cancers, however, the roles of MICAL-L2 in NSCLC pathogenesis remain to be explored. This study is designed to clarify the mechanisms by which MICAL-L2 participates in NSCLC cell proliferation. Materials and Methods: The expression levels of MICAL-L2 in human lung cancer samples were assessed by immunohistochemical staining. Cells were transfected with siRNA or plasmids to regulate MICAL-L2 expression. Cell proliferation was measured by EdU staining and CCK-8 assays. MICAL-L2 and phosphorylated/total c-Myc expression were examined by Western blotting analysis. Interaction between MICAL-L2 and c-Myc was assessed by immunofluorescence staining, Western blotting and co-immunoprecipitation assays. Western blotting, polyubiquitylation detection and protein stability assays were used to assess whether MICAL-L2 exerts its oncogenic effect via c-Myc. Results: We found that MICAL-L2 was highly expressed in human NSCLC. While overexpressing MICAL-L2 increased NSCLC cell proliferation, MICAL-L2 depletion decreased the proliferation of NSCLC cells, an effect that was linked to cell cycle arrest. MICAL-L2 physically interacted with the c-Myc protein and functioned to maintain nuclear c-Myc levels and prolonged its half-life. Knockdown of MICAL-L2 expression led to decreased c-Myc protein stability through accelerating polyubiquitylation of c-Myc and gave rise to c-Myc degradation. We further found that MICAL-L2 deubiquitinated c-Myc and blocked its degradation, presumably by inhibiting c-Myc phosphorylation at threonine residue 58. Conclusions: These results indicate that MICAL-L2 is a key regulator of c-Myc deubiquitination and stability in the nucleus, and this activity may be involved in promoting NSCLC cell proliferation.
Collapse
Affiliation(s)
- Pengxiang Min
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Lin Zhang
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yueyuan Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Chenxiang Qi
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yixuan Song
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Maria Bibi
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yadong Ma
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Xuyang Zhao
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Minjie Yu
- The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
23
|
Lu J, Huang XY, Wang YH, Xie JW, Wang JB, Lin JX, Chen QY, Cao LL, Li P, Huang CM, Zheng CH. POC1A acts as a promising prognostic biomarker associated with high tumor immune cell infiltration in gastric cancer. Aging (Albany NY) 2020; 12:18982-19011. [PMID: 33052878 PMCID: PMC7732308 DOI: 10.18632/aging.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/04/2020] [Indexed: 01/24/2023]
Abstract
The effect of POC1 centriolar protein A (POC1A) on gastric cancer (GC) has not been clearly defined. In this study, POC1A expression and clinical information in patients with GC were analyzed. Multiple databases were used to investigate the genes that were co-expressed with POC1A and genes whose changes co-occurred with genetic alternations of POC1A. Moreover, the TISIDB and TIMER databases were used to analyze immune infiltration. The GSE54129 GC dataset and LASSO regression model (tumor vs. normal) were employed, and 6 significant differentially expressed genes (LAMP5, CEBPB, ARMC9, PAOX, VMP1, POC1A) were identified. POC1A was selected for its high expression in adjacent tissues, which was confirmed with IHC. High POC1A expression was related to better overall and recurrence-free survival. GO and KEGG analyses demonstrated that POC1A may regulate the cell cycle, DNA replication and cell growth. Furthermore, POC1A was found to be correlated with immune infiltration levels in GC according to the TISIDB and TIMER databases. These findings indicate that POC1A acts as a tumor suppressor in GC by regulating the cell cycle and cell growth. In addition, POC1A preferentially regulates the immune infiltration of GC via several immune genes. However, the specific mechanism requires further study.
Collapse
Affiliation(s)
- Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Xiao-Yan Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yao-Hui Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
24
|
Yang W, Wang K, Ma J, Hui K, Lv W, Ma Z, Huan M, Luo L, Wang X, Li L, Chen Y. Inhibition of Androgen Receptor Signaling Promotes Prostate Cancer Cell Migration via Upregulation of Annexin A1 Expression. Arch Med Res 2020; 52:174-181. [PMID: 33059953 DOI: 10.1016/j.arcmed.2020.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 07/13/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent studies indicate that androgen deprivation therapy (ADT), the main therapeutic approach for metastatic prostate cancer (PCa), accelerates PCa invasion and metastasis. Annexin A1 (ANXA1) is a Ca2+-regulated phospholipid-binding protein that can promote PCa migration and invasion. AIM OF THE STUDY The aim of this study is to determine whether ANXA1 is regulated by ADT and participates in PCa progression after ADT, and to explore the possible mechanism of ANXA1-mediated PCa migration. METHODS Expression of ANXA1 and androgen receptor (AR) in PCa cell lines and tissues was detected, and the association between these two proteins were analyzed. Expression of ANXA1 was evaluated after AR knockdown or AR inhibition in PCa cell lines. Cell migration of PCa cell liness after ANXA1 knockdown or overexpression was determined by in vitro migration assay. Transcriptome analysis was used to explore the possible mechanism of ANXA1-mediated PCa migration. RESULTS ANXA1 expression in PCa cell lines and tissues was reversely associated with AR. In vitro studies revealed an increase in ANXA1 expression after AR knockdown or treatment with AR antagonist. Moreover, functional assays indicated that ANXA1 knockdown in PCa cells significantly inhibited cell migration, while ANXA1 overexpression in PCa cells significantly accelerated cell migration. Transcriptome analysis showed that ANXA1 regulated multiple genes involved in cell junction organization, such as CADM1, LIMCH1 and PPM1F. CONCLUSIONS Our results indicate that ADT might accelerate PCa metastasis via ANXA1 expression and PCa cell migration.
Collapse
Affiliation(s)
- Wenjie Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, PR China
| | - Jianbin Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Ke Hui
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Wei Lv
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Zhenkun Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, PR China
| | - Mengxi Huan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Lin Luo
- Department of Urology, 521 Hospital of Norinco Group, Xi'an, PR China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, PR China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, PR China.
| | - Yule Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, PR China; Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an, PR China.
| |
Collapse
|
25
|
Yin LM, Schnoor M, Jun CD. Structural Characteristics, Binding Partners and Related Diseases of the Calponin Homology (CH) Domain. Front Cell Dev Biol 2020; 8:342. [PMID: 32478077 PMCID: PMC7240100 DOI: 10.3389/fcell.2020.00342] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 01/01/2023] Open
Abstract
The calponin homology (CH) domain is one of the most common modules in various actin-binding proteins and is characterized by an α-helical fold. The CH domain plays important regulatory roles in both cytoskeletal dynamics and signaling. The CH domain is required for stability and organization of the actin cytoskeleton, calcium mobilization and activation of downstream pathways. The CH domain has recently garnered increased attention due to its importance in the onset of different diseases, such as cancers and asthma. However, many roles of the CH domain in various protein functions and corresponding diseases are still unclear. Here, we review current knowledge about the structural features, interactome and related diseases of the CH domain.
Collapse
Affiliation(s)
- Lei-Miao Yin
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and Meridian, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Michael Schnoor
- Molecular Biomedicine, Center for Investigation and Advanced Studies of the National Polytechnic Institute (Cinvestav), Mexico City, Mexico
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| |
Collapse
|
26
|
Zhang S, Wu X, Diao P, Wang C, Wang D, Li S, Wang Y, Cheng J. Identification of a prognostic alternative splicing signature in oral squamous cell carcinoma. J Cell Physiol 2019; 235:4804-4813. [PMID: 31637730 DOI: 10.1002/jcp.29357] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/07/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Shuting Zhang
- Jiangsu Key Laboratory of Oral Disease Nanjing Medical University Jiangsu China
| | - Xiang Wu
- Jiangsu Key Laboratory of Oral Disease Nanjing Medical University Jiangsu China
| | - Pengfei Diao
- Jiangsu Key Laboratory of Oral Disease Nanjing Medical University Jiangsu China
| | - Chenxing Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology Nanjing Medical University Nanjing China
| | - Dongmiao Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology Nanjing Medical University Nanjing China
| | - Sheng Li
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology Nanjing Medical University Nanjing China
| | - Yanling Wang
- Jiangsu Key Laboratory of Oral Disease Nanjing Medical University Jiangsu China
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Disease Nanjing Medical University Jiangsu China
- Department of Oral and Maxillofacial Surgery, Affiliated Hospital of Stomatology Nanjing Medical University Nanjing China
| |
Collapse
|
27
|
Min P, Zhao S, Liu L, Zhang Y, Ma Y, Zhao X, Wang Y, Song Y, Zhu C, Jiang H, Gu L, Du J. MICAL-L2 potentiates Cdc42-dependent EGFR stability and promotes gastric cancer cell migration. J Cell Mol Med 2019; 23:4475-4488. [PMID: 31034158 PMCID: PMC6533512 DOI: 10.1111/jcmm.14353] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/29/2019] [Accepted: 04/12/2019] [Indexed: 01/21/2023] Open
Abstract
Enhanced migration potential is a common characteristic of cancer cells induced by mechanisms that are incompletely defined. The present study was designed to investigate relationship of a new discovered cytoskeleton regulator MICAL‐L2 and the endogenous epidermal growth factor receptor (EGFR) signalling pathways in gastric cancer cell migration. Increased expression of MICAL‐L2 in gastric cancer cells up‐regulated EGFR protein level, accompanied by the increase of cell migration, whereas silencing MICAL‐L2 down‐regulated EGFR and inhibited cell migration. Expression of MICAL‐L2 was also shown positively correlated with the activation of HSP27/cytoskeleton and HSP27/β‐catenin signalling pathways that provide key mechanisms controlling cell migration. The up‐regulating effect of MICAL‐L2 on EGFR is mediated through a transcription‐independent mechanism that involves inhibiting EGFR protein degradation in lysosome. Further analysis indicated that Cdc42 activation contributed in maintaining the effect of MICAL‐L2 on EGFR stability. Furthermore analysis of clinic specimens revealed increased expression of MICAL‐L2 in carcinoma tissues and a positive correlation between MICAL‐L2 and EGFR expression levels. The above results indicate that MICAL‐L2 potentiates gastric cell migration via inhibiting EGFR degradation in lysosome via a Cdc42‐dependent manner that leads to the activation of EGFR/HSP27 signalling pathways.
Collapse
Affiliation(s)
- Pengxiang Min
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuo Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yadong Ma
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuyang Zhao
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yueyuan Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yixuan Song
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenchen Zhu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haonan Jiang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Luo Gu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|