1
|
Xu J, Chen S, Hao T, Liu G, Zhang K, Zhang C, He Y. MEX3A promotes colorectal cancer migration, invasion and EMT via regulating the Wnt/β-catenin signaling pathway. J Cancer Res Clin Oncol 2024; 150:319. [PMID: 38914858 PMCID: PMC11196291 DOI: 10.1007/s00432-024-05845-9] [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: 04/19/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Mex-3 RNA binding family members are well-established to be important in cancer development and progression. However, the functions of Mex-3 RNA binding family member A (MEX3A) in colorectal cancer (CRC) metastasis remain poorly understood. In this study, we aim to reveal the function and the mechanism of MEX3A in promoting CRC metastasis. METHODS We used multiple databases including TCGA database, UALCAN, LinkedOmics, CancerSEA, GeneMANIA and STRING database to investigate the expression, the functions and underlying molecular mechanism of MEX3A in CRC. Multiple experimental methods were adapted to determine the study, including real-time PCR (qPCR), immunohistochemistry (IHC), western blot (WB), transfection, transwell migration and invasion assays, immunofluorescence (IF). RESULTS We found that MEX3A was significantly upregulated and correlated to tumor stage and lymph nodal metastasis in CRC through bioinformatics analysis and tissue immunohistochemistry (IHC). The higher expression of MEX3A in CRC correlated with poor recurrence-free survival (RFS) and overall survival (OS). In vitro studies showed that knockdown of MEX3A suppressed EMT transition, invasion and metastasis of CRC cells. Mechanistically, we revealed that MEX3A promotes epithelial-mesenchymal transition (EMT), invasion and metastasis of CRC cells by upregulating the Wnt/β-catenin signaling pathway. CONCLUSION In conclusion, our study reveals that MEX3A promotes CRC migration, invasion and EMT via regulating the Wnt/β-catenin signaling pathway and could be a novel therapeutic target for this patient population.
Collapse
Affiliation(s)
- Jiannan Xu
- Center of Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Songyao Chen
- Center of Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Tengfei Hao
- Center of Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Guangyao Liu
- Center of Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Kai Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Changhua Zhang
- Center of Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Yulong He
- Center of Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
- Center of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
2
|
Wang D, Shi W, Qiu C. Construction of a TP53 mutation-associated ceRNA network as prognostic biomarkers in hepatocellular carcinoma. Heliyon 2024; 10:e30066. [PMID: 38737272 PMCID: PMC11088256 DOI: 10.1016/j.heliyon.2024.e30066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) continues to endanger human health worldwide. Regulatory networks of competing endogenous RNAs (ceRNAs) play important roles in HCC. TP53 is the second most often altered gene in HCC and has a significant role in regulating target genes such as miRNAs and lncRNAs. Methods Data from patients with TP53 mutation were collected through the cBioPortal database and differential analysis was performed to screen RNAs related to TP53 mutation. The lncRNA-miRNA-mRNA relationship was predicted by the miRcode, miRDB, and TargetScan databases. The ceRNA networks were screened and visualized by Cytoscape. Core ceRNA networks were generated by differential analysis, coexpression analysis, prognostic analysis and subcellular localization. Finally, methylation, mutation, PPI, GSEA, immunity and drug sensitivity analyses of MEX3A were performed to determine the role of MEX3A in HCC. Results We identified 1508 DEmRNAs, 85 DEmiRNAs and 931 DElncRNAs and obtained a ceRNA network including 28 lncRNAs, 4 miRNAs and 31 mRNAs. Twenty hub DERNAs in the TP53-altered-related ceRNA network were screened out by Cytoscape and the core ceRNA network (LINC00491/TCL6-hsa-miR-139-5p-MEX3A) was obtained by multiple analyses. In addition, we discovered that the methylation level of MEX3A was decreased and the mutation frequency was raised in HCC. Furthermore, elevated MEX3A expression was associated with alterations in the HCC immunological microenvironment. Conclusion We successfully constructed a reciprocal ceRNA network, which could provide new ideas for exploring HCC mechanisms and therapeutic approaches.
Collapse
Affiliation(s)
- Dong Wang
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou 213000, China
| | - Wenxiang Shi
- Department of Pediatric Cardiology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Chenjie Qiu
- Department of General Surgery, Changzhou Hospital of Traditional Chinese Medicine, Changzhou 213000, China
| |
Collapse
|
3
|
Deb R, Sengar GS, Sonowal J, Pegu SR, Das PJ, Singh I, Chakravarti S, Selvaradjou A, Attupurum N, Rajkhowa S, Gupta VK. Transcriptome signatures of host tissue infected with African swine fever virus reveal differential expression of associated oncogenes. Arch Virol 2024; 169:54. [PMID: 38381218 DOI: 10.1007/s00705-023-05959-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/27/2023] [Indexed: 02/22/2024]
Abstract
African swine fever (ASF) has emerged as a threat to swine production worldwide. Evasion of host immunity by ASF virus (ASFV) is well understood. However, the role of ASFV in triggering oncogenesis is still unclear. In the present study, ASFV-infected kidney tissue samples were subjected to Illumina-based transcriptome analysis. A total of 2463 upregulated and 825 downregulated genes were differentially expressed (p < 0.05). A literature review revealed that the majority of the differentially expressed host genes were key molecules in signaling pathways involved in oncogenesis. Bioinformatic analysis indicated the activation of certain oncogenic KEGG pathways, including basal cell carcinoma, breast cancer, transcriptional deregulation in cancer, and hepatocellular carcinoma. Analysis of host-virus interactions revealed that the upregulated oncogenic RELA (p65 transcription factor) protein of Sus scrofa can interact with the A238L (hypothetical protein of unknown function) of ASFV. Differential expression of oncogenes was confirmed by qRT-PCR, using the H3 histone family 3A gene (H3F3A) as an internal control to confirm the RNA-Seq data. The levels of gene expression indicated by qRT-PCR matched closely to those determined through RNA-Seq. These findings open up new possibilities for investigation of the mechanisms underlying ASFV infection and offer insights into the dynamic interaction between viral infection and oncogenic processes. However, as these investigations were conducted on pigs that died from natural ASFV infection, the role of ASFV in oncogenesis still needs to be investigated in controlled experimental studies.
Collapse
Affiliation(s)
- Rajib Deb
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India.
| | | | - Joyshikh Sonowal
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
- Multidisciplinary Research Unit, Jorhat Medical College and Hospital, Jorhat, Assam, 785001, India
| | - Seema Rani Pegu
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Pranab Jyoti Das
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India.
| | | | - Soumendu Chakravarti
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, UP, 243122, India
- Pirbright Institute, Ash Road, Pirbright, Surrey, United Kingdom
| | | | - Nitin Attupurum
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Swaraj Rajkhowa
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India
| | - Vivek Kumar Gupta
- ICAR-National Research Centre on Pig, Rani, Guwahati, Assam, 781131, India.
| |
Collapse
|
4
|
Zhang M, Cao L, Hou G, Lv X, Deng J. Investigation of the Potential Correlation Between RNA-Binding Proteins in the Evolutionarily Conserved MEX3 Family and Non-small-Cell Lung Cancer. Mol Biotechnol 2022:10.1007/s12033-022-00638-2. [DOI: 10.1007/s12033-022-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Abstract
Members of the MEX3 (muscle excess 3) family, uniquely characterised as mRNA binding proteins, play emerging roles in the post-transcriptional regulation of programmed biological processes, including tumour cell death and immune mechanisms, and have been shown to be involved in a variety of diseases. However, the role of MEX3 in non-small cell lung cancer (NSCLC) has not been fully elucidated. In this study, we found no significant changes in the sequence and copy number of the MEX3 gene through analysis using the COSMIC database, revealing its stability during malignancy development. Its expression in NSCLC was examined using the Oncomine™ database, and the prognosis of each member gene was analysed by Kaplan–Meier. The results showed that overexpression of MEX3A, MEX3B, MEX3C and MEX3D was associated with significantly worse OS in patients with LUAD, while overexpression of MEX3D was also associated with significantly worse OS in patients with LUSC. Afterwards, we applied the Tumour Immunology Estimation Resource (TIMER) tool to assess the correlation between different MEX3 and infiltrative immune cell infiltration. Ultimately, we found that most MEX3 members were highly expressed in NSCLC, with high expression suggesting poor prognosis and correlating with immune cell infiltration. The complexity and heterogeneity of NSCLC was understood through MEX3, setting the framework for the prognostic impact of MEX3 in NSCLC patients and the development of new targeted therapeutic strategies in the future.
Collapse
|
5
|
Nsengimana B, Khan FA, Ngowi EE, Zhou X, Jin Y, Jia Y, Wei W, Ji S. Processing body (P-body) and its mediators in cancer. Mol Cell Biochem 2022; 477:1217-1238. [PMID: 35089528 DOI: 10.1007/s11010-022-04359-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/11/2022] [Indexed: 12/24/2022]
Abstract
In recent years, processing bodies (P-bodies) formed by liquid-liquid phase separation, have attracted growing scientific attention due to their involvement in numerous cellular activities, including the regulation of mRNAs decay or storage. These cytoplasmic dynamic membraneless granules contain mRNA storage and decay components such as deadenylase and decapping factors. In addition, different mRNA metabolic regulators, including m6A readers and gene-mediated miRNA-silencing, are also associated with such P-bodies. Cancerous cells may profit from these mRNA decay shredders by up-regulating the expression level of oncogenes and down-regulating tumor suppressor genes. The main challenges of cancer treatment are drug resistance, metastasis, and cancer relapse likely associated with cancer stem cells, heterogeneity, and plasticity features of different tumors. The mRNA metabolic regulators based on P-bodies play a great role in cancer development and progression. The dysregulation of P-bodies mediators affects mRNA metabolism. However, less is known about the relationship between P-bodies mediators and cancerous behavior. The current review summarizes the recent studies on P-bodies mediators, their contribution to tumor development, and their potential in the clinical setting, particularly highlighting the P-bodies as potential drug-carriers such as exosomes to anticancer in the future.
Collapse
Affiliation(s)
- Bernard Nsengimana
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China
| | - Faiz Ali Khan
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China
| | - Ebenezeri Erasto Ngowi
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China
| | - Xuefeng Zhou
- Department of Oncology, Dongtai Affiliated Hospital of Nantong University, Dongtai, 224200, Jiangsu, People's Republic of China
| | - Yu Jin
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China
| | - Yuting Jia
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China
| | - Wenqiang Wei
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China.
| | - Shaoping Ji
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Henan, 475004, People's Republic of China.
| |
Collapse
|
6
|
Chu J, Li T, Li L, Fan H. MicroRNA-139-5p Suppresses Cell Malignant Behaviors in Breast Cancer through Targeting MEX3A. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6591541. [PMID: 34765013 PMCID: PMC8577887 DOI: 10.1155/2021/6591541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The study was designed to evaluate the underlying mechanism of microRNA-139-5p in breast cancer (BC). METHODS Expression statuses of microRNA-139-5p and MEX3A were measured by qRT-PCR and western blotting. The anticancer effect of microRNA-139-5p in vitro was tested by a set of assays. Interaction between microRNA-139-5p and MEX3A was validated by dual-luciferase detection. RESULTS MicroRNA-139-5p expression in BC cells was obviously low, while MEX3A was significantly overexpressed. MicroRNA-139-5p restrained proliferative, invasive, and migratory abilities of BC cells and increased apoptosis level of BC cells, while MEX3A exerted a promoting effect on BC cell growth. Dual-luciferase reporter detection confirmed that microRNA-139-5p bound to MEX3A 3'-UTR. CONCLUSIONS MicroRNA-139-5p inhibited the development of BC by targeting MEX3A. MicroRNA-139-5p/MEX3A may be a target for BC therapy.
Collapse
Affiliation(s)
- Jian Chu
- Department of General Surgery, The First People's Hospital of Yancheng, Yancheng, 224005 Jiangsu Province, China
- Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, China
| | - Tangya Li
- Department of General Surgery, The First People's Hospital of Yancheng, Yancheng, 224005 Jiangsu Province, China
- Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, China
| | - Lei Li
- Department of General Surgery, The First People's Hospital of Yancheng, Yancheng, 224005 Jiangsu Province, China
- Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, China
| | - Huiwen Fan
- Department of General Surgery, The First People's Hospital of Yancheng, Yancheng, 224005 Jiangsu Province, China
- Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, China
| |
Collapse
|
7
|
Bufalieri F, Basili I, Di Marcotullio L, Infante P. Harnessing the Activation of RIG-I Like Receptors to Inhibit Glioblastoma Tumorigenesis. Front Mol Neurosci 2021; 14:710171. [PMID: 34305530 PMCID: PMC8295747 DOI: 10.3389/fnmol.2021.710171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GB) is an incurable form of brain malignancy in an adult with a median survival of less than 15 months. The current standard of care, which consists of surgical resection, radiotherapy, and chemotherapy with temozolomide, has been unsuccessful due to an extensive inter- and intra-tumoral genetic and molecular heterogeneity. This aspect represents a serious obstacle for developing alternative therapeutic options for GB. In the last years, immunotherapy has emerged as an effective treatment for a wide range of cancers and several trials have evaluated its effects in GB patients. Unfortunately, clinical outcomes were disappointing particularly because of the presence of tumor immunosuppressive microenvironment. Recently, anti-cancer approaches aimed to improve the expression and the activity of RIG-I-like receptors (RLRs) have emerged. These innovative therapeutic strategies attempt to stimulate both innate and adaptive immune responses against tumor antigens and to promote the apoptosis of cancer cells. Indeed, RLRs are important mediators of the innate immune system by triggering the type I interferon (IFN) response upon recognition of immunostimulatory RNAs. In this mini-review, we discuss the functions of RLRs family members in the control of immune response and we focus on the potential clinical application of RLRs agonists as a promising strategy for GB therapy.
Collapse
Affiliation(s)
| | - Irene Basili
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Paola Infante
- Center For Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| |
Collapse
|
8
|
Comprehensive Analysis of Prognostic Value of MEX3A and Its Relationship with Immune Infiltrates in Ovarian Cancer. J Immunol Res 2021; 2021:5574176. [PMID: 34189143 PMCID: PMC8195639 DOI: 10.1155/2021/5574176] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 05/03/2021] [Accepted: 05/20/2021] [Indexed: 12/18/2022] Open
Abstract
MEX3A is a critical RNA-binding ubiquitin ligase that is upregulated in various types of cancer. However, the correlations of MEX3A with prognosis and its molecular mechanism in ovarian cancer (OC) remain unclear. The expression level, prognostic values, and the genetic variations of MEX3A were analyzed via Gene Expression Profiling Interactive Analysis (GEPIA) Oncomine, Kaplan-Meier plotter, and cBioPortal. We used the LinkedOmics database to investigate the functions of MEX3A coexpressed genes and performed visualizing gene interaction network analysis on the GeneMANIA website. The correlations between MEX3A and cancer immune infiltration were analyzed by the Tumor Immune Estimation Resource (TIMER) site and the TISIDB database. Furthermore, in vitro analysis was performed to evaluate the biological functions of MEX3A in OC cells. Our study showed that the expression of the MEX3A in OC was higher than in normal tissues; it had the greatest prognostic value in OC, and strong physical interaction with PABPC1, LAMTOR2, KHDRBS2, and IGF2BP2, which indicated the association between MEX3A and immune infiltration. We also found that MEX3A was negatively related to infiltrating levels of several types of immune cells, including macrophages, neutrophils, dendritic cells (DCs), B cells, and CD8+ T cells. Additionally, in vitro experiments demonstrated that MEX3A promotes proliferation and migration in OC cells. Taken together, MEX3A might influence the biological functions of OC cells by regulating the immune infiltration in the microenvironment as a prognostic biomarker and a potential therapeutic target.
Collapse
|
9
|
Lederer M, Müller S, Glaß M, Bley N, Ihling C, Sinz A, Hüttelmaier S. Oncogenic Potential of the Dual-Function Protein MEX3A. BIOLOGY 2021; 10:415. [PMID: 34067172 PMCID: PMC8151450 DOI: 10.3390/biology10050415] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
MEX3A belongs to the MEX3 (Muscle EXcess) protein family consisting of four members (MEX3A-D) in humans. Characteristic for MEX3 proteins is their domain structure with 2 HNRNPK homology (KH) domains mediating RNA binding and a C-terminal really interesting new gene (RING) domain that harbors E3 ligase function. In agreement with their domain composition, MEX3 proteins were reported to modulate both RNA fate and protein ubiquitination. MEX3 paralogs exhibit an oncofetal expression pattern, they are severely downregulated postnatally, and re-expression is observed in various malignancies. Enforced expression of MEX3 proteins in various cancers correlates with poor prognosis, emphasizing their oncogenic potential. The latter is supported by MEX3A's impact on proliferation, self-renewal as well as migration of tumor cells in vitro and tumor growth in xenograft studies.
Collapse
Affiliation(s)
- Marcell Lederer
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Simon Müller
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Markus Glaß
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Nadine Bley
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| | - Christian Ihling
- Center for Structural Mass Spectrometry, Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany; (C.I.); (A.S.)
| | - Andrea Sinz
- Center for Structural Mass Spectrometry, Department of Pharmaceutical Chemistry & Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany; (C.I.); (A.S.)
| | - Stefan Hüttelmaier
- Charles Tanford Protein Center, Faculty of Medicine, Institute of Molecular Medicine, Section for Molecular Cell Biology, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany; (S.M.).; (M.G.).; (N.B.); (S.H.)
| |
Collapse
|
10
|
Xu Y, Pan S, Chen H, Qian H, Wang Z, Zhu X. MEX3A suppresses proliferation and EMT via inhibiting Akt signaling pathway in cervical cancer. Am J Cancer Res 2021; 11:1446-1462. [PMID: 33948367 PMCID: PMC8085868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/11/2021] [Indexed: 06/12/2023] Open
Abstract
MEX3A, one member of the human MEX3 gene family, exerts different effects on a variety of human cancer cells. However, the biological functions and regulatory mechanism have not been explored in cervical cancer. In our study, we used multiple approaches to determine the functions and underlying molecular mechanism of MEX3A in cervical tumorigenesis, including CCK-8 assay, BrdU assay, FACS for cell cycle and apoptosis, wound healing assay, Transwell migration and invasion assays, immunohistochemistry (IHC) assay, Transfection, real-time RT-PCR and Western blotting analysis. IHC results showed that the expression levels of MEX3A were decreased in cervical cancer patients with advanced clinical stages and lymph node involvement. Moreover, upregulation of MEX3A attenuated cell proliferation, migration and invasion and induced cell cycle arrest at G0/G1 phase in human cervical cancer cells, whereas knockdown of MEX3A exhibited the opposite effects. Mechanistically, MEX3A exerted its tumor suppressive functions via inactivation of Akt signaling pathway and inhibiting epithelial to mesenchymal transition (EMT). Importantly, Akt activation by its activator SC79 reversed the biological functions of MEX3A overexpression. Furthermore, MEX3A inhibited tumor growth in xenograft models. Overall, our investigation suggested that MEX3A participated in antitumor activity in cervical cancer by inhibition of the Akt signaling pathway and EMT. Hence, targeting MEX3A might have a therapeutic potential to treat cervical cancer.
Collapse
Affiliation(s)
- Yichi Xu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou, People's Republic of China
| | - Shuya Pan
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou, People's Republic of China
| | - Hong Chen
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou, People's Republic of China
| | - Hongfei Qian
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou, People's Republic of China
| | - Zhiwei Wang
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou, People's Republic of China
| | - Xueqiong Zhu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University Wenzhou, People's Republic of China
| |
Collapse
|
11
|
The effects of MEX3A knockdown on proliferation, apoptosis and migration of osteosarcoma cells. Cancer Cell Int 2021; 21:197. [PMID: 33827584 PMCID: PMC8028067 DOI: 10.1186/s12935-021-01882-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/16/2021] [Indexed: 12/21/2022] Open
Abstract
Background Osteosarcoma is an aggressive malignant tumor which has attracted worldwide attention. MEX3A may be associated with tumors while has not yet seen its coverage on osteosarcoma. Herein, this study was to investigate the correlation between MEX3A and the progression of osteosarcoma. Methods Firstly, we determined that expression of MEX3A was significantly higher in osteosarcoma tissues than that in marginal bone by immunohistochemical staining. Additionally, MEX3A expression was downregulated by the RNAi‐mediated knockdown. The functions of MEX3A knockdown on proliferation, apoptosis, cell cycle, migration was assessed by MTT assay, flow cytometry, wound-healing assay and Transwell assay, respectively. Knockdown of MEX3A resulted in suppressing cell proliferation, increasing cell apoptosis, inducing the G2 phase cell cycle arrest, and attenuating cellular migration. Furthermore, mouse xenograft model confirmed inhibitory effects of MEX3A knockdown on osteosarcoma formation. Results The preliminary exploration on the molecular mechanism of MEX3A in osteosarcoma cells showed that the induction of apoptosis needs the participation of a series of apoptosis- associated factors, such as upregulation of Caspase 3, Caspase 8 and HSP60, downregulation of HSP27 and XIAP. Conclusions In summary, these findings predicated that therapy directed at decreasing MEX3A expression is a potential osteosarcoma treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01882-3.
Collapse
|
12
|
Yang C, Zhan H, Zhao Y, Wu Y, Li L, Wang H. MEX3A contributes to development and progression of glioma through regulating cell proliferation and cell migration and targeting CCL2. Cell Death Dis 2021; 12:14. [PMID: 33414423 PMCID: PMC7791131 DOI: 10.1038/s41419-020-03307-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/03/2023]
Abstract
Glioma is one of the most commonly diagnosed intracranial malignant tumors with extremely high morbidity and mortality, whose treatment was seriously limited because of the unclear molecular mechanism. In this study, in order to identify a novel therapeutic target for glioma treatment, we explored the functions and mechanism of MEX3A in regulating glioma. The immunohistochemical staining of MEX3A in glioma and normal tissues revealed the upregulation of MEX3A and further indicated the relationship between high MEX3A expression and higher malignancy as well as poorer prognosis of glioma. In vitro loss-of-function and gain-of-function experiments comprehensively demonstrated that MEX3A may promote glioma development through regulating cell proliferation, cell apoptosis, cell cycle, and cell migration. In vivo experiments also suggested the inhibition of glioma growth by MEX3A knockdown. Moreover, our mechanistic study identifies CCL2 as a potential downstream target of MEX3A, which possesses similar regulatory effects on glioma development with MEX3A and could attenuate the promotion of glioma induced by MEX3A overexpression. Overall, MEX3A was identified as a potential tumor promoter in glioma development and therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Chao Yang
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Haoqiang Zhan
- Department of Neurosurgery, The Six Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yiqing Zhao
- Department of Neurosurgery, TongJi hospital of TongJi Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
| | - Yasong Wu
- Department of Neurosurgery, TongJi hospital of TongJi Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130012, China
| | - Heping Wang
- Department of Neurosurgery, TongJi hospital of TongJi Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China.
| |
Collapse
|
13
|
Jasinski-Bergner S, Steven A, Seliger B. The Role of the RNA-Binding Protein Family MEX-3 in Tumorigenesis. Int J Mol Sci 2020; 21:ijms21155209. [PMID: 32717840 PMCID: PMC7432607 DOI: 10.3390/ijms21155209] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 12/16/2022] Open
Abstract
The muscle excess 3 (MEX-3) protein was first identified in Caenorhabditis elegans (C. elegans), and its respective homologues were also observed in vertebrates, including humans. It is a RNA-binding protein (RBP) with an additional ubiquitin E3 ligase function, which further acts as a post-transcriptional repressor through unknown mechanisms. In humans, MEX-3 proteins post-transcriptionally regulate a number of biological processes, including tumor immunological relevant ones. These have been shown to be involved in various diseases, including tumor diseases of distinct origins. This review provides information on the expression and function of the human MEX-3 family in healthy tissues, as well after malignant transformation. Indeed, the MEX-3 expression was shown to be deregulated in several cancers and to affect tumor biological functions, including apoptosis regulation, antigen processing, and presentation, thereby, contributing to the immune evasion of tumor cells. Furthermore, current research suggests MEX-3 proteins as putative markers for prognosis and as novel targets for the anti-cancer treatment.
Collapse
Affiliation(s)
| | | | - Barbara Seliger
- Correspondence: ; Tel.: +49-345-557-1357; Fax: +49-345-557-4055
| |
Collapse
|
14
|
Wang X, Shan YQ, Tan QQ, Tan CL, Zhang H, Liu JH, Ke NW, Chen YH, Liu XB. MEX3A knockdown inhibits the development of pancreatic ductal adenocarcinoma. Cancer Cell Int 2020; 20:63. [PMID: 32140076 PMCID: PMC7048143 DOI: 10.1186/s12935-020-1146-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/19/2020] [Indexed: 02/08/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDA) is one of the most serious causes of death in the world due to its high mortality and inefficacy treatments. MEX3A was first identified in nematodes and was associated with tumor formation and may promote cell proliferation and tumor metastasis. So far, nothing is known about the relationship between MEX3A and PDA. Methods In this study, the expression level of MEX3A in PDA tissues was measured by immunohistochemistry. The qRT-PCR and western blot were used to identify the constructed MEX3A knockdown cell lines, which was further used to construct mouse xenotransplantation models. Cell proliferation, colony formation, cell apoptosis and migration were detected by MTT, colony formation, flow cytometry and Transwell. Results This study showed that MEX3A expression is significantly upregulated in PDA and associated with tumor grade. Loss-of-function studies showed that downregulation of MEX3A could inhibit cell growth in vitro and in vivo. Moreover, it was demonstrated that knockdown of MEX3A in PDA cells promotes apoptosis by regulating apoptosis-related factors, and inhibits migration through influencing EMT. At the same time, the regulation of PDA progression by MEX3A involves changes in downstream signaling pathways including Akt, p-Akt, PIK3CA, CDK6 and MAPK9. Conclusions We proposed that MEX3A is associated with the prognosis and progression of PDA,which can be used as a potential therapeutic target.
Collapse
Affiliation(s)
- Xing Wang
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Yu-Qiang Shan
- 2Department of Hangzhou First People's Hospital, No. 261, Huansha Road, Hangzhou, 310006 Zhejiang China
| | - Qing-Quan Tan
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Chun-Lu Tan
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Hao Zhang
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Jin-Heng Liu
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Neng-Wen Ke
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Yong-Hua Chen
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| | - Xu-Bao Liu
- 1Department of Pancreatic Surgery, West China Hospital, Sichuan University, No 37 Guo Xue Alley, Chengdu, 610041 Sichuan China
| |
Collapse
|
15
|
Bufalieri F, Caimano M, Lospinoso Severini L, Basili I, Paglia F, Sampirisi L, Loricchio E, Petroni M, Canettieri G, Santoro A, D’Angelo L, Infante P, Di Marcotullio L. The RNA-Binding Ubiquitin Ligase MEX3A Affects Glioblastoma Tumorigenesis by Inducing Ubiquitylation and Degradation of RIG-I. Cancers (Basel) 2020; 12:cancers12020321. [PMID: 32019099 PMCID: PMC7072305 DOI: 10.3390/cancers12020321] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GB) is the most malignant primary brain tumor in humans, with an overall survival of approximatively 15 months. The molecular heterogeneity of GB, as well as its rapid progression, invasiveness and the occurrence of drug-resistant cancer stem cells, limits the efficacy of the current treatments. In order to develop an innovative therapeutic strategy, it is mandatory to identify and characterize new molecular players responsible for the GB malignant phenotype. In this study, the RNA-binding ubiquitin ligase MEX3A was selected from a gene expression analysis performed on publicly available datasets, to assess its biological and still-unknown activity in GB tumorigenesis. We find that MEX3A is strongly up-regulated in GB specimens, and this correlates with very low protein levels of RIG-I, a tumor suppressor involved in differentiation, apoptosis and innate immune response. We demonstrate that MEX3A binds RIG-I and induces its ubiquitylation and proteasome-dependent degradation. Further, the genetic depletion of MEX3A leads to an increase of RIG-I protein levels and results in the suppression of GB cell growth. Our findings unveil a novel molecular mechanism involved in GB tumorigenesis and suggest MEX3A and RIG-I as promising therapeutic targets in GB.
Collapse
Affiliation(s)
- Francesca Bufalieri
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
| | - Miriam Caimano
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
| | - Ludovica Lospinoso Severini
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
| | - Irene Basili
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
| | - Francesco Paglia
- Dipartimento di Neurologia e Psichiatria, Neurochirurgia, Sapienza University, Viale dell’Università 30, 00185 Rome, Italy; (F.P.); (L.S.); (A.S.); (L.D.)
| | - Luigi Sampirisi
- Dipartimento di Neurologia e Psichiatria, Neurochirurgia, Sapienza University, Viale dell’Università 30, 00185 Rome, Italy; (F.P.); (L.S.); (A.S.); (L.D.)
| | - Elena Loricchio
- Center for Life Nano Science (CLNS@Sapienza), Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| | - Marialaura Petroni
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti-Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Antonio Santoro
- Dipartimento di Neurologia e Psichiatria, Neurochirurgia, Sapienza University, Viale dell’Università 30, 00185 Rome, Italy; (F.P.); (L.S.); (A.S.); (L.D.)
| | - Luca D’Angelo
- Dipartimento di Neurologia e Psichiatria, Neurochirurgia, Sapienza University, Viale dell’Università 30, 00185 Rome, Italy; (F.P.); (L.S.); (A.S.); (L.D.)
| | - Paola Infante
- Center for Life Nano Science (CLNS@Sapienza), Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
- Correspondence: (P.I.); (L.D.M.); Tel.: +39-06-49255132 (P.I.); +39-06-49255657 (L.D.M.); Fax: +39-06-49255660 (L.D.M.)
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy; (F.B.); (M.C.); (L.L.S.); (I.B.); (M.P.); (G.C.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti-Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
- Correspondence: (P.I.); (L.D.M.); Tel.: +39-06-49255132 (P.I.); +39-06-49255657 (L.D.M.); Fax: +39-06-49255660 (L.D.M.)
| |
Collapse
|
16
|
Yang D, Jiao Y, Li Y, Fang X. Clinical characteristics and prognostic value of MEX3A mRNA in liver cancer. PeerJ 2020; 8:e8252. [PMID: 31998552 PMCID: PMC6979405 DOI: 10.7717/peerj.8252] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MEX3A is an RNA-binding proteins (RBPs) that promotes the proliferation, invasion, migration and viability of cancer cells. The aim of this study was to explore the clinicopathological characteristics and prognostic significance of MEX3A mRNA expression in liver cancer. METHODS RNA-Seq and clinical data were collected from The Cancer Genome Atlas (TCGA). Boxplots were used to represent discrete variables of MEX3A. Chi-square tests were used to analyze the correlation between clinical features and MEX3A expression. Receiver operating characteristic (ROC) curves were used to confirm diagnostic ability. Independent prognostic ability and values were assessed using Kaplan-Meier curves and Cox analysis. RESULTS We acquired MEX3A RNA-Seq from 50 normal liver tissues and 373 liver cancer patients along with clinical data. We found that MEX3A was up-regulated in liver cancer which increased according to histological grade (p < 0.001). MEX3A showed moderate diagnostic ability for liver cancer (AUC = 0.837). Kaplan-Meier curves and Cox analysis revealed that the high expression of MEX3A was significantly associated with poor survival (OS and RFS) (p < 0.001). Moreover, MEX3A was identified as an independent prognostic factor of liver cancer (p < 0.001). CONCLUSIONS MEX3A expression shows promise as an independent predictor of liver cancer prognosis.
Collapse
Affiliation(s)
- Dingquan Yang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanqing Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|