1
|
Chen Y, Chen Y, Li Q, Liu H, Han J, Zhang H, Cheng L, Lin G. Short C-terminal Musashi-1 proteins regulate pluripotency states in embryonic stem cells. Cell Rep 2023; 42:113308. [PMID: 37858462 DOI: 10.1016/j.celrep.2023.113308] [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: 05/10/2023] [Revised: 09/04/2023] [Accepted: 10/03/2023] [Indexed: 10/21/2023] Open
Abstract
The RNA-binding protein Musashi-1 (MSI1) regulates the proliferation and differentiation of adult stem cells. However, its role in embryonic stem cells (ESCs) and early embryonic development remains poorly understood. Here, we report the presence of short C-terminal MSI1 (MSI1-C) proteins in early mouse embryos and mouse ESCs, but not in human ESCs, under conventional culture conditions. In mouse embryos and mESCs, deletion of MSI1-C together with full-length MSI1 causes early embryonic developmental arrest and pluripotency dissolution. MSI1-C is induced upon naive induction and facilitates hESC naive pluripotency acquisition, elevating the pluripotency of primed hESCs toward a formative-like state. MSI1-C proteins are nuclear localized and bind to RNAs involved in DNA-damage repair (including MLH1, BRCA1, and MSH2), conferring on hESCs better survival in human-mouse interspecies cell competition and prolonged ability to form blastoids. This study identifies MSI1-C as an essential regulator in ESC pluripotency states and early embryonic development.
Collapse
Affiliation(s)
- Youwei Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical Center for Brain and Spinal Cord Research, Medical School, Tongji University, Shanghai, China
| | - Ying Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qianyan Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Huahua Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jiazhen Han
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hailin Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical Center for Brain and Spinal Cord Research, Medical School, Tongji University, Shanghai, China.
| | - Gufa Lin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China; Clinical Center for Brain and Spinal Cord Research, Medical School, Tongji University, Shanghai, China.
| |
Collapse
|
2
|
Fang T, Sun H, Sun X, He Y, Tang P, Gong L, Yu Z, Liu L, Xie S, Wang T, Xu Z, Yi S, An G, Xu Y, Zhu G, Qiu L, Hao M. Exosome miRNAs profiling in serum and prognostic evaluation in patients with multiple myeloma. BLOOD SCIENCE 2023; 5:196-208. [PMID: 37546707 PMCID: PMC10400059 DOI: 10.1097/bs9.0000000000000160] [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: 12/14/2022] [Accepted: 04/18/2023] [Indexed: 08/08/2023] Open
Abstract
MicroRNAs (MiRNAs) carried by exosomes play pivotal roles in the crosstalk between cell components in the tumor microenvironment. Our study aimed at identifying the expression profile of exosomal miRNAs (exo-miRNAs) in the serum of multiple myeloma (MM) patients and investigating the regulation networks and their potential functions by integrated bioinformatics analysis. Exosomes in serum from 19 newly diagnosed MM patients and 9 healthy donors were isolated and the miRNA profile was investigated by small RNA sequencing. Differential expression of exo-miRNAs was calculated and target genes of miRNAs were predicted. CytoHubba was applied to identify the hub miRNAs and core target genes. The LASSO Cox regression model was used to develop the prognostic model, and the ESTIMATE immune score was calculated to investigate the correlation between the model and immune status in MM patients. The top six hub differentially expressed serum exo-miRNAs were identified. 513 target genes of the six hub exo-miRNAs were confirmed to be differentially expressed in MM cells in the Zhan Myeloma microarray dataset. Functional enrichment analysis indicated that these target genes were mainly involved in mRNA splicing, cellular response to stress, and deubiquitination. 13 core exo-miRNA target genes were applied to create a novel prognostic signature to provide risk stratification for MM patients, which is associated with the immune microenvironment of MM patients. Our study comprehensively investigated the exo-miRNA profiles in MM patients. A novel prognostic signature was constructed to facilitate the risk stratification of MM patients with distinct outcomes.
Collapse
Affiliation(s)
- Teng Fang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Hao Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Xiyue Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yi He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Peixia Tang
- Hematology Department, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou 350001, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Shiyi Xie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Tingyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Zhenshu Xu
- Hematology Department, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou 350001, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Guoqing Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
- Gobroad Healthcare Group, Beijing 100072, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| |
Collapse
|
3
|
Getu AA, Zhou M, Cheng SY, Tan M. The mammalian Sterile 20-like kinase 4 (MST4) signaling in tumor progression: Implications for therapy. Cancer Lett 2023; 563:216183. [PMID: 37094736 PMCID: PMC10642761 DOI: 10.1016/j.canlet.2023.216183] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 04/26/2023]
Abstract
Cancer is a leading cause of death in humans, with a complex and dynamic nature that makes it challenging to fully comprehend and treat. The Mammalian Sterile 20-Like Kinase 4 (MST4 or STK26) is a serine/threonine-protein kinase that plays a crucial role in cell migration and polarity in both normal and tumor cells via activation of intracellular signaling molecules and pathways. MST4 is involved in tumor cell proliferation, migration and invasion, epithelial-mesenchymal transition (EMT), survival, and cancer metastasis through modulation of downstream signaling pathways including the extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) pathways. Additionally, MST4 interacts with programmed cell death 10 (PDCD10) to promote tumor proliferation and migration. MST4 phosphorylates autophagy related 4B cysteine peptidase (ATG4B) to mediate autophagy signaling, promote tumor cell survival and proliferation, and contribute to treatment resistance. Taken together, MST4 functions as an oncogene and is a promising therapeutic target which deserves further exploration.
Collapse
Affiliation(s)
- Ayechew A Getu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Ming Zhou
- Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, Lou & Jean Malnati Brain Tumor Institute at Northwestern Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Ming Tan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 406040, Taiwan; Institute of Biochemistry & Molecular Biology, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
4
|
The Role of Different Types of microRNA in the Pathogenesis of Breast and Prostate Cancer. Int J Mol Sci 2023; 24:ijms24031980. [PMID: 36768298 PMCID: PMC9916830 DOI: 10.3390/ijms24031980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Micro ribonucleic acids (microRNAs or miRNAs) form a distinct subtype of non-coding RNA and are widely recognized as one of the most significant gene expression regulators in mammalian cells. Mechanistically, the regulation occurs through microRNA binding with its response elements in the 3'-untranslated region of target messenger RNAs (mRNAs), resulting in the post-transcriptional silencing of genes, expressing target mRNAs. Compared to small interfering RNAs, microRNAs have more complex regulatory patterns, making them suitable for fine-tuning gene expressions in different tissues. Dysregulation of microRNAs is well known as one of the causative factors in malignant cell growth. Today, there are numerous data points regarding microRNAs in different cancer transcriptomes, the specificity of microRNA expression changes in various tissues, and the predictive value of specific microRNAs as cancer biomarkers. Breast cancer (BCa) is the most common cancer in women worldwide and seriously impairs patients' physical health. Its incidence has been predicted to rise further. Mounting evidence indicates that microRNAs play key roles in tumorigenesis and development. Prostate cancer (PCa) is one of the most commonly diagnosed cancers in men. Different microRNAs play an important role in PCa. Early diagnosis of BCa and PCa using microRNAs is very useful for improving individual outcomes in the framework of predictive, preventive, and personalized (3P) medicine, thereby reducing the economic burden. This article reviews the roles of different types of microRNA in BCa and PCa progression.
Collapse
|
5
|
Arora R, Kim JH, Getu AA, Angajala A, Chen YL, Wang B, Kahn AG, Chen H, Reshi L, Lu J, Zhang W, Zhou M, Tan M. MST4: A Potential Oncogene and Therapeutic Target in Breast Cancer. Cells 2022; 11:cells11244057. [PMID: 36552828 PMCID: PMC9777386 DOI: 10.3390/cells11244057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The mammalian STE 20-like protein kinase 4 (MST4) gene is highly expressed in several cancer types, but little is known about the role of MST4 in breast cancer, and the function of MST4 during epithelial-mesenchymal transition (EMT) has not been fully elucidated. Here we report that overexpression of MST4 in breast cancer results in enhanced cell growth, migration, and invasion, whereas inhibition of MST4 expression significantly attenuates these properties. Further study shows that MST4 promotes EMT by activating Akt and its downstream signaling molecules such as E-cadherin/N-cadherin, Snail, and Slug. MST4 also activates AKT and its downstream pro-survival pathway. Furthermore, by analyzing breast cancer patient tissue microarray and silicon datasets, we found that MST4 expression is much higher in breast tumor tissue compared to normal tissue, and significantly correlates with cancer stage, lymph node metastasis and a poor overall survival rate (p < 0.05). Taken together, our findings demonstrate the oncogenic potential of MST4 in breast cancer, highlighting its role in cancer cell proliferation, migration/invasion, survival, and EMT, suggesting a possibility that MST4 may serve as a novel therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Ritu Arora
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Jin-Hwan Kim
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
- Markey Cancer Center, Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40508, USA
| | - Ayechew A. Getu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar P.O. Box 196, Ethiopia
| | - Anusha Angajala
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | - Yih-Lin Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
| | - Bin Wang
- Department of Mathematics and Statistics, University of South Alabama, Mobile, AL 36688, USA
| | - Andrea G. Kahn
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hong Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
| | - Latif Reshi
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
| | - Jianrong Lu
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Wenling Zhang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Ming Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 410013, China
| | - Ming Tan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 406040, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
- Correspondence:
| |
Collapse
|
6
|
Sulforaphane Suppresses the Nicotine-Induced Expression of the Matrix Metalloproteinase-9 via Inhibiting ROS-Mediated AP-1 and NF-κB Signaling in Human Gastric Cancer Cells. Int J Mol Sci 2022; 23:ijms23095172. [PMID: 35563563 PMCID: PMC9099819 DOI: 10.3390/ijms23095172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 02/01/2023] Open
Abstract
Sulforaphane, a natural phytochemical compound found in various cruciferous vegetables, has been discovered to present anti-cancer properties. Matrix metalloproteinase-9 (MMP-9) plays a crucial role in gastric cancer metastasis. However, the role of sulforaphane in MMP-9 expression in gastric cancer is not yet defined. Nicotine, a psychoactive alkaloid found in tobacco, is associated with the development of gastric cancer. Here, we found that sulforaphane suppresses the nicotine-mediated induction of MMP-9 in human gastric cancer cells. We discovered that reactive oxygen species (ROS) and MAPKs (p38 MAPK, Erk1/2) are involved in nicotine-induced MMP-9 expression. AP-1 and NF-κB are the critical transcription factors in MMP-9 expression. ROS/MAPK (p38 MAPK, Erk1/2) and ROS functioned as upstream signaling of AP-1 and NF-κB, respectively. Sulforaphane suppresses the nicotine-induced MMP-9 by inhibiting ROS-mediated MAPK (p38 MAPK, Erk1/2)/AP-1 and ROS-mediated NF-κB signaling axes, which in turn inhibit cell invasion in human gastric cancer AGS cells. Therefore, the current study provides valuable evidence for developing sulforaphane as a new anti-invasion strategy for human gastric cancer therapy.
Collapse
|
7
|
Gurer T, Aytekin A, Caki E, Gezici S. miR-485-3p and miR-4728-5p as Tumor Suppressors in Pathogenesis of Colorectal Cancer. Mol Biol 2022. [DOI: 10.1134/s0026893322030062] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
8
|
Splice and Dice: Intronic microRNAs, Splicing and Cancer. Biomedicines 2021; 9:biomedicines9091268. [PMID: 34572454 PMCID: PMC8465124 DOI: 10.3390/biomedicines9091268] [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: 08/30/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/17/2022] Open
Abstract
Introns span only a quarter of the human genome, yet they host around 60% of all known microRNAs. Emerging evidence indicates the adaptive advantage of microRNAs residing within introns is attributed to their complex co-regulation with transcription and alternative splicing of their host genes. Intronic microRNAs are often co-expressed with their host genes, thereby providing functional synergism or antagonism that is exploited or decoupled in cancer. Additionally, intronic microRNA biogenesis and the alternative splicing of host transcript are co-regulated and intertwined. The importance of intronic microRNAs is under-recognized in relation to the pathogenesis of cancer.
Collapse
|
9
|
Caputo M, Cansby E, Kumari S, Kurhe Y, Nair S, Ståhlman M, Kulkarni NM, Borén J, Marschall HU, Blüher M, Mahlapuu M. STE20-Type Protein Kinase MST4 Controls NAFLD Progression by Regulating Lipid Droplet Dynamics and Metabolic Stress in Hepatocytes. Hepatol Commun 2021; 5:1183-1200. [PMID: 34278168 PMCID: PMC8279465 DOI: 10.1002/hep4.1702] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/08/2021] [Accepted: 02/14/2021] [Indexed: 12/27/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as a leading cause of chronic liver disease worldwide, primarily because of the massive global increase in obesity. Despite intense research efforts in this field, the factors that govern the initiation and subsequent progression of NAFLD are poorly understood, which hampers the development of diagnostic tools and effective therapies in this area of high unmet medical need. Here we describe a regulator in molecular pathogenesis of NAFLD: STE20-type protein kinase MST4. We found that MST4 expression in human liver biopsies was positively correlated with the key features of NAFLD (i.e., hepatic steatosis, lobular inflammation, and hepatocellular ballooning). Furthermore, the silencing of MST4 attenuated lipid accumulation in human hepatocytes by stimulating β-oxidation and triacylglycerol secretion, while inhibiting fatty acid influx and lipid synthesis. Conversely, overexpression of MST4 in human hepatocytes exacerbated fat deposition by suppressing mitochondrial fatty acid oxidation and triacylglycerol efflux, while enhancing lipogenesis. In parallel to these reciprocal alterations in lipid storage, we detected substantially decreased or aggravated oxidative/endoplasmic reticulum stress in human hepatocytes with reduced or increased MST4 levels, respectively. Interestingly, MST4 protein was predominantly associated with intracellular lipid droplets in both human and rodent hepatocytes. Conclusion: Together, our results suggest that hepatic lipid droplet-decorating protein MST4 is a critical regulatory node governing susceptibility to NAFLD and warrant future investigations to address the therapeutic potential of MST4 antagonism as a strategy to prevent or mitigate the development and aggravation of this disease.
Collapse
Affiliation(s)
- Mara Caputo
- Department of Chemistry and Molecular BiologyUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Emmelie Cansby
- Department of Chemistry and Molecular BiologyUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Sima Kumari
- Department of Chemistry and Molecular BiologyUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Yeshwant Kurhe
- Department of Chemistry and Molecular BiologyUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Syam Nair
- Institute of Neuroscience and Physiology, and Institute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine/Wallenberg LaboratoryInstitute of MedicineUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Nagaraj M Kulkarni
- Department of Chemistry and Molecular BiologyUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg LaboratoryInstitute of MedicineUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg LaboratoryInstitute of MedicineUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| | | | - Margit Mahlapuu
- Department of Chemistry and Molecular BiologyUniversity of Gothenburg and Sahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
10
|
Zhang X, Song L, Huang Y, Han S, Hou M, Li H. Downregulation of MST4 Underlies a Novel Inhibitory Role of MicroRNA Let-7a in the Progression of Retinoblastoma. Invest Ophthalmol Vis Sci 2021; 61:28. [PMID: 32539131 PMCID: PMC7415300 DOI: 10.1167/iovs.61.6.28] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose Retinoblastoma (RB) is the most common intraocular malignancy in children. Deregulation of several microRNAs (miRNAs) has been identified in RB. However, the specific effect of let-7a on RB remains unclear. The present study aims to explore the effect of let-7a on malignant biological behaviors of RB cells and angiogenesis in RB. Methods The expressions of let-7a and mammalian sterile-20 like kinase 4 (MST4) in RB were determined with the use of real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Next, in order to explore effects of let-7a and MST4 on RB cellular functions, RB cells were transfected with let-7a-mimic, let-7a inhibitor, si-MST4, or co-transfected with let-7a-mimic and oe-MST4 plasmids. Subsequently, the interaction among let-7a, MST4, and the MAPK signaling pathway was evaluated by RT-qPCR, dual-luciferase reporter gene assay, and Western blot analysis. Finally, the effects of let-7a and MST4 were further confirmed in vivo by injecting nude mice with RB cells stably expressing let-7a agomir or sh-MST4. Results Rb tissues and cells presented with downregulated Let-7a and upregulated MST4. Let-7a negatively targeted MST4 to block the activation of the MAPK signaling pathway. Upregulation of let-7a promoted apoptosis, and facilitated proliferation, angiogenesis, migration, and invasion of RB cells by decreasing MST4. Elevation of let-7a or silencing MST4 restricted angiogenesis and tumorigenesis in RB mice. Conclusions Taken together, let-7a inhibits angiogenesis in RB by silencing MST4 and inhibiting the MAPK signaling pathway.
Collapse
|
11
|
Yu H, Zhang W, Han P, Yang B, Feng X, Zhou P, Zhu X, Zhou B, Chen W, Qian J, Yu J. MST4 Regulates Epithelial-Mesenchymal Transition of Choriocarcinoma by Mediating TGF-β1 Expression. Onco Targets Ther 2020; 13:11935-11946. [PMID: 33244239 PMCID: PMC7683511 DOI: 10.2147/ott.s269168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background Mammalian Ste20-like kinase 4 (MST4), also known as serine/threonine kinase 26 (STK26), promotes development of several cancers and is found to be highly expressed in the placenta. However, in choriocarcinoma that originated from the placenta, the expression of MST4 was undetermined and its mechanism was unknown. In this study, the expression of MST4 in choriocarcinoma as well as the underlying mechanism was explored. Purpose To detect the expression of MST4 in patient samples and mechanism of mediating EMT by MST4 in choriocarcinoma. Patients and Methods The metastatic lesions of choriocarcinoma (n=17) and volunteer villus (n=17) were collected to determine MST4 expression using immunohistochemistry and H&E staining. We use siRNA and lentiviral vector to knockdown MST4 and use plasmid to overexpress MST4 in choriocarcinoma. Then, we apply real-time polymerase chain reaction (RT-PCR), Western blot assay and immunofluorescence assay to detect target protein expressions. Cell invasion and migration and cell proliferation were detected by transwell assay and wound healing assay and CCK-8 and cell colony formation. Results MST4 is lowly expressed in the metastatic lesions of choriocarcinoma patients when compared with normal villus. Knockdown of MST4 activated epithelial–mesenchymal transition (EMT) process, significantly increasing the ability of invasion and migration in choriocarcinoma cell lines (JAR and JEG-3). In contrast, the EMT process was restrained in choriocarcinoma cell lines with overexpressed MST4. Meanwhile, genome-wide gene expression array, Western blot and ELISA revealed that tumor growth factor-beta 1 (TGF-β1) has significantly increased. The EMT process and metastatic prompting biofunction were reversed after using TGF-β1 inhibitor (LY364947) in the choriocarcinoma cell lines with MST4 knockdown. Conclusion Our studies demonstrated that MST4 was lowly expressed in patient samples. Additionally, JAR and JEG-3 increase cell invasion and migration ability while there is no influence on cell proliferation with MST4 knockdown. Conversely, the metastatic ability of JAR and JEG-3 was decreased with overexpressed MST4. Moreover, TGF-β1 was a key factor after MST4 knockdown. In conclusion, MST4 affects choriocarcinoma EMT by mediating TGF-β1 expression.
Collapse
Affiliation(s)
- Hanxi Yu
- Department of Gynecology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Weichen Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Peilin Han
- Department of Gynecology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Beng Yang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Xiaode Feng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Ping Zhou
- Department of Gynecology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Xiaoxu Zhu
- Department of Gynecology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Bingqian Zhou
- Department of Gynecology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Pujiang 322200, People's Republic of China
| | - Wei Chen
- Department of Medical Oncology, Tongde Hospital of Zhejiang Province, Hangzhou 310012, People's Republic of China
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| | - Jun Yu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery First Affiliated Hospital, The First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou 310006, People's Republic of China
| |
Collapse
|
12
|
Hu Y, Zhang X, Gong C, Li J. Aberrant expression of miR-4728 in patients with non-small cell lung cancer and its regulatory effects on tumor progression in tumor cells. Exp Ther Med 2020; 20:15. [PMID: 32934680 PMCID: PMC7471878 DOI: 10.3892/etm.2020.9141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common malignant tumor with poor prognosis and an increasing number of cases. MicroRNA (miR)-4728 is related with the progression of various types of cancer, and is dysregulated in NSCLC, which indicates that miR-4728 may serve as a biomarker for NSCLC. The present study aimed to investigate the clinical significance of miR-4728 in NSCLC diagnosis and prognosis, and to explore the biological function of miR-4728 in NSCLC progression. Serum and tissue samples were collected from 122 patients with NSCLC. By conducting reverse transcription-quantitative PCR, the Cell Counting Kit-8 assay and Transwell assays, the expression of miR-4728 and its effect on NSCLC cell proliferation, migration and invasion were investigated. The diagnostic value of miR-4728 was evaluated by plotting a receiver operating characteristic curve, and Kaplan-Meier and Cox regression analyses were conducted to assess the prognostic value of miR-4728. miR-4728 was significantly downregulated in NSCLC serum and tissue samples compared with healthy controls, with a relatively high diagnostic accuracy and ability to predict poor overall survival time in patients with NSCLC. By conducting gain- and loss-of-function experiments, the results indicated that miR-4728 knockdown significantly promoted NSCLC cell proliferation, migration and invasion compared with the inhibitor negative control (NC) group. By contrast, miR-4728 overexpression displayed the opposite effect on NSCLC cell proliferation, migration and invasion. The present study indicated that miR-4728 was downregulated in NSCLC and may serve as a candidate diagnostic and prognostic biomarker. NSCLC cell proliferation, migration and invasion were inhibited by miR-4728 overexpression compared with the mimic NC group, which suggested that miR-4728 may serve as a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Ying Hu
- Department of Blood Transfusion, Qilu Hospital Huantai Branch, Zibo, Shandong 256400, P.R. China
| | - Xinfang Zhang
- Clinical Laboratory, Qilu Hospital Huantai Branch, Zibo, Shandong 256400, P.R. China
| | - Cuixue Gong
- Outpatient Dressing Room, Zibo Central Hospital, Zibo, Shandong 255000, P.R. China
| | - Jianzhao Li
- Department of Pathology, Zibo Central Hospital, Zibo, Shandong 255000, P.R. China
| |
Collapse
|
13
|
miR-6086 inhibits ovarian cancer angiogenesis by downregulating the OC2/VEGFA/EGFL6 axis. Cell Death Dis 2020; 11:345. [PMID: 32393810 PMCID: PMC7214437 DOI: 10.1038/s41419-020-2501-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
miRNAs have emerged as a pivotal component of gene regulatory networks, mediating cytokines secretion, cell cycle, and differentiation regulation. However, how miRNAs collaborate with transcription factors and downstream effector proteins that determine the fate of ovarian cancer cells remains to be understood, especially regarding to mechanism of tumor angiogenesis regulation. Based on the qRT-PCR and IHC analysis, we found that miR-6086 was maintained a very low level both in ovarian cancer cell lines and tissues. Further, we identified OC2 and EGFL6 as the direct targets of miR-6086 by luciferase assay and we observed an inverse relationship between the expression of miR-6086 and the OC2/VEGFA/EGFL6 axis. The Western blotting analysis suggested that OC2 could directly upregulate VEGFA and indirectly up-regulate EGFL6 through VEGFA. Moreover, miR-6086 could indirectly downregulate VEGFA through OC2. In addition, miR-6086, siOC2 and siEGFL6 could negatively regulate the tumor growth and angiogenesis of ovarian cancer (Skov3) in the animal studies, with the inhibition rates of 77.07%, 69.89%, and 73.62%, respectively (**p < 0.01). Moreover, the tumor cell proliferation, migration, and invasion of ovarian cancer cell lines (Caov3 and Skov3) and vascular formation (HUVECs) were significantly suppressed in vitro, by decreasing the AKT/MAPK pathways (*p < 0.05). Taken together, our results reveal that miR-6086 can suppress the angiogenesis networks in ovarian cancer by down-regulating the OC2/VEGFA/EGFL6 axis, directly or indirectly, which may provide potential targets for tumor therapeutics.
Collapse
|
14
|
Søkilde R, Persson H, Ehinger A, Pirona AC, Fernö M, Hegardt C, Larsson C, Loman N, Malmberg M, Rydén L, Saal L, Borg Å, Vallon-Christerson J, Rovira C. Refinement of breast cancer molecular classification by miRNA expression profiles. BMC Genomics 2019; 20:503. [PMID: 31208318 PMCID: PMC6580620 DOI: 10.1186/s12864-019-5887-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/06/2019] [Indexed: 02/06/2023] Open
Abstract
Background Accurate classification of breast cancer using gene expression profiles has contributed to a better understanding of the biological mechanisms behind the disease and has paved the way for better prognostication and treatment prediction. Results We found that miRNA profiles largely recapitulate intrinsic subtypes. In the case of HER2-enriched tumors a small set of miRNAs including the HER2-encoded mir-4728 identifies the group with very high specificity. We also identified differential expression of the miR-99a/let-7c/miR-125b miRNA cluster as a marker for separation of the Luminal A and B subtypes. High expression of this miRNA cluster is linked to better overall survival among patients with Luminal A tumors. Correlation between the miRNA cluster and their precursor LINC00478 is highly significant suggesting that its expression could help improve the accuracy of present day’s signatures. Conclusions We show here that miRNA expression can be translated into mRNA profiles and that the inclusion of miRNA information facilitates the molecular diagnosis of specific subtypes, in particular the clinically relevant sub-classification of luminal tumors. Electronic supplementary material The online version of this article (10.1186/s12864-019-5887-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rolf Søkilde
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden
| | - Helena Persson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden
| | - Anna Ehinger
- Clinical Pathology, Laboratory Medicine, Skåne University Hospital, Lund, Sweden
| | - Anna Chiara Pirona
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden.,German Cancer Research Center DKFZ, Division of Functional Genome Analysis, Heidelberg, Germany
| | - Mårten Fernö
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden
| | - Cecilia Hegardt
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden
| | - Christer Larsson
- Division of Translational Cancer Research, Lund University, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden
| | - Niklas Loman
- Division of Oncology, Skåne University Hospital, Lund, Sweden
| | - Martin Malmberg
- Division of Oncology, Skåne University Hospital, Lund, Sweden
| | - Lisa Rydén
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Lao Saal
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden
| | - Johan Vallon-Christerson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden
| | - Carlos Rovira
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Scheelevägen 2, 223 81, Lund, Sweden. .,BioCARE, Strategic Cancer Research Program, Lund, Sweden.
| |
Collapse
|
15
|
Intronic miR-744 Inhibits Glioblastoma Migration by Functionally Antagonizing Its Host Gene MAP2K4. Cancers (Basel) 2018; 10:cancers10110400. [PMID: 30366472 PMCID: PMC6266622 DOI: 10.3390/cancers10110400] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/16/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
Background: The second intron of Mitogen-Activated Protein Kinase Kinase 4 (MAP2K4), an important hub in the pro-invasive MAPK pathway, harbors miR-744. There is accumulating evidence that intronic micro-RNAs (miRNAs) are capable of either supporting or restraining functional pathways of their host genes, thereby creating intricate regulative networks. We thus hypothesized that miR-744 regulates glioma migration by interacting with its host’s pathways. Methods: Patients’ tumor specimens were obtained stereotactically. MiR-744 was overexpressed in U87, T98G, and primary glioblastoma (GBM) cell lines. Cell mobility was studied using migration and Boyden chamber assays. Protein and mRNA expression was quantified by SDS-PAGE and qRT-PCR. Interactions of miR-744 and 3’UTRs were analyzed by luciferase reporter assays, and SMAD2/3, p38, and beta-Catenin activities by TOP/FOPflash reporter gene assays. Results: As compared to a normal brain, miR-744 levels were dramatically decreased in GBM samples and in primary GBM cell lines. Astrocytoma WHO grade II/III exhibited intermediate expression levels. Re-expression of miR-744 in U87, T98G, and primary GBM cell lines induced focal growth and impaired cell mobility. Luciferase activity of 3’UTR reporter constructs revealed the pro-invasive factors TGFB1 and DVL2 as direct targets of miR-744. Re-expression of miR-744 reduced levels of TGFB1, DVL2, and the host MAP2K4, and mitigated activity of TGFB1 and DVL2 downstream targets SMAD2/3 and beta-Catenin. TGFB1 knock-down repressed MAP2K4 expression. Conclusion: MiR-744 acts as an intrinsic brake on its host. It impedes MAP2K4 functional pathways through simultaneously targeting SMAD-, beta-Catenin, and MAPK signaling networks, thereby strongly mitigating pro-migratory effects of MAP2K4. MiR-744 is strongly repressed in glioma, and its re-expression might attenuate tumor invasiveness.
Collapse
|
16
|
Wang W, Lin P, Yao H, Jia X, Sun J, Feng L. MicroRNA-4728 serves as a suppressor and antagonist of oncogenic MAPK in Burkitt lymphoma. Saudi J Biol Sci 2018; 25:982-985. [PMID: 30108451 PMCID: PMC6088107 DOI: 10.1016/j.sjbs.2018.05.010] [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: 03/29/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 11/18/2022] Open
Abstract
Objective This study identified the biological role of miR-4728 in Burkitt lymphoma (BL) process. Methods Ramos cells were used to analyze MicroRNA-4728 (miR-4728) biological functions. MiR-4728 expression was investigated in 14 randomly chosen tumor tissues and 12 noncancerous tissues by qRT-PCR. Cyquant assay was used to monitor cell proliferation. Colony formation assay was performed to study the effectiveness of miR-4728 on the proliferation of cells. The effects of miR-4728 on MAPK signaling pathway were detected by luciferase reporter assay. The significance of differences between groups were evaluated by SPSS. Results In this study, MiRNA-4728 was observed to down-regulated in BL tissues compared to the noncancerous tissues. Additionally, miR-4728 inhibited Ramos cell proliferation. Moreover, miR-4728 overexpression also decreased the MAPK signaling activity. Conclusion Our results suggested that miR-4728 serves as a suppressor and antagonist of oncogenic MAPK in Burkitt lymphoma. The appropriate regulation of miR-4728 might be vital to improve BL treatment.
Collapse
Affiliation(s)
- Wei Wang
- Baoding First Central Hospital, Baoding 071000, PR China
| | - Peng Lin
- Baoding First Central Hospital, Baoding 071000, PR China
| | - Haiying Yao
- Baoding First Central Hospital, Baoding 071000, PR China
| | - Xi Jia
- Baoding First Central Hospital, Baoding 071000, PR China
| | - Jirui Sun
- Baoding First Central Hospital, Baoding 071000, PR China
| | - Lin Feng
- Pavlovian Medical College, St. Petersburg 190121, Russia
| |
Collapse
|
17
|
Liu Z, Zhang J, Gao J, Li Y. MicroRNA-4728 mediated regulation of MAPK oncogenic signaling in papillary thyroid carcinoma. Saudi J Biol Sci 2018; 25:986-990. [PMID: 30108452 PMCID: PMC6088101 DOI: 10.1016/j.sjbs.2018.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 12/24/2022] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer that accounts for 85% of thyroid cancers. MicroRNAs (miRNAs) have been reported to play important roles in the biological processes in cancer. In this study, we analyzed the biological role of miR-4728 in human PTC process in human PTC cell lines in vitro. MiRNA-4728 was observed to down-regulated in human PTC tissues and PTC cell lines. Additionally, miR-4728 inhibited PTC cell proliferation. Further study demonstrated SOS1 was repressed by miR-4728 and overexpression of miR-4728 down-regulated both the mRNA and protein levels of SOS1. Moreover, miR-4728 overexpression also decreased the MAPK signaling activity. These observations suggested that miR-4728 could inhibit the process of human PTC through regulating MAPK signaling pathway. And, appropriate regulation of miR-4728 might be vital to improve human PTC treatment.
Collapse
Affiliation(s)
- Zhibao Liu
- Hebei Cangzhou Central Hospital, Cangzhou 061001, PR China
| | - Jinghua Zhang
- Hebei Cangzhou Central Hospital, Cangzhou 061001, PR China
| | - Jinghua Gao
- Hebei Cangzhou Central Hospital, Cangzhou 061001, PR China
| | - Yunnan Li
- Massey University, Private Bag 11, 222 Palmerston North, 4442, New Zealand
| |
Collapse
|
18
|
Marchese S, Polo A, Ariano A, Velotto S, Costantini S, Severino L. Aflatoxin B1 and M1: Biological Properties and Their Involvement in Cancer Development. Toxins (Basel) 2018; 10:E214. [PMID: 29794965 PMCID: PMC6024316 DOI: 10.3390/toxins10060214] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 12/12/2022] Open
Abstract
Aflatoxins are fungal metabolites found in feeds and foods. When the ruminants eat feedstuffs containing Aflatoxin B1 (AFB1), this toxin is metabolized and Aflatoxin M1 (AFM1) is excreted in milk. International Agency for Research on Cancer (IARC) classified AFB1 and AFM1 as human carcinogens belonging to Group 1 and Group 2B, respectively, with the formation of DNA adducts. In the last years, some epidemiological studies were conducted on cancer patients aimed to evaluate the effects of AFB1 and AFM1 exposure on cancer cells in order to verify the correlation between toxin exposure and cancer cell proliferation and invasion. In this review, we summarize the activation pathways of AFB1 and AFM1 and the data already reported in literature about their correlation with cancer development and progression. Moreover, considering that few data are still reported about what genes/proteins/miRNAs can be used as damage markers due to AFB1 and AFM1 exposure, we performed a bioinformatic analysis based on interaction network and miRNA predictions to identify a panel of genes/proteins/miRNAs that can be used as targets in further studies for evaluating the effects of the damages induced by AFB1 and AFM1 and their capacity to induce cancer initiation.
Collapse
Affiliation(s)
- Silvia Marchese
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| | - Andrea Polo
- Unità di Farmacologia Sperimentale, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", 80131 Napoli, Italy.
| | - Andrea Ariano
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| | - Salvatore Velotto
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| | - Susan Costantini
- Unità di Farmacologia Sperimentale, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", 80131 Napoli, Italy.
| | - Lorella Severino
- Unità di Farmacologia e Tossicologia-Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli "Federico II", 80138 Napoli, Italy.
| |
Collapse
|
19
|
Slattery ML, Mullany LE, Sakoda LC, Wolff RK, Samowitz WS, Herrick JS. The MAPK-Signaling Pathway in Colorectal Cancer: Dysregulated Genes and Their Association With MicroRNAs. Cancer Inform 2018; 17:1176935118766522. [PMID: 29636593 PMCID: PMC5888819 DOI: 10.1177/1176935118766522] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Indexed: 12/11/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways regulate many cellular functions including cell proliferation and apoptosis. We examined associations of differential gene and microRNA (miRNA) expression between carcinoma and paired normal mucosa for 241 genes in the KEGG-identified MAPK-signaling pathway among 217 colorectal cancer (CRC) cases. Gene expression data (RNA-Seq) and miRNA expression data (Agilent Human miRNA Microarray V19.0; Agilent Technologies Inc., Santa Clara, CA, USA) were analyzed. We first identified genes most strongly associated with CRC using a fold change (FC) of >1.50 or <0.67) that were statistically significant after adjustment for multiple comparisons. We then determined miRNAs associated with dysregulated genes and through miRNA:mRNA (messenger RNA) seed region matches discerned genes with a greater likelihood of having a direct biological association. Ninety-nine genes had a meaningful FC for all CRC, microsatellite unstable–specific tumors, or microsatellite stable–specific tumors. Thirteen dysregulated genes were associated with miRNAs, totaling 68 miRNA:mRNA associations. Thirteen of the miRNA:mRNA associations had seed region matches where the differential expression between the miRNA and mRNA was inversely related suggesting a direct association as a result of their binding. Several direct associations, upstream of ERK1/ERK2, JNK, and p38, were found for PDGFRA with 7 miRNAs; RASGRP3 and PRKCB with miR-203a; and TGFBR1 with miR-6071 and miR-2117. Other associations between miRNAs and mRNAs are most likely indirect, resulting from feedback and feed forward loops. Our results suggest that miRNAs may alter MAPK signaling through direct binding with key genes in this pathway. We encourage others to validate results in targeted CRC experiments that can help solidify important therapeutic targets.
Collapse
Affiliation(s)
| | - Lila E Mullany
- School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Roger K Wolff
- School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Wade S Samowitz
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | | |
Collapse
|
20
|
Coamplification of miR-4728 protects HER2-amplified breast cancers from targeted therapy. Proc Natl Acad Sci U S A 2018; 115:E2594-E2603. [PMID: 29476008 PMCID: PMC5856537 DOI: 10.1073/pnas.1717820115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In HER2-amplified breast cancers, HER2 inhibitors have been very successful as adjuvant therapy but not as monotherapy. Here, we demonstrate that coamplification of a HER2 intronic miRNA causes intrinsic resistance to HER2 inhibitors by indirectly down-regulating the pro-apoptotic NOXA. Importantly, coinhibition with MCL-1 inhibitors overcomes this resistance. HER2 (ERBB2) amplification is a driving oncogenic event in breast cancer. Clinical trials have consistently shown the benefit of HER2 inhibitors (HER2i) in treating patients with both local and advanced HER2+ breast cancer. Despite this benefit, their efficacy as single agents is limited, unlike the robust responses to other receptor tyrosine kinase inhibitors like EGFR inhibitors in EGFR-mutant lung cancer. Interestingly, the lack of HER2i efficacy occurs despite sufficient intracellular signaling shutdown following HER2i treatment. Exploring possible intrinsic causes for this lack of response, we uncovered remarkably depressed levels of NOXA, an endogenous inhibitor of the antiapoptotic MCL-1, in HER2-amplified breast cancer. Upon investigation of the mechanism leading to low NOXA, we identified a micro-RNA encoded in an intron of HER2, termed miR-4728, that targets the mRNA of the Estrogen Receptor α (ESR1). Reduced ESR1 expression in turn prevents ERα-mediated transcription of NOXA, mitigating apoptosis following treatment with the HER2i lapatinib. Importantly, resistance can be overcome with pharmacological inhibition of MCL-1. More generally, while many cancers like EGFR-mutant lung cancer are driven by activated kinases that when drugged lead to robust monotherapeutic responses, we demonstrate that the efficacy of targeted therapies directed against oncogenes active through focal amplification may be mitigated by coamplified genes.
Collapse
|
21
|
Zhang Y, Zhang Y, Chen M, Liu C, Xiang C. DUSP1 is involved in the progression of small cell carcinoma of the prostate. Saudi J Biol Sci 2017; 25:858-862. [PMID: 30108432 PMCID: PMC6088105 DOI: 10.1016/j.sjbs.2017.09.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/28/2017] [Accepted: 09/30/2017] [Indexed: 02/07/2023] Open
Abstract
Small cell carcinoma of the prostate (SCCP) is a rare and the most aggressive variant of prostate cancer. There is no effective cure or treatment for SCCP. Therefore, there is an urgent need for new therapy to improve the prognosis of patients with SCCP. DUSP1 is a dual specific phosphatase with an increasingly recognized in tumor biology. Altered expression of DUSP1 induced changes in the expression of genes involved in various biological pathways, including cell-cell signaling and angiogenesis. To understand more about the role of DUSP1 in SCCP, we evaluated the biological function and associated regulatory mechanism of DUSP1. In this study, DUSP1 was significantly down-regulated in human SCCP compared with the non-carcinoma tissues (P < 0.05). Overexpression of DUSP1 was found to suppress MAPK signaling and cell proliferation in PC-3 cells. Additionally, silencing of DUSP1 enhanced MAPK signaling and PC-3 cell proliferation. Moreover, it was observed that DUSP1 blocked the phosphorylation of p38 MAPK induced by anisomycin. Taken together, this investigation suggests that DUSP1 is involved in the progression of SCCP and may provide a new therapeutic target for SCCP treatment.
Collapse
Affiliation(s)
- Yajing Zhang
- The First Hospital of Shijiazhuang, Shijiangzhuang 050011, PR China
| | - Yan Zhang
- The First Hospital of Shijiazhuang, Shijiangzhuang 050011, PR China
| | - Meng Chen
- Department of Rheumatology, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Ci Liu
- The First Hospital of Shijiazhuang, Shijiangzhuang 050011, PR China
| | - Cheng Xiang
- The First Hospital of Shijiazhuang, Shijiangzhuang 050011, PR China
| |
Collapse
|
22
|
Lu M, Wang T, He M, Cheng W, Yan T, Huang Z, Zhang L, Zhang H, Zhu W, Zhu Y, Liu P. Tumor suppressor role of miR-3622b-5p in ERBB2-positive cancer. Oncotarget 2017; 8:23008-23019. [PMID: 28160563 PMCID: PMC5410281 DOI: 10.18632/oncotarget.14968] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/10/2017] [Indexed: 12/31/2022] Open
Abstract
Over-expression or amplification of ERBB2 is observed in multifarious carcinomas. However, the molecular mechanism of ERBB2 downregulation in ERBB2-positive cancers remains obscure. This experiment investigated the suppressive role of miR-3622b-5p in ERBB2-positive breast and gastric cancers. The luciferase activity of ERBB2 3′-untranslated region-based reporters constructed in HEK-293T, SK-BR-3 and MCF-10A cells suggested that ERBB2 was the target gene of miR-3622b-5p. Over-expressed miR-3622b-5p reduced the protein level of ERBB2, weakened the activation of mTORC1/S6, and induced the apoptosis of ERBB2-positive cancer cells. MiR-3622b-5p was significantly down-regulated in breast and gastric cancer tissues. This down-regulation in ERBB2-positive breast and gastric cancer tissues was more obvious than that in ERBB2-negative breast and gastric cancer tissues. MiR-3622b-5p turned ERBB2-positive cancer cells more vulnerable to the apoptosis induced by cisplatin and 5-fluorouracil. Taken together, miR-3622b-5p is involved in the proliferation and apoptosis of human ERBB2-positive cancer cells via targeting ERBB2/mTORC1 signaling pathway.
Collapse
Affiliation(s)
- Mingjie Lu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Tongshan Wang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Mingfeng He
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wenfang Cheng
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Ting Yan
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing 211166, PR China
| | - Zebo Huang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Lan Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Huo Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wei Zhu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 211166, PR China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, PR China
| | - Ping Liu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| |
Collapse
|
23
|
Li C, Li X, Gao S, Li C, Ma L. MicroRNA-133a Inhibits Proliferation of Gastric Cancer Cells by Downregulating ERBB2 Expression. Oncol Res 2017; 25:1169-1176. [PMID: 28109082 PMCID: PMC7840978 DOI: 10.3727/096504017x14847395834985] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Gastric cancer is the fourth most common type of cancer and the second highest leading cause of cancer-related deaths worldwide. It has already been established that miR-133a is involved in gastric cancer. In this study, we investigated the molecular mechanisms by which miR-133a inhibits the proliferation of gastric cancer cells. We analyzed the proliferative capacity of human gastric cancer cells SNU-1 using an MTT assay. Cell apoptosis was determined using flow cytometry. The expression levels of ERBB2, p-ERK1/2, and p-AKT in SNU-1 cells were determined using Western blot analysis. To confirm that ERBB2 is a direct target of miR-133a, a luciferase reporter assay was performed. Results showed that miR-133a overexpression inhibited SNU-1 cell proliferation and increased apoptosis. ERBB2 was a direct target of miR-133a, and it was negatively regulated by miR-133a. Interestingly, ERBB2 silencing has a similar impact to miR-133a overexpression, in that it significantly induced apoptosis and inhibited ERK and AKT activation. Our study showed that miR-133a inhibits the proliferation of gastric cancer cells by downregulating the expression of ERBB2 and its downstream signaling molecules p-ERK1/2 and p-AKT. Therefore, miR-133a might be used as a therapeutic target for treating gastric cancer.
Collapse
Affiliation(s)
- Chang Li
- *Department of Gastrointestinal Internal Medicine, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Xiaoping Li
- †Department of Pediatrics, The First Hospital of Jilin University, Changchun, P.R. China
| | - Shuohui Gao
- *Department of Gastrointestinal Internal Medicine, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Chang Li
- ‡Department of Cadre’s Ward, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| | - Lianjun Ma
- §Endoscopy Center, China–Japan Union Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
24
|
Hinske LC, Heyn J, Hübner M, Rink J, Hirschberger S, Kreth S. Intronic miRNA-641 controls its host Gene's pathway PI3K/AKT and this relationship is dysfunctional in glioblastoma multiforme. Biochem Biophys Res Commun 2017; 489:477-483. [PMID: 28576488 DOI: 10.1016/j.bbrc.2017.05.175] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/29/2017] [Indexed: 01/17/2023]
Abstract
MicroRNAs have established their role as important regulators of the epigenome. A considerable number of human miRNA genes are found in intronic regions of protein-coding host genes, in many cases adopting their regulatory circuitry. However, emerging evidence foreshadows an unprecedented importance for this relationship: Intronic miRNAs may protect the cell from overactivation of the respective host pathway, a setting that may trigger tumor development. AKT2 is a well-known proto-oncogene central to the PI3K/AKT pathway. This pathway is known to promote tumor growth and survival, especially in glioblastoma. Its intronic miRNA, hsa-miR-641, is scarcely investigated, however. We hypothesized that miR-641 regulates its host AKT2 and that this regulation may become dysfunctional in glioblastoma. We found that indeed miR-641 expression differs significantly between GBM tissue and normal brain samples, and that transfection of glioma cells with miR-641 antagonizes the PI3K/AKT pathway. Combining clinical samples, cell cultures, and biomolecular methods, we could show that miR-641 doesn't affect AKT2's expression levels, but down-regulates kinases that are necessary for AKT2-activation, thereby affecting its functional state. We also identified NFAT5 as a miR-641 regulated central factor to trigger the expression of these kinases and subsequently activate AKT2. In summary, our study is the first that draws a connecting line between the proto-oncogene AKT2 and its intronic miRNA miR-641 with implication for glioblastoma development.
Collapse
Affiliation(s)
- Ludwig Christian Hinske
- Department of Anesthesiology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany.
| | - Jens Heyn
- Department of Anesthesiology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany.
| | - Max Hübner
- Department of Anesthesiology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany; Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany.
| | - Jessica Rink
- Department of Anesthesiology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany; Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany.
| | - Simon Hirschberger
- Department of Anesthesiology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany; Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany.
| | - Simone Kreth
- Department of Anesthesiology, University Hospital of the Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany; Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-University Munich, Marchioninistraße 15, D-81377 Munich, Germany.
| |
Collapse
|
25
|
Newie I, Søkilde R, Persson H, Jacomasso T, Gorbatenko A, Borg Å, de Hoon M, Pedersen SF, Rovira C. HER2-encoded mir-4728 forms a receptor-independent circuit with miR-21-5p through the non-canonical poly(A) polymerase PAPD5. Sci Rep 2016; 6:35664. [PMID: 27752128 PMCID: PMC5067774 DOI: 10.1038/srep35664] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/04/2016] [Indexed: 12/24/2022] Open
Abstract
We previously reported that the human HER2 gene encodes the intronic microRNA mir-4728, which is overexpressed together with its oncogenic host gene and may act independently of the HER2 receptor. More recently, we also reported that the oncogenic miR-21-5p is regulated by 3' tailing and trimming by the non-canonical poly(A) polymerase PAPD5 and the ribonuclease PARN. Here we demonstrate a dual function for the HER2 locus in upregulation of miR-21-5p; while HER2 signalling activates transcription of mir-21, miR-4728-3p specifically stabilises miR-21-5p through inhibition of PAPD5. Our results establish a new and unexpected oncogenic role for the HER2 locus that is not currently being targeted by any anti-HER2 therapy.
Collapse
Affiliation(s)
- Inga Newie
- Department of Clinical Sciences, Lund, Division of Oncology and Pathology, Lund University Cancer Center, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden
| | - Rolf Søkilde
- Department of Clinical Sciences, Lund, Division of Oncology and Pathology, Lund University Cancer Center, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden
| | - Helena Persson
- Department of Clinical Sciences, Lund, Division of Oncology and Pathology, Lund University Cancer Center, Lund, Sweden
| | - Thiago Jacomasso
- Department of Clinical Sciences, Lund, Division of Oncology and Pathology, Lund University Cancer Center, Lund, Sweden
| | - Andrej Gorbatenko
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Åke Borg
- Department of Clinical Sciences, Lund, Division of Oncology and Pathology, Lund University Cancer Center, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden.,CREATE Health, Strategic Centre for Translational Cancer Research, Lund, Sweden
| | - Michiel de Hoon
- Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Stine F Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Carlos Rovira
- Department of Clinical Sciences, Lund, Division of Oncology and Pathology, Lund University Cancer Center, Lund, Sweden.,BioCARE, Strategic Cancer Research Program, Lund, Sweden.,CREATE Health, Strategic Centre for Translational Cancer Research, Lund, Sweden
| |
Collapse
|
26
|
Mutlu M, Saatci Ö, Ansari SA, Yurdusev E, Shehwana H, Konu Ö, Raza U, Şahin Ö. miR-564 acts as a dual inhibitor of PI3K and MAPK signaling networks and inhibits proliferation and invasion in breast cancer. Sci Rep 2016; 6:32541. [PMID: 27600857 PMCID: PMC5013276 DOI: 10.1038/srep32541] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/11/2016] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of PI3K and MAPK pathways promotes uncontrolled cell proliferation, apoptotic inhibition and metastasis. Individual targeting of these pathways using kinase inhibitors has largely been insufficient due to the existence of cross-talks between these parallel cascades. MicroRNAs are small non-coding RNAs targeting several genes simultaneously and controlling cancer-related processes. To identify miRNAs repressing both PI3K and MAPK pathways in breast cancer, we re-analyzed our previous miRNA mimic screen data with reverse phase protein array (RPPA) output, and identified miR-564 inhibiting both PI3K and MAPK pathways causing markedly decreased cell proliferation through G1 arrest. Moreover, ectopic expression of miR-564 blocks epithelial-mesenchymal transition (EMT) and reduces migration and invasion of aggressive breast cancer cells. Mechanistically, miR-564 directly targets a network of genes comprising AKT2, GNA12, GYS1 and SRF, thereby facilitating simultaneous repression of PI3K and MAPK pathways. Notably, combinatorial knockdown of these target genes using a cocktail of siRNAs mimics the phenotypes exerted upon miR-564 expression. Importantly, high miR-564 expression or low expression of target genes in combination is significantly correlated with better distant relapse-free survival of patients. Overall, miR-564 is a potential dual inhibitor of PI3K and MAPK pathways, and may be an attractive target and prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Merve Mutlu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Özge Saatci
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Suhail A Ansari
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Emre Yurdusev
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Huma Shehwana
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Özlen Konu
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Umar Raza
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| | - Özgür Şahin
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, 06800 Ankara, Turkey
| |
Collapse
|
27
|
Shi Z, Jiao S, Zhou Z. STRIPAK complexes in cell signaling and cancer. Oncogene 2016; 35:4549-57. [PMID: 26876214 DOI: 10.1038/onc.2016.9] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/24/2015] [Accepted: 12/24/2015] [Indexed: 12/28/2022]
Abstract
Striatin-interacting phosphatase and kinase (STRIPAK) complexes are striatin-centered multicomponent supramolecular structures containing both kinases and phosphatases. STRIPAK complexes are evolutionarily conserved and have critical roles in protein (de)phosphorylation. Recent studies indicate that STRIPAK complexes are emerging mediators and regulators of multiple vital signaling pathways including Hippo, MAPK (mitogen-activated protein kinase), nuclear receptor and cytoskeleton remodeling. Different types of STRIPAK complexes are extensively involved in a variety of fundamental biological processes ranging from cell growth, differentiation, proliferation and apoptosis to metabolism, immune regulation and tumorigenesis. Growing evidence correlates dysregulation of STRIPAK complexes with human diseases including cancer. In this review, we summarize the current understanding of the assembly and functions of STRIPAK complexes, with a special focus on cell signaling and cancer.
Collapse
Affiliation(s)
- Z Shi
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - S Jiao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Z Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|