1
|
Nishida M, Sato A, Shimizu A, Rahman N, Wada A, Kageyama S, Ogita H. EphA-Mediated Regulation of Stomatin Expression in Prostate Cancer Cells. Cancer Med 2024; 13:e70276. [PMID: 39377541 PMCID: PMC11459579 DOI: 10.1002/cam4.70276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND AND AIMS Tumor growth and progression are affected by interactions between tumor cells and stromal cells within the tumor microenvironment. We previously showed that the expression of an integral membrane protein, called stomatin, was increased in cancer cells following their association with stromal cells. Additionally, stomatin impaired the Akt signaling pathway to suppress tumor growth. However, it remains unclear how stomatin expression is regulated. To explore this, we examined the cell surface molecules that can transduce the intercellular communication signals between cancer cells and stromal cells. RESULTS Among these molecules, EphA3 and EphA7 receptors and their ligand ephrin-A5 were found to be expressed in prostate cancer cells, but not in prostate stromal cells. Cell-to-cell contact of prostate cancer cells through the EphA-ephrin-A interaction suppressed stomatin expression, while knockdown of EphA3/7 or ephrin-A5 increased stomatin expression. This increase contributed to an inhibition of prostate cancer cell proliferation. Intracellularly, the binding of ephrin-A to EphA attenuated extracellular signaling-regulated kinase (ERK) activation that promoted stomatin expression. Furthermore, ELK1 and ELK4, which are Ets family transcription factors phosphorylated by ERK, were involved in the induction of stomatin expression. We also found that higher Gleason score prostate cancer tissue samples had increased activation of EphA, while the stomatin expression and activated ERK and ELK levels were all low. In the mouse xenograft tumor samples generated by implantation of prostate cancer cells, EphA3 phosphorylation was attenuated and the ERK-ELK signaling and stomatin expression were enhanced in the area where stromal cells infiltrated the tumor. CONCLUSION The EphA-mediated signaling suppresses the ERK-ELK pathway, leading to the reduction of stomatin expression that affects prostate cancer malignancy.
Collapse
Affiliation(s)
- Masanari Nishida
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Nor Idayu A. Rahman
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| | - Akinori Wada
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Susumu Kageyama
- Department of UrologyShiga University of Medical ScienceOtsuJapan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular BiologyShiga University of Medical ScienceOtsuJapan
| |
Collapse
|
2
|
Alam SK, Wang L, Zhu Z, Hoeppner LH. IKKα promotes lung adenocarcinoma growth through ERK signaling activation via DARPP-32-mediated inhibition of PP1 activity. NPJ Precis Oncol 2023; 7:33. [PMID: 36966223 PMCID: PMC10039943 DOI: 10.1038/s41698-023-00370-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/08/2023] [Indexed: 03/27/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 80-85% cases of lung cancer cases. Diagnosis at advanced stages is common, after which therapy-refractory disease progression frequently occurs. Therefore, a better understanding of the molecular mechanisms that control NSCLC progression is necessary to develop new therapies. Overexpression of IκB kinase α (IKKα) in NSCLC correlates with poor patient survival. IKKα is an NF-κB-activating kinase that is important in cell survival and differentiation, but its regulation of oncogenic signaling is not well understood. We recently demonstrated that IKKα promotes NSCLC cell migration by physically interacting with dopamine- and cyclic AMP-regulated phosphoprotein, Mr 32000 (DARPP-32), and its truncated splice variant, t-DARPP. Here, we show that IKKα phosphorylates DARPP-32 at threonine 34, resulting in DARPP-32-mediated inhibition of protein phosphatase 1 (PP1), subsequent inhibition of PP1-mediated dephosphorylation of ERK, and activation of ERK signaling to promote lung oncogenesis. Correspondingly, IKKα ablation in human lung adenocarcinoma cells reduced their anchorage-independent growth in soft agar. Mice challenged with IKKα-ablated HCC827 cells exhibited less lung tumor growth than mice orthotopically administered control HCC827 cells. Our findings suggest that IKKα drives NSCLC growth through the activation of ERK signaling via DARPP-32-mediated inhibition of PP1 activity.
Collapse
Affiliation(s)
- Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
| | - Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zhu Zhu
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Yang Q, Zhu L, Ye M, Zhang B, Zhan P, Li H, Zou W, Liu J. Tumor Suppressor 4.1N/EPB41L1 is Epigenetic Silenced by Promoter Methylation and MiR-454-3p in NSCLC. Front Genet 2022; 13:805960. [PMID: 35795202 PMCID: PMC9251189 DOI: 10.3389/fgene.2022.805960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 04/08/2022] [Indexed: 12/24/2022] Open
Abstract
Non–small-cell lung cancer (NSCLC) is divided into three major histological types, namely, lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), and large-cell lung carcinoma (LCLC). We previously identified that 4.1N/EPB41L1 acts as a tumor suppressor and is reduced in NSCLC patients. In the current study, we explored the underlying epigenetic mechanisms of 4.1N/EPB41L1 reduction in NSCLC. The 4.1N/EPB41L1 gene promoter region was highly methylated in LUAD and LUSC patients. LUAD patients with higher methylation level in the 4.1N/EPB41L1 gene promoter (TSS1500, cg13399773 or TSS200, cg20993403) had a shorter overall survival time (Log-rank p = 0.02 HR = 1.509 or Log-rank p = 0.016 HR = 1.509), whereas LUSC patients with higher methylation level in the 4.1N/EPB41L1 gene promoter (TSS1500 cg13399773, TSS1500 cg07030373 or TSS200 cg20993403) had a longer overall survival time (Log-rank p = 0.045 HR = 0.5709, Log-rank p = 0.018 HR = 0.68 or Log-rank p = 0.014 HR = 0.639, respectively). High methylation of the 4.1N/EPB41L1 gene promoter appeared to be a relatively early event in LUAD and LUSC. DNA methyltransferase inhibitor 5-Aza-2′-deoxycytidine restored the 4.1N/EPB41L1 expression at both the mRNA and protein levels. MiR-454-3p was abnormally highly expressed in NSCLC and directly targeted 4.1N/EPB41L1 mRNA. MiR-454-3p expression was significantly correlated with 4.1N/EPB41L1 expression in NSCLC patients (r = −0.63, p < 0.0001). Therefore, we concluded that promoter hypermethylation of the 4.1N/EPB41L1 gene and abnormally high expressed miR-454-3p work at different regulation levels but in concert to restrict 4.1N/EPB41L1 expression in NSCLC. Taken together, this work contributes to elucidate the underlying epigenetic disruptions of 4.1N/EPB41L1 deficiency in NSCLC.
Collapse
Affiliation(s)
- Qin Yang
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- School of Medical Laboratory, Shao Yang University, Shaoyang, China
| | - Lin Zhu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan Univers ity, Changsha, China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Peihe Zhan
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Hui Li
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Molecular Science and Biomedicine Laboratory, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, China
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan Univers ity, Changsha, China
- *Correspondence: Jing Liu, ; Wen Zou, ; Hui Li,
| | - Wen Zou
- Department of Oncology, The Second Xiangya Hospital of Central South University, Central South University, Changsha, China
- *Correspondence: Jing Liu, ; Wen Zou, ; Hui Li,
| | - Jing Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- *Correspondence: Jing Liu, ; Wen Zou, ; Hui Li,
| |
Collapse
|
4
|
Liu X, Yang J, Yang C, Huang X, Han M, Kang F, Li J. Morphine promotes the malignant biological behavior of non-small cell lung cancer cells through the MOR/Src/mTOR pathway. Cancer Cell Int 2021; 21:622. [PMID: 34823532 PMCID: PMC8613927 DOI: 10.1186/s12935-021-02334-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Morphine, a µ-opioid receptor (MOR) agonist, has been shown to be related to the activity of cancer cells, and a higher morphine dosage reduces the survival time of patients with lung cancer. However, the effect of morphine on the malignant behavior of lung cancer cells remains unclear. The aim of this study was to investigate the specific molecular mechanism by which morphine regulates the malignant biological behavior of non-small cell lung cancer. METHODS Immunofluorescence staining and Western blot analyses were performed to detect MOR expression. H460 non-small cell lung cancer cells were used in this study, and cell proliferation, the cell cycle and apoptosis were evaluated using Cell Counting Kit-8 (CCK-8) and flow cytometry assays, respectively. Cell migration and invasion were detected using wound healing and Transwell assays. The effect of morphine on lung cancer development in vivo was examined by performing a xenograft tumor assay following morphine treatment. RESULTS Morphine promoted the growth of H460 cells both in vivo and in vitro. Morphine enhanced cell migration and invasion, modified cell cycle progression through the S/G2 transition and exerted an antiapoptotic effect on H460 cells. Additionally, morphine increased Rous sarcoma oncogene cellular homolog (Src) phosphorylation and activated the phosphoinositide 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway. Treatment with the MOR antagonist methylnaltrexone (MNTX) and the Src inhibitor protein phosphatase 1 (PP1) reduced the phosphorylation induced by morphine. Furthermore, MNTX, PP1, and the PI3K/AKT inhibitor deguelin reversed the antiapoptotic effect of morphine on lung cancer cells. CONCLUSION Morphine promotes the malignant biological behavior of H460 cells by activating the MOR and Src/mTOR signaling pathways.
Collapse
Affiliation(s)
- Xingyun Liu
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, 230036, China
| | - Jia Yang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Chengwei Yang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Xiang Huang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Mingming Han
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Fang Kang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Juan Li
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, 230036, China.
| |
Collapse
|
5
|
Yang Q, Liu J, Wang Z. 4.1N-Mediated Interactions and Functions in Nerve System and Cancer. Front Mol Biosci 2021; 8:711302. [PMID: 34589518 PMCID: PMC8473747 DOI: 10.3389/fmolb.2021.711302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/16/2021] [Indexed: 01/05/2023] Open
Abstract
Scaffolding protein 4.1N is a neuron-enriched 4.1 homologue. 4.1N contains three conserved domains, including the N-terminal 4.1-ezrin-radixin-moesin (FERM) domain, internal spectrin–actin–binding (SAB) domain, and C-terminal domain (CTD). Interspersed between the three domains are nonconserved domains, including U1, U2, and U3. The role of 4.1N was first reported in the nerve system. Then, extensive studies reported the role of 4.1N in cancers and other diseases. 4.1N performs numerous vital functions in signaling transduction by interacting, locating, supporting, and coordinating different partners and is involved in the molecular pathogenesis of various diseases. In this review, recent studies on the interactions between 4.1N and its contactors (including the α7AChr, IP3R1, GluR1/4, GluK1/2/3, mGluR8, KCC2, D2/3Rs, CASK, NuMA, PIKE, IP6K2, CAM 1/3, βII spectrin, flotillin-1, pp1, and 14-3-3) and the 4.1N-related biological functions in the nerve system and cancers are specifically and comprehensively discussed. This review provides critical detailed mechanistic insights into the role of 4.1N in disease relationships.
Collapse
Affiliation(s)
- Qin Yang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,School of Medical Laboratory, Shao Yang University, Shaoyang, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Zi Wang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
6
|
Ning S, Hua L, Ji Z, Fan D, Meng X, Li Z, Wang Q, Guo Z. Protein 4.1 family and ion channel proteins interact to regulate the process of heart failure in rats. Acta Histochem 2021; 123:151748. [PMID: 34271280 DOI: 10.1016/j.acthis.2021.151748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 02/06/2023]
Abstract
Heart failure (HF) is a major cause of death in cardiovascular diseases worldwide, and its molecular mechanisms and effective prevention strategies remain to be further studied. The myocardial cytoskeleton plays a pivotal role in many heart diseases. However, little is known about the function of the membrane cytoskeleton 4.1 protein family and related regulatory mechanisms in the pathogenesis of HF. In this study, we detected the localization and expression of the protein 4.1 family and ion channel proteins in a rat HF model induced by doxorubicin (DOX), and studied the interactions between them. Our results showed that compared with the control group, the HF group displayed an increased expression level of protein 4.1R and decreased levels of protein 4.1 G and 4.1 N. The Nav1.5 protein levels were significantly increased, while the SERCA2a and Cav1.2 protein levels were significantly decreased in the HF group. Furthermore, there is co-localization and interaction between protein 4.1R and Nav1.5, protein 4.1 G and SERCA2a, protein 4.1 N and Cav1.2, respectively. Taken together, the results indicated that the protein 4.1 family might be involved in the occurrence and development of HF through its interaction with ion channel proteins, suggesting that 4.1 proteins may serve as a novel therapeutic target for HF.
Collapse
Affiliation(s)
- Shuwei Ning
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Lei Hua
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhenyu Ji
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Dandan Fan
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangguang Meng
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhiying Li
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Qian Wang
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China
| | - Zhikun Guo
- Zhengzhou Key Laboratory, Zhengzhou No. 7 People's Hospital, Zhengzhou, 450016, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
7
|
Matos B, Howl J, Jerónimo C, Fardilha M. Modulation of serine/threonine-protein phosphatase 1 (PP1) complexes: A promising approach in cancer treatment. Drug Discov Today 2021; 26:2680-2698. [PMID: 34390863 DOI: 10.1016/j.drudis.2021.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/23/2021] [Accepted: 08/05/2021] [Indexed: 01/21/2023]
Abstract
Cancer is the second leading cause of death worldwide. Despite the availability of numerous therapeutic options, tumor heterogeneity and chemoresistance have limited the success of these treatments, and the development of effective anticancer therapies remains a major focus in oncology research. The serine/threonine-protein phosphatase 1 (PP1) and its complexes have been recognized as potential drug targets. Research on the modulation of PP1 complexes is currently at an early stage, but has immense potential. Chemically diverse compounds have been developed to disrupt or stabilize different PP1 complexes in various cancer types, with the objective of inhibiting disease progression. Beneficial results obtained in vitro now require further pre-clinical and clinical validation. In conclusion, the modulation of PP1 complexes seems to be a promising, albeit challenging, therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Bárbara Matos
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal; Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - John Howl
- Molecular Pharmacology Group, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4050-513 Porto, Portugal
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
8
|
Zhu LM, Zeng D, Lei XC, Huang J, Deng YF, Ji YB, Liu J, Dai FF, Li YZ, Shi DD, Zhu YQ, Dai AG, Wang Z. KLF2 regulates neutrophil migration by modulating CXCR1 and CXCR2 in asthma. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165920. [PMID: 32800946 DOI: 10.1016/j.bbadis.2020.165920] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 07/01/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
Neutrophils are key inflammatory cells in the immunopathogenesis of asthma. Neutrophil migration can be initiated through activation of the CXCR1 and CXCR2 receptors by CXC chemokines, such as IL-8. Although transcription factor KLF2 has been found to maintain T cell migration patterns through repression of several chemokine receptors, whether KLF2 can regulate neutrophil migration via modulation of CXCR1 and CXCR2 is unknown. Here, we aimed to explore the functions of KLF2, CXCR1 and CXCR2 in neutrophil migration in asthma and to establish a regulatory role of KLF2 for CXCR1/2. We demonstrate that with asthma aggravation, the percentages and migration rates of peripheral blood neutrophils gradually increased in asthmatic patients and the guinea pig asthma model. Correspondingly, both the KLF2 mRNA and protein levels in neutrophils were gradually reduced. While CXCR1 and CXCR2 expression was negatively correlated with KLF2. In vitro knockdown of KLF2 dramatically increased the migration of HL-60-drived neutrophil-like cells, which was accompanied by an increase in the CXCR1 and CXCR2 mRNA and protein expression levels. Taken together, our results indicate that decreased KLF2 aggravates asthma progression by promoting neutrophil migration, which is associated with the transcriptional upregulation of CXCR1 and CXCR2. The KLF2 and/or CXCR1/2 expression levels may represent an indicator of asthma severity.
Collapse
Affiliation(s)
- Li-Ming Zhu
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China; Institute of Respiratory Disease, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China.
| | - Dan Zeng
- Institute of Respiratory Disease, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Xue-Chun Lei
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Jin Huang
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Yan-Feng Deng
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Yu-Bin Ji
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Jing Liu
- Molecular Biology Research Center, School of life Sciences, Central South University, Changsha 410008, China
| | - Fang-Fang Dai
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Yu-Zhu Li
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Dan-Dan Shi
- Department of Geriatric Respiratory Medicine, Hunan Provincial People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha 410016, China
| | - Ying-Qun Zhu
- Department of Respiratory Medicine, The Third Hospital of Changsha, Changsha 410015, China
| | - Ai-Guo Dai
- Institute of Respiratory Disease, Changsha medical University, Changsha 410219, China
| | - Zi Wang
- Molecular Biology Research Center, School of life Sciences, Central South University, Changsha 410008, China; Key Laboratory of Nanobiological Technology of Chinese Minisitry of Health, Xiangya Hospital, Central South Universeity, Changsha 410008, China.
| |
Collapse
|
9
|
Loss of 4.1N in epithelial ovarian cancer results in EMT and matrix-detached cell death resistance. Protein Cell 2020; 12:107-127. [PMID: 32448967 PMCID: PMC7862473 DOI: 10.1007/s13238-020-00723-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/23/2020] [Indexed: 01/01/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the leading causes of death from gynecologic cancers and peritoneal dissemination is the major cause of death in patients with EOC. Although the loss of 4.1N is associated with increased risk of malignancy, its association with EOC remains unclear. To explore the underlying mechanism of the loss of 4.1N in constitutive activation of epithelial-mesenchymal transition (EMT) and matrix-detached cell death resistance, we investigated samples from 268 formalin-fixed EOC tissues and performed various in vitro and in vivo assays. We report that the loss of 4.1N correlated with progress in clinical stage, as well as poor survival in EOC patients. The loss of 4.1N induces EMT in adherent EOC cells and its expression inhibits anoikis resistance and EMT by directly binding and accelerating the degradation of 14-3-3 in suspension EOC cells. Furthermore, the loss of 4.1N could increase the rate of entosis, which aggravates cell death resistance in suspension EOC cells. Moreover, xenograft tumors in nude mice also show that the loss of 4.1N can aggravate peritoneal dissemination of EOC cells. Single-agent and combination therapy with a ROCK inhibitor and a 14-3-3 antagonist can reduce tumor spread to varying degrees. Our results not only define the vital role of 4.1N loss in inducing EMT, anoikis resistance, and entosis-induced cell death resistance in EOC, but also suggest that individual or combined application of 4.1N, 14-3-3 antagonists, and entosis inhibitors may be a promising therapeutic approach for the treatment of EOC.
Collapse
|
10
|
Tang Z, Xiong D, Song J, Ye M, Liu J, Wang Z, Zhang L, Xiao X. Antitumor Drug Combretastatin-A4 Phosphate Aggravates the Symptoms of Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Front Pharmacol 2020; 11:339. [PMID: 32265711 PMCID: PMC7106770 DOI: 10.3389/fphar.2020.00339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 03/06/2020] [Indexed: 12/17/2022] Open
Abstract
Ulcerative colitis (UC) is an idiopathic inflammatory bowel disease (IBD) that causes long-lasting inflammation and ulcers in the innermost lining of the colon and rectum. Previous studies demonstrated that resveratrol suppresses colitis and colon cancer associated with colitis by improving glucose metabolism, but resveratrol use is limited by its low oral bioavailability. Combretastatin-A4 phosphate (CA4P) is a vascular-disrupting agent with antitumor activity. CA4P is structurally similar to resveratrol, but whether CA4P has the same effect as resveratrol on UC is not clear. In this study, we examined the pharmacological effects of CA4P administration on dextran sulfate sodium (DSS)-induced inflammation in a mouse model of UC. C57BL/6 mice were administered 2.5% DSS in the drinking water to induce acute UC. CA4P (11 mg/kg/d) was injected intraperitoneally daily. The Disease Activity Index (DAI) score and histological score were evaluated to determine the severity of UC. Colon tissues and blood samples were collected for histological analyses. The results show that CA4P plus DSS significantly decreased colon length (P < 0.05 versus DSS+PBS group) and body weight (P < 0.001 versus PBS group), while increased spleen weight (P < 0.01 versus DSS+PBS group), DAI score (P < 0.01 versus DSS+PBS group), and histological score (P < 0.01 versus DSS+PBS group). Moreover, CA4P exacerbated the pathological features of colitis and significantly increased proinflammatory cytokines (IL-1β, IL-6, TNF-α) and inflammatory cells (neutrophil, lymphocyte, monocyte). These findings reveal that CA4P aggravates the symptoms of DSS-induced UC and provide a key reference for the potential of CA4P as an anticancer drug.
Collapse
Affiliation(s)
- Zhengshan Tang
- Hepatobiliary and Enteric Surgery Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Dehui Xiong
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Jianhui Song
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering for Theranostics, Hunan University, Changsha, China
| | - Jing Liu
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Zi Wang
- Hepatobiliary and Enteric Surgery Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Zhang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Xiaojuan Xiao
- School of Life Sciences & Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| |
Collapse
|
11
|
Han X, Wang X, Li H, Zhang H. Mechanism of microRNA-431-5p- EPB41L1 interaction in glioblastoma multiforme cells. Arch Med Sci 2019; 15:1555-1564. [PMID: 31749885 PMCID: PMC6855151 DOI: 10.5114/aoms.2019.88274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/13/2017] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is a kind of malignant brain tumor prevalent in adults, with the characteristics well adapted to poorly immunogenic and hypoxic conditions. Effective treatment of GBM is impeded due to the high proliferation, migration and invasion of GBM cells. GBM cells migrate by degrading the extracellular matrix, so it is difficult to have GBM cells eradicated completely by surgery. This study aims to confirm that miR-431-5p could influence the proliferation, invasion and migration of human glioblastoma multiforme cells by targeting EPB41L1 (erythrocyte membrane protein band 4.1). MATERIAL AND METHODS The expression levels of miR-431-5p and EPB41L1 were detected in GBM cells and tissues using qRT-PCR. Dual luciferase reporter gene assay and western blot were applied to confirm the targeting relationship between miR-431-5p and EPB41L1. GBM cell line U87 was used in MTT, flow cytometry, Transwell, and wound healing assays to determine cell proliferation, migration and invasion. RESULTS MiR-431-5p was overexpressed in GBM tissues while EPB41L1 was under-expressed. The results of dual luciferase reporter gene assay and western blot demonstrated that miR-431-5p could target EPB41L1 and suppress its expression. Down-regulating the expression of miR-431-5p or up-regulating the expression of EPB41L1 could inhibit the proliferation, invasion and migration but promote the apoptosis of GBM cells. CONCLUSIONS MiR-431-5p facilitated the progression of GBM by inhibiting EPB41L1 expression.
Collapse
Affiliation(s)
- Xiaoyong Han
- Third Department of Neurosurgery, CangZhou Central Hospital, CangZhou, Hebei, China
| | - Xirui Wang
- Third Department of Neurosurgery, CangZhou Central Hospital, CangZhou, Hebei, China
| | - Hui Li
- Department of Surgery, Dongguang County Chinese Traditional Medicine Hospital, CangZhou, Hebei, China
| | - Hui Zhang
- Third Department of Neurosurgery, CangZhou Central Hospital, CangZhou, Hebei, China
| |
Collapse
|
12
|
Li H, Liu J, Cao W, Xiao X, Liang L, Liu-Smith F, Wang W, Liu H, Zhou P, Ouyang R, Yuan Z, Liu J, Ye M, Zhang B. C-myc/miR-150/EPG5 axis mediated dysfunction of autophagy promotes development of non-small cell lung cancer. Am J Cancer Res 2019; 9:5134-5148. [PMID: 31410206 PMCID: PMC6691579 DOI: 10.7150/thno.34887] [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: 03/14/2019] [Accepted: 06/16/2019] [Indexed: 12/24/2022] Open
Abstract
Rationale: Lung cancer is the leading cause of cancer death worldwide, and treatment options are limited to mainly cytotoxic agents. Here we reveal a novel role of miR-150 in non-small cell lung cancer (NSCLC) development and seek potential new therapeutic targets. Methods: The miR-150-mediated autophagy dysfunction in NSCLC cells were examined using molecular methods in vitro and in vivo. The upstream regulatory element and downstream target of miR-150 were identified in vitro and validated in vivo. Potential therapeutic methods (anti-c-myc or anti-miR-150) were tested in vitro and in vivo. Clinical relevance of the c-myc/miR-150/EPG5 axis in NSCLC was validated in human clinical samples and large genomics database. Results: miR-150 blocked the fusion of autophagosomes and lysosomes through directly repressing EPG5. The miR-150-mediated autophagy defect induced ER stress and increased cellular ROS levels and DNA damage response, and promoted NSCLC cell proliferation and tumor growth. Knockdown of EPG5 promoted NSCLC cell proliferation, and attenuated the effects of miR-150. c-myc gene was identified as a miR-150 transcriptional factor which increased miR-150 accumulation, therefore pharmacologically or genetically inhibiting c-myc/miR-150 expression significantly inhibited NSCLC cell growth in vitro and in vivo. Both c-myc and miR-150 were significantly over-expressed in NSCLC, while EPG5 was down-regulated in NSCLC. Expression levels of these molecules were well correlated, and also well correlated with patient survival. Conclusions: Our findings suggest that c-myc/miR-150/EPG5 mediated dysfunction of autophagy contributes to NSCLC development, which may provide a potential new diagnostic and therapeutic target in NSCLC.
Collapse
|
13
|
Feng G, Guo K, Yan Q, Ye Y, Shen M, Ruan S, Qiu S. Expression of Protein 4.1 Family in Breast Cancer: Database Mining for 4.1 Family Members in Malignancies. Med Sci Monit 2019; 25:3374-3389. [PMID: 31063460 PMCID: PMC6524556 DOI: 10.12659/msm.914085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/25/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The protein 4.1 family is a family of cytoskeletal proteins that play an important role in maintaining normal cell morphology and cell adhesion, migration, division, and intercellular signaling. The main aim of this study was to explore the prognostic significance of the protein 4.1 family in breast cancer (BC) patients and to provide new biomarkers and therapeutic targets for the diagnosis and treatment of BC. MATERIAL AND METHODS The expression of 4.1 family members in various tumor types was compared to normal controls using the ONCOMINE and GOBO databases. The prognostic significance of the 4.1 family in BC patients was determined by Kaplan-Meier Plotter. RESULTS EPB41L2 (4.1G) was expressed at higher levels in normal tissues compared with BC patients for all 4.1 family members. In survival analysis, 4.1G and EPB41 (4.1R) mRNA high expressions were associated with better survival in BC patients. Moreover, 4.1G high expression was significantly associated with longer overall survival (OS) in luminal A and protracted relapse-free survival (RFS) in luminal B subtype BC patients who received Tamoxifen treatment. In addition, high expression of each 4.1 family member also showed better prognostic value in different molecular subtypes of BC. CONCLUSIONS These results indicate that the protein 4.1 family can be regarded as novel biomarkers and potential therapeutic targets for BC. Further research is needed to explore the detailed biological functions.
Collapse
Affiliation(s)
- Guan Feng
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Kaibo Guo
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Qingying Yan
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Ye Ye
- Hangzhou Vocational and Technical College, Hangzhou, Zhejiang, P.R. China
| | - Minhe Shen
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Shanming Ruan
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Shengliang Qiu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
14
|
Zhang Y, Gao H, Zhou W, Sun S, Zeng Y, Zhang H, Liang L, Xiao X, Song J, Ye M, Yang Y, Zhao J, Wang Z, Liu J. Targeting c-met receptor tyrosine kinase by the DNA aptamer SL1 as a potential novel therapeutic option for myeloma. J Cell Mol Med 2018; 22:5978-5990. [PMID: 30353654 PMCID: PMC6237600 DOI: 10.1111/jcmm.13870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 08/03/2018] [Indexed: 12/13/2022] Open
Abstract
Hepatocyte growth factor (HGF)/c‐met pathway activation has been implicated in the pathogenesis of multiple myeloma (MM), and blocking this pathway has been considered a rational therapeutic strategy for treating MM. Aptamers are single‐stranded nucleic acid molecules that fold into complex 3D structures and bind to a variety of targets. Recently, it was reported that DNA aptamer SL1 exhibited high specificity and affinity for c‐met and inhibited HGF/c‐met signaling in SNU‐5 cells. However, as the first c‐met‐targeted DNA aptamer to be identified, application of SL1 to myeloma treatment requires further investigation. Here, we explore the potential application of SL1 in MM. Our results indicated that c‐met expression is gradually increased in MM patients and contributes to poor outcomes. SL1 selectively bound to c‐met‐positive MM cells but not to normal B cells and suppressed the growth, migration and adhesion of MM cells in vitro in a co‐culture model performed with HS5 cells, wherein SL1 inhibited HGF‐induced activation of c‐met signaling. In vivo and ex vivo fluorescence imaging showed that SL1 accumulated in the c‐met positive tumour areas. In addition, SL1 was active against CD138+ primary MM cells and displayed a synergistic inhibition effect with bortezomib. Collectively, our data suggested that SL1 could be beneficial as a c‐met targeted antagonist in MM.
Collapse
Affiliation(s)
- Yibin Zhang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China.,Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China.,Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Hongmei Gao
- Nursing Department, Xiangya Hospital, Central South University, Changsha, China
| | - Weihua Zhou
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Sunming Sun
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Yayue Zeng
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Hui Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Long Liang
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jianhui Song
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Hunan University, Changsha, China
| | - Yujia Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Jingfeng Zhao
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
| | - Zi Wang
- Key Laboratory of Nanobiological Technology of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China.,Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| | - Jing Liu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
15
|
Protein 4.1N is required for the formation of the lateral membrane domain in human bronchial epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1143-1151. [PMID: 29428502 DOI: 10.1016/j.bbamem.2018.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/16/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022]
Abstract
The membrane skeleton forms a scaffold on the cytoplasmic side of the plasma membrane. The erythrocyte membrane represents an archetype of such structural organization. It has been documented that a similar membrane skeleton also exits in the Golgi complex. It has been previously shown that βII spectrin and ankyrin G are localized at the lateral membrane of human bronchial epithelial cells. Here we show that protein 4.1N is also located at the lateral membrane where it associates E-cadherin, β-catenin and βII spectrin. Importantly, depletion of 4.1N by RNAi in human bronchial epithelial cells resulted in decreased height of lateral membrane, which was reversed following re-expression of mouse 4.1N. Furthermore, although the initial phase of lateral membrane biogenesis proceeded normally in 4.1N-depleted cells, the final height of the lateral membrane of 4.1N-depleted cells was shorter compared to that of control cells. Our findings together with previous findings imply that 4.1N, βII spectrin and ankyrin G are structural components of the lateral membrane skeleton and that this skeleton plays an essential role in the assembly of a fully functional lateral membrane.
Collapse
|
16
|
Chang Y, Yan W, Sun C, Liu Q, Wang J, Wang M. miR-145-5p inhibits epithelial-mesenchymal transition via the JNK signaling pathway by targeting MAP3K1 in non-small cell lung cancer cells. Oncol Lett 2017; 14:6923-6928. [PMID: 29344125 DOI: 10.3892/ol.2017.7092] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 07/03/2017] [Indexed: 01/13/2023] Open
Abstract
Lung cancer is one of the most common types of tumors and the leading cause of cancer-associated mortality in the world. Additionally, non-small cell lung cancer (NSCLC) accounts for ~80% of all lung cancer cases. Epithelial-mesenchymal transition (EMT) is an important cell biological process, which is associated with cancer migration, metastasis, asthma and fibrosis in the lung. In the present study, it was revealed that miR-145-5p was able to suppress EMT by inactivating the c-Jun N-terminal kinase (JNK) signaling pathway in NSCLC cells. Mitogen-activated protein kinase kinase kinase 1 (MAP3K1) was predicted and confirmed to be a novel target of miR-145-5p. Overexpression of MAP3K1 was able to reverse the inhibition of EMT induced by miR-145-5p via the JNK signaling pathway. Overall, the results revealed that miR-145-5p inhibits EMT via the JNK signaling pathway by targeting MAP3K1 in NSCLC cells.
Collapse
Affiliation(s)
- Yongmei Chang
- Department of Respiratory Medicine, Guangdong No. 2 Provincial People Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Wensen Yan
- Department of Respiratory Medicine, Guangdong No. 2 Provincial People Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Cong Sun
- Department of Respiratory Medicine, Guangdong No. 2 Provincial People Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Qingfeng Liu
- Department of Respiratory Medicine, Guangdong No. 2 Provincial People Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Jun Wang
- Department of Respiratory Medicine, Guangdong No. 2 Provincial People Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Mingzhi Wang
- Department of Cardiothoracic Surgery, Guangdong No. 2 Provincial People Hospital, Guangzhou, Guangdong 510317, P.R. China
| |
Collapse
|
17
|
Li H, Ouyang R, Wang Z, Zhou W, Chen H, Jiang Y, Zhang Y, Li H, Liao M, Wang W, Ye M, Ding Z, Feng X, Liu J, Zhang B. MiR-150 promotes cellular metastasis in non-small cell lung cancer by targeting FOXO4. Sci Rep 2016; 6:39001. [PMID: 27976702 PMCID: PMC5157020 DOI: 10.1038/srep39001] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/15/2016] [Indexed: 12/22/2022] Open
Abstract
Previous studies have shown that dysregulation of microRNA-150 (miR-150) is associated with aberrant proliferation of human non-small cell lung cancer (NSCLC) cells. However, whether miR-150 has a critical role in NSCLC cell metastasis is unknown. Here, we reveal that the critical pro-metastatic role of miR-150 in the regulation of epithelial-mesenchymal-transition (EMT) through down-regulation of FOXO4 in NSCLC. In vitro, miR-150 targets 3'UTR region of FOXO4 mRNA, thereby negatively regulating its expression. Clinically, the expression of miR-150 was frequently up-regulated in metastatic NSCLC cell lines and clinical specimens. Contrarily, FOXO4 was frequently down-regulated in NSCLC cell lines and clinical specimens. Functional studies show that ectopic expression of miR-150 enhanced tumor cell metastasis in vitro and in a mouse xenograft model, and triggered EMT-like changes in NSCLC cells (including E-cadherin repression, N-cadherin and Vimentin induction, and mesenchymal morphology). Correspondingly, FOXO4 knockdown exhibited pro-metastatic and molecular effects resembling the effect of miR-150 over-expression. Moreover, NF-κB/snail/YY1/RKIP circuitry regulated by FOXO4 were likely involved in miR-150-induced EMT event. Simultaneous knockdown of miR-150 and FOXO4 abolished the phenotypic and molecular effects caused by individual knockdown of miR-150. Therefore, our study provides previously unidentified pro-metastatic roles and mechanisms of miR-150 in NSCLC.
Collapse
Affiliation(s)
- Hui Li
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Ruoyun Ouyang
- Department of Respiratory Medicine, Respiratory Disease Research Institute, Second XiangYa Hospital of Central South University, Changsha, 410011, China
| | - Zi Wang
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Weihua Zhou
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Huiyong Chen
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Yawen Jiang
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Yibin Zhang
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Hui Li
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Mengting Liao
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Weiwei Wang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha, 410082, China
| | - Zhigang Ding
- Center for Experimental Medicine, the Third Xiangya Hospital, Central South University, Changsha, 400083, China
| | - Xueping Feng
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Jing Liu
- The State Key Laboratory of Medical Genetics &School of Life Sciences, Central South University, Changsha, 410078, China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| |
Collapse
|
18
|
Wang H, Wu S, Huang S, Yin S, Zou G, Huang K, Zhang Z, Tang A, Wen W. Follistatin-like protein 1 contributes to dendritic cell and T-lymphocyte activation in nasopharyngeal carcinoma patients by altering nuclear factor κb and Jun N-terminal kinase expression. Cell Biochem Funct 2016; 34:554-562. [PMID: 27859422 PMCID: PMC5215428 DOI: 10.1002/cbf.3227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/01/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023]
Abstract
Follistatin‐like protein 1 (FSTL1) is a newly characterized protein that can regulate the immune response in various ways. Dendritic cells (DCs) are central to immune regulation. In this study, we explored the impact of FSTL1 on DC activity in nasopharyngeal carcinoma (NPC) patients. The surface expression of CD40, CD86, and HLA‐DR on DCs was analyzed and showed significantly elevated expression levels, indicating DC maturity. After FSTL1 was added to DCs collected from NPC patients (n = 50), controls (n = 47), and healthy donors (n = 10), interferon γ secretion and T‐cell receptor expression in cytotoxic T lymphocytes were also investigated. In the experimental groups, the expression of the critical immune protein nuclear factor (NF)‐κb was upregulated, whereas Jun N‐terminal kinase (JNK) was downregulated. Our findings demonstrate that FSTL1 plays a critical role in immune regulation, enhancing the antigen presentation ability of DCs by up‐regulating NF‐κb expression and down‐regulating JNK expression.
Collapse
Affiliation(s)
- Hong Wang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Senyong Wu
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital, Guigang, Guangxi, China
| | - Shiping Huang
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital, Guigang, Guangxi, China
| | - Shaolin Yin
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital, Guigang, Guangxi, China.,Department of Otolaryngology, The Cooperation of Chinese and Western Medicine Hospital in Guangzhou, Guangzhou, China
| | - Guilong Zou
- Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital, Guigang, Guangxi, China.,Department of Otolaryngology, The People's Hospital of Hezhou, Guangxi, China
| | - Kuan'en Huang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,Department of Otorhinolaryngology Head and Neck Surgery, The People's Hospital, Guigang, Guangxi, China
| | - Zhe Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.,Department of Otolaryngology, The Cooperation of Chinese and Western Medicine Hospital in Guangzhou, Guangzhou, China
| | - Anzhou Tang
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wensheng Wen
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
19
|
Wang FF, Wang S, Xue WH, Cheng JL. microRNA-590 suppresses the tumorigenesis and invasiveness of non-small cell lung cancer cells by targeting ADAM9. Mol Cell Biochem 2016; 423:29-37. [PMID: 27770372 DOI: 10.1007/s11010-016-2822-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 09/08/2016] [Indexed: 12/22/2022]
Abstract
microRNAs (miRNAs), a family of small non-coding RNA molecules, are implicated in cancer growth and progression. In the present study, we examined the expression and biological roles of miR-590 in non-small cell lung cancer (NSCLC). Compared to normal lung tissues, miR-590 expression was downregulated in primary NSCLCs and, to a greater extent, in corresponding brain metastases. NSCLC cell lines with high metastatic potential had significantly (P < 0.05) lower levels of miR-590 than those with low metastatic potential. Re-expression of miR-590 suppressed NSCLC cell proliferation, colony formation, migration, and invasion in vitro and tumorigenesis in vivo. In contrast, inhibition of miR-590 enhanced the migration and invasion of NSCLC cells. Mechanistic studies revealed that a disintegrin and metalloproteinase 9 (ADAM9) was a direct target of miR-590. Delivery of miR-590 mimic was found to decrease endogenous ADAM9 expression in NSCLC cells. Enforced expression of a miRNA-resistant form of ADAM9 significantly restored the aggressive behaviors in miR-590-overexpressing NSCLC cells. Taken together, our data reveal miR-590 as a tumor suppressor in NSCLC, which is at least partially mediated through targeting of ADAM9. Restoration of miR-590 may provide a promising therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Fei-Fei Wang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Song Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wen-Hua Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jing-Liang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
20
|
The Fra-1-miR-134-SDS22 feedback loop amplifies ERK/JNK signaling and reduces chemosensitivity in ovarian cancer cells. Cell Death Dis 2016; 7:e2384. [PMID: 27685628 PMCID: PMC5059884 DOI: 10.1038/cddis.2016.289] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/04/2016] [Accepted: 08/10/2016] [Indexed: 12/12/2022]
Abstract
The Fra-1 transcription factor is frequently upregulated in multiple types of tumors. Here we found that Fra-1 promotes miR-134 expression. miR-134 activates JNK and ERK by targeting SDS22, which in turn induces Fra-1 expression and leads to miR-134 upregulation. In addition, miR-134 augmented H2AX S139 phosphorylation by activating JNK and promoted non-homologous end joining (NHEJ)-mediated DNA repair. Therefore, ectopic miR-134 expression reduced chemosensitivity in ovarian cancer cells. Furthermore, miR-134 promotes cell proliferation, migration and invasion of ovarian cancer cells, and enhances tumor growth in vivo. Of particular significance, both Fra-1 and miR-134 are upregulated in ovarian cancer tissues, and Fra-1 and miR-134 expression is positively correlated. High levels of miR-134 expression were associated with a reduced median survival of ovarian cancer patients. Our study revealed that a Fra-1-miR-134 axis drives a positive feedback loop that amplifies ERK/JNK signaling and reduces chemosensitivity in ovarian cancer cells.
Collapse
|
21
|
4.1N is involved in a flotillin-1/β-catenin/Wnt pathway and suppresses cell proliferation and migration in non-small cell lung cancer cell lines. Tumour Biol 2016; 37:12713-12723. [DOI: 10.1007/s13277-016-5146-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/11/2016] [Indexed: 01/14/2023] Open
|
22
|
Wang Z, Zhang J, Zeng Y, Sun S, Zhang J, Zhang B, Zhu M, Ouyang R, Ma B, Ye M, An X, Liu J. Knockout of 4.1B triggers malignant transformation in SV40T-immortalized mouse embryo fibroblast cells. Mol Carcinog 2016; 56:538-549. [DOI: 10.1002/mc.22515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 05/30/2016] [Accepted: 06/14/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Zi Wang
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| | - Jingxin Zhang
- College of Life Sciences; Zhengzhou University; Zhengzhou China
| | - Yayue Zeng
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| | - Shuming Sun
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| | - Ji Zhang
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| | - Bin Zhang
- Department of Histology and Embryology; Xiangya School of Medicine, Central South University; Changsha China
| | - Min Zhu
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| | - Ruoyun Ouyang
- Department of Respiratory Medicine, Respiratory Disease Research Institute; Second XiangYa Hospital of Central South University; Changsha China
| | - Bianyin Ma
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology; College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University; Changsha China
| | - Xiuli An
- College of Life Sciences; Zhengzhou University; Zhengzhou China
- Laboratory of Membrane Biology; New York Blood Center; New York New York
| | - Jing Liu
- The State Key Laboratory of Medical Genetics and School of Life Sciences; Central South University; Changsha China
| |
Collapse
|
23
|
Choi Y, Park J, Choi Y, Ko YS, Yu DA, Kim Y, Pyo JS, Jang BG, Kim MA, Kim WH, Lee BL. c-Jun N-terminal kinase activation has a prognostic implication and is negatively associated with FOXO1 activation in gastric cancer. BMC Gastroenterol 2016; 16:59. [PMID: 27268017 PMCID: PMC4895928 DOI: 10.1186/s12876-016-0473-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 05/27/2016] [Indexed: 12/27/2022] Open
Abstract
Background Since the biological function of c-Jun N-terminal kinase (JNK) in gastric cancer remains unclear, we investigated the clinical significance of JNK activation and its association with FOXO1 activation. Methods Immunohistochemical tissue array analysis of 483 human gastric cancer specimens was performed, and the results of the immunostaining were quantified. The correlation between JNK activation (nuclear staining for pJNK) and clinicopathological features, the proliferation index, prognosis or FOXO1 inactivation (cytoplasmic staining for pFOXO1) was analyzed. The SNU-638 gastric cancer cell line was used for in vitro analysis. Results Nuclear staining of pJNK was found in 38 % of the gastric carcinomas and was higher in the early stages of pTNM (P < 0.001). pJNK staining negatively correlated with lymphatic invasion (P = 0.034) and positively correlated with intestinal type by Lauren’s classification (P = 0.037), Ki-67-labeling index (P < 0.001), cyclin D1 (P = 0.045), cyclin E (P < 0.001) and pFOXO1 (P < 0.001). JNK activation correlated with a longer patients survival (P =0.008) and patients with a JNK-active and FOXO1-inactive tumor had a higher survival rate than the remainder of the population (P = 0.004). In vitro analysis showed that JNK inhibition by SP600125 in SNU-638 cells decreased cyclin D1 protein expression and increased FOXO1 activation. Further, JNK inhibition markedly suppressed colony formation, which was partially restored by FOXO1 shRNA expression. Conclusions Our results indicate that JNK activation may serve as a valuable prognostic factor in gastric cancer, and that it is implicated in gastric tumorigenesis, at least in part, through FOXO1 inhibition.
Collapse
Affiliation(s)
- Youngsun Choi
- Department of Tumor Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Jinju Park
- Department of Tumor Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Yiseul Choi
- Department of Tumor Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Young San Ko
- Departments of Anatomy, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Da-Ae Yu
- Departments of Anatomy, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Younghoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 110-746, South Korea
| | - Bo Gun Jang
- Department of Pathology, Jeju National University Hospital, Jeju, 690-767, South Korea
| | - Min A Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | - Byung Lan Lee
- Department of Tumor Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, 110-799, South Korea. .,Departments of Anatomy, Seoul National University College of Medicine, Seoul, 110-799, South Korea. .,Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, 110-799, South Korea.
| |
Collapse
|