1
|
Wang W, Jin W, Liu X, Zheng L. Circ_0002577/miR-126-5p/MACC1 axis promotes endometrial carcinoma progression by regulation of proliferation, migration, invasion, and apoptosis of endometrial carcinoma cells. Arch Gynecol Obstet 2022; 306:481-491. [PMID: 35103833 DOI: 10.1007/s00404-022-06412-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/04/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Endometrial carcinoma (EC) is a common female reproductive malignant tumor. Circular RNAs (circRNAs) have been reported to participate in tumorigenesis, including EC. Therefore, this study was designed to clarify the role and underlying molecular mechanisms of circ_0002577 in EC. METHODS The expression levels of circ_0002577, miR-126-5p, and metastasis associated in colon cancer 1 (MACC1) was determined by real-time quantitative polymerase chain reaction (RT-qPCR) assay. The protein expression was quantified by western blot assay. The proliferation of EC cells was assessed by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT) and colony-forming assays. The migration and invasion of EC cells was measured by transwell assay. The apoptosis was determined by flow cytometry assay. Dual-luciferase reporter assay and RNA pull-down assays were performed to confirm the relationship between miR-126-5p and circ_0002577 or MACC1. The influence of circ_0002577 inhibition on tumor growth was assessed by xenograft experiment. RESULTS Circ_0002577 and MACC1 were increased while miR-126-5p was decreased in EC tissues and cells. Loss-of-functional experiment revealed that silencing of circ_0002577 inhibited the proliferation, migration, and invasion while induced apoptosis of EC cells, which were overturned by overexpression of MACC1. The upregulation of miR-126-5p also impeded proliferation and mobility while induced apoptosis of EC cells. MiR-126-5p, negatively regulating MACC1 expression, was a functional target of circ_0002577 in EC cells. Moreover, we also confirmed that suppression of circ_0002577 repressed tumor growth in vivo. CONCLUSION The contributions of the circ_0002577 in EC were contributed to its interactions with miR-126-5p and MACC1, which offered a new perspective to the roles of circ_0002577 in EC.
Collapse
Affiliation(s)
- Wenzhi Wang
- Department of Gynaecology, Shaanxi Provincial People's Hospital, No. 256, Youyi West Road, Xi'an City, 710000, Shaanxi Province, China.
| | - Wen Jin
- Department of Gynaecology, Shaanxi Provincial People's Hospital, No. 256, Youyi West Road, Xi'an City, 710000, Shaanxi Province, China
| | - Xiaoying Liu
- Department of Gynaecology, Shaanxi Provincial People's Hospital, No. 256, Youyi West Road, Xi'an City, 710000, Shaanxi Province, China
| | - Lei Zheng
- College of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Cheng H, Zhou L, Long Y, Xiang J, Chen L. MACC1 Is Associated With Epithelial-Mesenchymal Transition and Can Predict Poor Prognosis in Nasopharyngeal Carcinoma. Front Oncol 2021; 11:644120. [PMID: 33854976 PMCID: PMC8039464 DOI: 10.3389/fonc.2021.644120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 03/11/2021] [Indexed: 12/28/2022] Open
Abstract
Background Given the reported correlation between the oncogene metastasis-associated in colon cancer 1 (MACC1) and nasopharyngeal carcinoma (NPC), as well as between MACC1 and epithelial–mesenchymal transition (EMT), we speculated that EMT is a likely causative link between MACC1 expression and poor NPC prognosis. Thus, we aim to clarify the relationship between MACC1 and EMT in NPC prognosis. Material and Methods We performed immunohistochemical examination of tissue sections from 128 NPC patients that were divided into six groups corresponding to high and low protein expression of MACC1 and two EMT-related proteins, vimentin and E-cadherin, and Kaplan–Meier (KM) survival analyses were performed. Results KM survival analysis showed that upregulation of MACC1 and vimentin and downregulation of E-cadherin were significantly associated with reduced survival in NPC. Short hairpin RNA (shRNA) interference and immunoblotting in the NPC cell line HNE-1 led to increased E-cadherin but decreased vimentin levels. MACC1 overexpression was significantly correlated with poor 5-year overall survival, metastasis-free survival, and disease-free survival (P<0.05) but not with poor relapse-free survival (P>0.05). Univariate analyses revealed that MACC1, E-cadherin, and vimentin levels along with T and N tumor classifications and cancer staging are significant prognostic factors of NPC (P<0.05). Conclusion Our findings showed the association between MACC1 and EMT in NPC malignancy and support the role of MACC1 as a prognostic biomarker and molecular target for NPC treatment.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Radiation Oncology, Nanfang Hospital of Southern Medical University, Guangzhou, China.,Department of Nasopharyngeal Carcinoma, The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, China
| | - Linxiang Zhou
- Department of Nasopharyngeal Carcinoma, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Yalan Long
- Department of Nasopharyngeal Carcinoma, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Juanjuan Xiang
- Department of Nasopharyngeal Carcinoma, The First People's Hospital of Chenzhou, University of South China, Chenzhou, China
| | - Longhua Chen
- Department of Radiation Oncology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Sun L. COPS8 in cutaneous melanoma: an oncogene that accelerates the malignant development of tumor cells and predicts poor prognosis. Biosci Biotechnol Biochem 2021; 85:242-250. [PMID: 33604618 DOI: 10.1093/bbb/zbaa017] [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: 05/08/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022]
Abstract
This study aimed to investigate the roles of COP9 signalosome subunit 8 (COPS8) and its underlying mechanism in cutaneous melanoma. Bioinformatics tools were utilized to analyze the expression of COPS8 in cutaneous melanoma, while Kaplan-Meier analysis was employed to assess the correlation between COPS8 and patients' overall survival. The proliferation, migration, and invasion of cells were estimated by CCK8, colony formation, and Transwell assays. Western blot was used to check the expression of epithelial-mesenchymal transition (EMT)-related proteins. Results showed that COPS8 was up-regulated and predicted a poor clinical outcome for cutaneous melanoma patients. Knockdown of COPS8 inhibited cutaneous melanoma cell proliferation, migration and invasion, whereas overexpression of COPS8 resulted in the opposite outcomes. The up-regulation of E-cadherin and down-regulation of N-cadherin, vimentin, and snail were caused by silencing COPS8 while their expression showed contrary trends in cells with overexpressed COPS8. Collectively, COPS8 is up-regulated and promotes cutaneous melanoma progression via regulating EMT.
Collapse
Affiliation(s)
- Liangliang Sun
- Department of Dermatology, Daqing Oilfield General Hospital, No. 9 ZhongKang street, Daqing City, Heilongjiang Province, P. R. China
| |
Collapse
|
4
|
Hu X, Yuan L, Ma T. Mechanisms of JAK-STAT signaling pathway mediated by CXCL8 gene silencing on epithelial-mesenchymal transition of human cutaneous melanoma cells. Oncol Lett 2020; 20:1973-1981. [PMID: 32724443 PMCID: PMC7377181 DOI: 10.3892/ol.2020.11706] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022] Open
Abstract
Effect of CXCL8 gene silencing-mediated JAK-STAT signaling pathway on epithelial-mesenchymal transition (EMT) of human cutaneous melanoma cells was explored. Eighty patients with cutaneous melanoma were enrolled in the study. Cells were transfected accordingly and divided into five groups: The blank group (human cutaneous melanoma cells), NC group (human cutaneous melanoma cells + blank vector plasmid transfection), CXCL8 siRNA group (human cutaneous melanoma cells + CXCL8 silent expression vector plasmid transfection), AG490 group (human cutaneous melanoma cells + JAK-STAT signal pathway inhibitor transfection), CXCL8 siRNA + AG490 group (human cutaneous melanoma cells + JAK-STAT signaling pathway inhibitor + CXCL8 silent expression vector plasmid transfection). The expression levels of CXCL8, JAK2, STAT3, epithelial cadherin (E-cadherin), neurotrophic cadherin (N-cadherin) and vimentin in tissues and cells were detected by RT-qPCR and western blot analysis. CCK-8 and flow cytometry were used to detect cell proliferation and apoptosis. Compared with adjacent normal tissues, the expression of E-cadherin in human cutaneous melanoma tissues was significantly decreased, whereas the expression of CXCL8, JAK2, STAT3, vimentin and N-cadherin was significantly increased (P<0.05). Compared with the blank group, CXCL8 siRNA group and CXCL8 siRNA + AG490 group had significantly lower expression of CXCL8 (P<0.05). Compared with the blank group, the expression levels of JAK2, STAT3, vimentin and N-cadherin in CXCL8 siRNA group, AG490 group and CXCL8 siRNA + AG490 group were decreased, the expression of E-cadherin was increased, the cell proliferation ability was decreased and apoptosis was increased (P<0.05). Compared with CXCL8 siRNA group, the expression of JAK2, STAT3, vimentin and N-cadherin in CXCL8 siRNA + AG490 group were significantly decreased, the expression of E-cadherin was significantly increased, cell proliferation ability was decreased and apoptosis was increased (P<0.05). In conclusion, CXCL8 gene expression silencing may inhibit EMT and cell proliferation while promoting cell apoptosis of human cutaneous melanoma cells by inhibiting the activation of JAK-STAT signaling pathway.
Collapse
Affiliation(s)
- Xiaorui Hu
- Department of Burn and Plastic Surgery, General Hospital of Ningxia Medical University, Yinchuan, Gansu 750001, P.R. China
| | - Lili Yuan
- Department of Plastic Surgery, Qingyang People's Hospital of Gansu Province, Qingyang, Gansu 745000, P.R. China
| | - Teng Ma
- Department of Traumatic Orthopaedics, General Hospital of Ningxia Medical University, Yinchuan, Gansu 750001, P.R. China
| |
Collapse
|
5
|
Hohmann T, Hohmann U, Kolbe MR, Dahlmann M, Kobelt D, Stein U, Dehghani F. MACC1 driven alterations in cellular biomechanics facilitate cell motility in glioblastoma. Cell Commun Signal 2020; 18:85. [PMID: 32503676 PMCID: PMC7275321 DOI: 10.1186/s12964-020-00566-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/23/2020] [Indexed: 12/31/2022] Open
Abstract
Background Metastasis-associated in colon cancer 1 (MACC1) is an established marker for metastasis and tumor cell migration in a multitude of tumor entities, including glioblastoma (GBM). Nevertheless, the mechanism underlying the increased migratory capacity in GBM is not comprehensively explored. Methods We performed live cell and atomic force microscopy measurements to assess cell migration and mechanical properties of MACC1 overexpressing GBM cells. We quantified MACC1 dependent dynamics of 3D aggregate formation. For mechanistic studies we measured the expression of key adhesion molecules using qRT-PCR, and MACC1 dependent changes in short term adhesion to fibronectin and laminin. We then determined changes in sub-cellular distribution of integrins and actin in dependence of MACC1, but also in microtubule and intermediate filament organization. Results MACC1 increased the migratory speed and elastic modulus of GBM cells, but decreased cell-cell adhesion and inhibited the formation of 3D aggregates. These effects were not associated with altered mRNA expression of several key adhesion molecules or altered short-term affinity to laminin and fibronectin. MACC1 did neither change the organization of the microtubule nor intermediate filament cytoskeleton, but resulted in increased amounts of protrusive actin on laminin. Conclusion MACC1 overexpression increases elastic modulus and migration and reduces adhesion of GBM cells thereby impeding 3D aggregate formation. The underlying molecular mechanism is independent on the organization of microtubules, intermediate filaments and several key adhesion molecules, but depends on adhesion to laminin. Thus, targeting re-organization of the cytoskeleton and cell motility via MACC1 may offer a treatment option to impede GBM spreading. Video Abstract
Collapse
Affiliation(s)
- Tim Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany
| | - Urszula Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany
| | - Marc R Kolbe
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany
| | - Mathias Dahlmann
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany.
| |
Collapse
|
6
|
Jurmeister P, Bockmayr M, Treese C, Stein U, Lenze D, Jöhrens K, Friedling F, Dietel M, Klauschen F, Marsch W, Fiedler E, von Laffert M. Immunohistochemical analysis of Bcl-2, nuclear S100A4, MITF and Ki67 for risk stratification of early-stage melanoma - A combined IHC score for melanoma risk stratification. J Dtsch Dermatol Ges 2020; 17:800-808. [PMID: 31437373 DOI: 10.1111/ddg.13917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/16/2019] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVES Overall survival (OS) in patients with early-stage malignant melanoma differs. To date, there are no established prognostic markers. We aimed to contribute to a better understanding of potential prognostic immunohistochemical markers for risk stratification. PATIENTS AND METHODS 161 surgically resected early-stage malignant melanomas (stage pT1 and pT2) were analyzed for expression of 20 different proteins using immunohistochemistry. The results were correlated with OS. The cohort was randomly split into a discovery and a validation cohort. RESULTS High Bcl-2 expression, high nuclear S100A4 expression as well as a Ki67 proliferation index of ≥ 20 % were associated with shorter OS. Strong MITF immunoreactivity was a predictor for favorable prognosis. A combination of these four markers resulted in a multi-marker score with significant prognostic value in multivariate survival analysis (HR: 3.704; 95 % CI 1.484 to 9.246; p = 0.005). Furthermore, the score was able to differentiate a low-risk group with excellent OS rates (five-year survival rate: 100 %), an intermediate-risk group (five-year survival rate: 81.8 %) and a high-risk group (five-year survival rate: 52.6 %). The prognostic value was confirmed within the validation cohort. CONCLUSIONS Combined immunohistochemical analysis of Bcl-2, nuclear S100A4, Ki67 and MITF could contribute to better risk stratification of early-stage malignant melanoma patients.
Collapse
Affiliation(s)
- Philipp Jurmeister
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Charité Comprehensive Cancer Center, Berlin, Germany
| | - Michael Bockmayr
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Treese
- Berlin Institute of Health (BIH), Berlin, Germany.,Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Department of Gastroenterology, Infectious Diseases, Rheumatology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dido Lenze
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Franziska Friedling
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Manfred Dietel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frederick Klauschen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Marsch
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Eckhard Fiedler
- Department of Dermatology and Venereology, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Maximilian von Laffert
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
7
|
Jin T, Liu M, Liu Y, Li Y, Xu Z, He H, Liu J, Zhang Y, Ke Y. Lcn2-derived Circular RNA (hsa_circ_0088732) Inhibits Cell Apoptosis and Promotes EMT in Glioma via the miR-661/RAB3D Axis. Front Oncol 2020; 10:170. [PMID: 32154171 PMCID: PMC7047435 DOI: 10.3389/fonc.2020.00170] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Glioma is the most common malignant tumor of the central nervous system, and often displays invasive growth. Recently, circular RNA (circRNA), which is a novel non-coding type of RNA, has been shown to play a vital role in glioma tumorigenesis. However, the functions and mechanism of lipocalin-2 (Lcn2)-derived circular RNA (hsa_circ_0088732) in glioma progression remain unclear. Methods: We evaluated hsa_circ_0088732 expression by fluorescence in situ hybridization (FISH), Sanger sequencing, and PCR assays. Cell apoptosis was evaluated by flow cytometry and Hoechst 33258 staining. Transwell migration and invasion assays were performed to measure cell metastasis and viability. In addition, the target miRNA of hsa_circ_0088732 and the target gene of miR-661 were predicted by a bioinformatics analysis, and the interactions were verified by dual-luciferase reporter assays. RAB3D expression was analyzed by an immunochemistry assay, and E-cadherin, N-cadherin, and vimentin protein expression were examined by western blot assays. A mouse xenograft model was developed and used to analyze the effects of hsa_circ_0088732 on glioma growth in vivo. Results: We verified that hsa_circ_0088732 is circular and highly expressed in glioma tissues. Knockdown of hsa_circ_0088732 induced glioma cell apoptosis and inhibited glioma cell migration, invasion, and epithelial-mesenchymal transition (EMT). We found that hsa_circ_0088732 negatively regulated miR-661 by targeting miR-661, and RAB3D was a target gene of miR-661. In addition, inhibition of miR-661 promoted glioma cell metastasis and suppressed cell apoptosis. Knockdown of RAB3D induced cell apoptosis and suppressed cell metastasis. Moreover, hsa_circ_0088732 accelerated glioma progression through its effects on the miR-661/RAB3D axis. Finally, results from a mouse xenograft model confirmed that knockdown of hsa_circ_0088732 induced miR-661 expression, resulting in suppression of RAB3D expression and inhibition of tumor growth in vivo. Conclusion: We demonstrated that hsa_circ_0088732 facilitated glioma progression by sponging miR-661 to increase RAB3D expression. This study provides a theoretical basis for understanding the development and occurrence of glioma, as well as for the development of targeted drugs.
Collapse
Affiliation(s)
- Tao Jin
- The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Mingfa Liu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Yan Liu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Yuanzhi Li
- Department of Neurosurgery, Affiliated Hengyang Hospital of Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Zhennan Xu
- Department of Neurosurgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Haoqi He
- The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Liu
- The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuxuan Zhang
- The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiquan Ke
- The National Key Clinical Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, The Engineering Technology Research Center of Education Ministry of China, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Decoding and targeting the molecular basis of MACC1-driven metastatic spread: Lessons from big data mining and clinical-experimental approaches. Semin Cancer Biol 2019; 60:365-379. [PMID: 31430556 DOI: 10.1016/j.semcancer.2019.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Metastasis remains the key issue impacting cancer patient survival and failure or success of cancer therapies. Metastatic spread is a complex process including dissemination of single cells or collective cell migration, penetration of the blood or lymphatic vessels and seeding at a distant organ site. Hundreds of genes involved in metastasis have been identified in studies across numerous cancer types. Here, we analyzed how the metastasis-associated gene MACC1 cooperates with other genes in metastatic spread and how these coactions could be exploited by combination therapies: We performed (i) a MACC1 correlation analysis across 33 cancer types in the mRNA expression data of TCGA and (ii) a comprehensive literature search on reported MACC1 combinations and regulation mechanisms. The key genes MET, HGF and MMP7 reported together with MACC1 showed significant positive correlations with MACC1 in more than half of the cancer types included in the big data analysis. However, ten other genes also reported together with MACC1 in the literature showed significant positive correlations with MACC1 in only a minority of 5 to 15 cancer types. To uncover transcriptional regulation mechanisms that are activated simultaneously with MACC1, we isolated pan-cancer consensus lists of 1306 positively and 590 negatively MACC1-correlating genes from the TCGA data and analyzed each of these lists for sharing transcription factor binding motifs in the promotor region. In these lists, binding sites for the transcription factors TELF1, ETS2, ETV4, TEAD1, FOXO4, NFE2L1, ELK1, SP1 and NFE2L2 were significantly enriched, but none of them except SP1 was reported in combination with MACC1 in the literature. Thus, while some of the results of the big data analysis were in line with the reported experimental results, hypotheses on new genes involved in MACC1-driven metastasis formation could be generated and warrant experimental validation. Furthermore, the results of the big data analysis can help to prioritize cancer types for experimental studies and testing of combination therapies.
Collapse
|
9
|
Jurmeister P, Bockmayr M, Treese C, Stein U, Lenze D, Jöhrens K, Friedling F, Dietel M, Klauschen F, Marsch W, Fiedler E, Laffert M. Immunhistochemische Analyse von Bcl‐2, nukleärem S100A4, MITF und Ki67 zur Risikostratifizierung von Melanomen im Frühstadium – ein kombinierter immunhistochemischer Score. J Dtsch Dermatol Ges 2019; 17:800-809. [DOI: 10.1111/ddg.13917_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/16/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Philipp Jurmeister
- Institut für PathologieCharité – Universitätsmedizin Berlin
- Charité Comprehensive Cancer Center Berlin
| | - Michael Bockmayr
- Institut für PathologieCharité – Universitätsmedizin Berlin
- Klinik für pädiatrische Hämatologie und OnkologieUniversitätsklinikum Hamburg‐Eppendorf Hamburg
| | - Christoph Treese
- Berlin Institute of Health (BIH) Berlin
- Experimentelles und klinisches ForschungszentrumCharité – Universitätsmedizin Berlinund Max‐Delbrück‐Zentrum für molekulare Medizin Berlin
- Medizinische Klinik für GastroenterologieInfektiologie und RheumatologieCharité – Universitätsmedizin Berlin Campus Benjamin Franklin Berlin
| | - Ulrike Stein
- Experimentelles und klinisches ForschungszentrumCharité – Universitätsmedizin Berlinund Max‐Delbrück‐Zentrum für molekulare Medizin Berlin
- Deutsches Konsortium für Translationale Krebsforschung (DKTK) Heidelberg
| | - Dido Lenze
- Institut für PathologieCharité – Universitätsmedizin Berlin
| | | | - Franziska Friedling
- Universitätsklinik und Poliklinik für Dermatologie und Venerologie Halle (Saale)Martin‐Luther‐Universität Halle‐Wittenberg Halle (Saale)
| | - Manfred Dietel
- Institut für PathologieCharité – Universitätsmedizin Berlin
| | | | - Wolfgang Marsch
- Universitätsklinik und Poliklinik für Dermatologie und Venerologie Halle (Saale)Martin‐Luther‐Universität Halle‐Wittenberg Halle (Saale)
| | - Eckhard Fiedler
- Universitätsklinik und Poliklinik für Dermatologie und Venerologie Halle (Saale)Martin‐Luther‐Universität Halle‐Wittenberg Halle (Saale)
| | - Maximilian Laffert
- Institut für PathologieCharité – Universitätsmedizin Berlin
- Berlin Institute of Health (BIH) Berlin
| |
Collapse
|
10
|
Radhakrishnan H, Walther W, Zincke F, Kobelt D, Imbastari F, Erdem M, Kortüm B, Dahlmann M, Stein U. MACC1-the first decade of a key metastasis molecule from gene discovery to clinical translation. Cancer Metastasis Rev 2019; 37:805-820. [PMID: 30607625 DOI: 10.1007/s10555-018-9771-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Deciphering the paths to metastasis and identifying key molecules driving this process is one important issue for understanding and treatment of cancer. Such a key driver molecule is Metastasis Associated in Colon Cancer 1 (MACC1). A decade long research on this evolutionarily conserved molecule with features of a transcription factor as well as an adapter protein for versatile protein-protein interactions has shown that it has manifold properties driving tumors to their metastatic stage. MACC1 transcriptionally regulates genes involved in epithelial-mesenchymal transition (EMT), including those which are able to directly induce metastasis like c-MET, impacts tumor cell migration and invasion, and induces metastasis in solid cancers. MACC1 has proven as a valuable biomarker for prognosis of metastasis formation linked to patient survival and gives promise to also act as a predictive marker for individualized therapies in a broad variety of cancers. This review discusses the many features of MACC1 in the context of the hallmarks of cancer and the potential of this molecule as biomarker and novel therapeutic target for restriction and prevention of metastasis.
Collapse
Affiliation(s)
- Harikrishnan Radhakrishnan
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Fabian Zincke
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Francesca Imbastari
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Müge Erdem
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Mathias Dahlmann
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Consortium (DKTK), German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
11
|
Zhang R, Shi H, Ren F, Liu Z, Ji P, Zhang W, Wang W. Down-regulation of miR-338-3p and Up-regulation of MACC1 Indicated Poor Prognosis of Epithelial Ovarian Cancer Patients. J Cancer 2019; 10:1385-1392. [PMID: 31031848 PMCID: PMC6485222 DOI: 10.7150/jca.29502] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
Objective To detect the expression of microRNA-338-3p (miR-338-3p) and MET transcriptional regulator MACC1 (MACC1) gene in different ovarian tissues, to analyze their relationships, their correlations to the clinicopathologic characteristics of epithelial ovarian cancer and their significant to the progression of ovarian cancer. Methods The expression of miR-338-3p and MACC1 gene in 20 specimens of normal ovarian tissues, 20 specimens of benign epithelial ovarian tumor and 65 specimens of epithelial ovarian cancer was detected by real-time PCR method. Their interrelationships and their correlations to the clinicopathologic characteristics of epithelial ovarian cancer were analyzed. Risk factors of recurrence and death were discussed by binary Logistic regression analysis. The relations between miR-338-3p and MACC1 expression and the survival of ovarian cancer were measured by Kaplan-Meier analysis. Results The expressions of miR-338-3p and MACC1 gene in epithelial ovarian cancer tissues were (0.331±0.038) and (0.774±0.025), significant differences were noted between epithelial ovarian cancer and normal ovarian tissues, benign ovarian tumors (F=77.916, P=1.205E-18; F=77.945, P=1.187E-18). In different ovarian tissues, miR-338-3p expression was negatively correlated to MACC1 expression (r = -0.968, P<0.0001). In epithelial ovarian cancer, lower expression of miR-338-3p and higher expression of MACC1 were associated with more advanced FIGO stage, higher histological grade and developed lymph node metastasis. Down-regulation of miR-338-3p was related with the recurrence (P=0. 005, OR=12.862, 95%CI: 2.120~78.026) and death (P=0. 007, OR=12.837, 95%CI: 2.205~81.389) of ovarian cancer patients, which was showed by binary Logistic regression analysis. Compared to other patients, the overall survival rate and progression free survival rate of patients with lower miR-338-3p and higher MACC1 expression were obviously poorer (χ2=16.955, P=7.219E-5; χ2=18.929, P=2.828E-5). Conclusions Down-regulation of miR-338-3p and up-regulation of MACC1 gene were closely related with the poor prognosis of epithelial ovarian cancer patients, which could served as bio-markers of the progression and recurrence of ovarian cancer.
Collapse
Affiliation(s)
- Ruitao Zhang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China.,Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Huirong Shi
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Fang Ren
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Zheying Liu
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Pengcheng Ji
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Weiwei Zhang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Wenwen Wang
- Department of Gynecology, First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
12
|
Liang T, Wang X, Li P, Cao Y, Feng E, You G. HOXC8: a predictive glioma biomarker that induces epithelia-mesenchymal transition. Chin Neurosurg J 2018; 4:24. [PMID: 32922885 PMCID: PMC7398260 DOI: 10.1186/s41016-018-0132-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/07/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The transcription factor Homeobox C8 (HOXC8) is overexpressed and regulates many important genes involved in the proliferation and invasion of many malignant tumors. However, the function of HOXC8 in gliomas remains unclear. METHOD Based on the Chinese Glioma Genome Atlas (CGGA) set, HOXC8 expression is negatively correlated with overall survival (OS). Small interfering RNA (si-HOXC8) was used to downregulate the mRNA and protein expression levels of HOXC8 to assess glioma cell proliferation, migration and invasion. RESULTS Patients with higher HOXC8 levels showed poorer prognosis. DAVID analysis results indicated that HOXC8 was related to cell cycle, cell adhesion and immune response. In U251 and LN229 glioma cells treated with small interfering RNA for HOXC8 (si-HOXC8) for gene knockdown, significantly lower cell capacity of growth, migration and invasion was observed. Moreover, HOXC8 knockdown could reduce the protein expression of classical epithelial mesenchymal transition (EMT) related markers. CONCLUSION HOXC8 may play an important role in glioma proliferation, migration and invasion. These findings indicated that HOXC8 may constitute a novel target for glioma treatment.
Collapse
Affiliation(s)
- Tingyu Liang
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100020 China
| | | | - Peiliang Li
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100020 China
| | - Yang Cao
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100020 China
| | - Enshan Feng
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, 100020 China
| | - Gan You
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 TiantanXili, Dongcheng District, Beijing, 100050 China
- Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, No. 6 TiantanXili, Dongcheng District, Beijing, 100050 China
| |
Collapse
|
13
|
Lin G, Yu B, Liang Z, Li L, Qu S, Chen K, Zhou L, Lu Q, Sun Y, Zhu X. Silencing of c-jun decreases cell migration, invasion, and EMT in radioresistant human nasopharyngeal carcinoma cell line CNE-2R. Onco Targets Ther 2018; 11:3805-3815. [PMID: 30013361 PMCID: PMC6038861 DOI: 10.2147/ott.s162700] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Previously, we found that c-jun was highly expressed in radiation-resistant human nasopharyngeal carcinoma cells (CNE-2R) compared with human nasopharyngeal carcinoma cells (CNE-2). MATERIALS AND METHODS In this study, we first used the scratch assays and transwell assays to detect the migration and invasion of CNE-2R and CNE-2 cells and tested the epithelial mesenchymal transformation (EMT)-related proteins E-cadherin and N-cadherin by Western blot analysis. Subsequently, c-jun was knocked down to establish the effect of c-jun on EMT, migration, and invasion of CNE-2R cells both in vitro and in vivo. RESULTS A high EMT level, CNE-2R cells were more capable of migration and invasion than CNE-2 cells. Moreover, silencing of c-jun has upregulated the expression of E-cadherin and downregulated N-cadherin in CNE-2R cells, and subsequently the migration and invasion capacity of the cells was decreased. Consistent with in vitro results, in vivo studies indicated that the c-jun silencing reduced pulmonary migration of CNE-2R cells. Immunohistochemistry of lung metastatic tumor tissue showed that E-cadherin was upregulated, and N-cadherin was downregulated. CONCLUSION These findings suggest that silencing of c-jun in CNE-2R cells reduces cells migration, invasion, and EMT both in vitro and in vivo.
Collapse
Affiliation(s)
- Guoxiang Lin
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Binbin Yu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Zhongguo Liang
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Ling Li
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530021, People's Republic of China,
| | - Song Qu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530021, People's Republic of China,
| | - Kaihua Chen
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Lei Zhou
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Qiteng Lu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Yongchu Sun
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| | - Xiaodong Zhu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Guangxi Medical University and Cancer Institute of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, People's Republic of China,
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
- Key Laboratory of High-Incidence-Tumor Prevention and Treatment (Guangxi Medical University), Ministry of Education, Nanning, Guangxi 530021, People's Republic of China,
- Department of Oncology, Affiliated Wuming Hospital of Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China,
| |
Collapse
|
14
|
Wang Y, Zhang S, Liu J, Fang B, Yao J, Cheng B. Matrine inhibits the invasive and migratory properties of human hepatocellular carcinoma by regulating epithelial‑mesenchymal transition. Mol Med Rep 2018; 18:911-919. [PMID: 29845189 PMCID: PMC6059723 DOI: 10.3892/mmr.2018.9023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
Matrine has been reported to be an effective anti-tumor therapy; however, the anti-metastatic effects of matrine on hepatocellular carcinoma (HCC) and the molecular mechanism(s) involved remain unclear. Therefore, the aims of the present study were to evaluate the effects of matrine on hepatoma and to determine the associated mechanism(s) involved. In the present study, matrine was confirmed to prevent the proliferation of HCC cells and it was observed that matrine also inhibited the migratory, and invasive capabilities of HCC at non-toxic concentrations. Additionally, matrine increased epithelial-cadherin expression and decreased the expression levels of vimentin, matrix metalloproteinase (MMP)2, MMP9, zinc finger protein SNAI1 and zinc finger protein SNAI2. These results indicate that the anti-metastatic effect of matrine may be associated with epithelial-mesenchymal transition (EMT). Furthermore, matrine can increase phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN (PTEN) expression and reduce phosphorylated-protein kinase B (Akt) levels. In conclusion, these results suggested that matrine is a potential therapeutic agent that can suppress cancer-associated invasion and migration via PTEN/Akt-dependent inhibition of EMT.
Collapse
Affiliation(s)
- Yuwen Wang
- Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong 518110, P.R. China
| | - Shujun Zhang
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jia Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Biaobiao Fang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Jie Yao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| | - Binglin Cheng
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150000, P.R. China
| |
Collapse
|
15
|
Ozturk E, Aksoy SAK, Ugras N, Tunca B, Ceylan S, Tezcan G, Yilmazlar T, Yerci O, Egeli U, Cecener G. Coexistence of MACC1 and NM23-H1 dysregulation and tumor budding promise early prognostic evidence for recurrence risk of early-stage colon cancer. APMIS 2018; 126:99-108. [DOI: 10.1111/apm.12801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Ersin Ozturk
- Department of General Surgery; Medical Faculty; Uludag University; Bursa Turkey
| | - Secil AK Aksoy
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
| | - Nesrin Ugras
- Department of Pathology; Medical Faculty; Uludag University; Bursa Turkey
| | - Berrin Tunca
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
| | - Serkan Ceylan
- Department of General Surgery; Medical Faculty; Uludag University; Bursa Turkey
| | - Gulcin Tezcan
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
- Institute of Fundamental Medicine and Biology; Kazan Federal University; Kazan Tatarstan Russia
| | - Tuncay Yilmazlar
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
| | - Omer Yerci
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
| | - Unal Egeli
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
| | - Gulsah Cecener
- Department of Medical Biology; Medical Faculty; Uludag University; Bursa Turkey
| |
Collapse
|
16
|
Yang Z, Ji L, Jiang G, Liu R, Liu Z, Yang Y, Ma Q, Zhao H. FL118, a novel camptothecin analogue, suppressed migration and invasion of human breast cancer cells by inhibiting epithelial-mesenchymal transition via the Wnt/β-catenin signaling pathway. Biosci Trends 2018; 12:40-46. [DOI: 10.5582/bst.2017.01288] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Zhihong Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Lixia Ji
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Guohui Jiang
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Ranran Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Zhantao Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Yuecheng Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Qingxia Ma
- Department of Pharmacology, School of Pharmacy, Qingdao University
| | - Hongqin Zhao
- Department of Pharmacology, School of Pharmacy, Qingdao University
| |
Collapse
|
17
|
Dong G, Wang M, Gu G, Li S, Sun X, Li Z, Cai H, Zhu Z. MACC1 and HGF are associated with survival in patients with gastric cancer. Oncol Lett 2017; 15:3207-3213. [PMID: 29435059 DOI: 10.3892/ol.2017.7710] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 11/02/2017] [Indexed: 02/06/2023] Open
Abstract
Metastasis-associsated in colon cancer 1 (MACC1), a newly identified oncogene, promotes tumor cell proliferation and invasion. In the present study, the expression of MACC1, hepatocyte growth factor (HGF) and its receptor, MET proto-oncogene (c-Met), was investigated in human gastric cancer tissues and adjacent normal tissues by immunohistochemistry. The association between the expression levels of the proteins and the clinicopathological parameters of the tumors were statistically analyzed. Furthermore, lentiviral particles expressing MACC1 were used to infect the hepatic satellite cell (HSC) line LX2. The expression of α-smooth muscle actin (SMA), HGF, matrix metallopeptidase (MMP)-2 and MMP-9 in human HSCs was examined by western blotting and reverse transcription-quantitative polymerase chain reaction. Transwell assays were used to measure the effect of MACC1-infected or non-infected HSCs on the migration and invasion abilities of MKN45 and MKN74 gastric carcinoma cells in vitro. The results demonstrated that positive protein expression of MACC1, HGF and c-Met was significantly higher in human gastric cancer tissues compared with adjacent normal tissues. Positive expression of MACC1 and c-Met in gastric cancer tissues had no correlation with the sex, age, tumor location and peritoneal metastasis of patients, but was significantly correlated with tumor size, depth of tumor invasion, lymph node metastasis, TNM stage, histological differentiation, and overall (5 years) and disease-free survival (5 years). Positive expression of each MACC1, HGF and c-Met protein was demonstrated to be positively correlated with each other in human gastric cancer tissues. Western blotting results confirmed that MACC1 protein was overexpressed in MACC1-overexpressing lentivirus-infected HSCs. Overexpression of MACC1 significantly increased HGF, MMP-2, MMP-9 and α-SMA expression levels in HSCs. Results from the Transwell assays indicated an increase in the number of MKN45 or MKN74 cells migrating towards MACC1-overexpressing HSCs, compared with control HSCs. These findings suggested that MACC1 may regulate the expression of HGF, MMP-2 and MMP-9 in HSCs, and may thus promote migration and invasion of gastric carcinoma cells. MACC1, HGF and c-Met might cooperatively participate in the malignant progression of gastric cancer. In conclusion, MACC1 might serve as a useful molecular target for the diagnosis, treatment and prognosis of gastric cancer.
Collapse
Affiliation(s)
- Guokai Dong
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Man Wang
- Department of Medical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Guangfu Gu
- Department of Medical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Shanshan Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Xiaoming Sun
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Zhouru Li
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Hongxing Cai
- Department of Forensic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Zhengqiu Zhu
- Department of Medical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
18
|
Fan JY, Zhang Y, Guo Q. MACC1 regulatory network in tumor metastasis. Shijie Huaren Xiaohua Zazhi 2017; 25:989-995. [DOI: 10.11569/wcjd.v25.i11.989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The MACC1 gene was firstly identified in colorectal cancer. Recently, abnormal upregulation of MACC1 has been detected in multiple tumors. The expression of MACC1 is shown to be positively associated with tumor metastasis, but negatively with prognosis of patients, and it represents a potential therapeutic target for anti-tumor strategies. MACC1 has increasingly emerged as a key regulator in metastatic processes, and it has been identified to be able to maintain multiple tumor-associated signaling pathways, transactivate oncogenic genes, and regulate epithelial-mesenchymal transition and tumor vascularization. On the other hand, MACC1 is regulated and influenced by non-coding RNAs and SNPs. The present review will summarize the recent progress in understanding the role of the MACC1 regulatory network in tumor metastasis.
Collapse
|
19
|
Bai C, Liang S, Wang Y, Jiao B. Knocking down TCF8 inhibits high glucose- and angiotensin II-induced epithelial to mesenchymal transition in podocytes. Biosci Trends 2017; 11:77-84. [PMID: 28111379 DOI: 10.5582/bst.2016.01224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Epithelial to mesenchymal transition (EMT) is a physiological phenomenon in mammalian embryogenesis by which epithelial cells become mesenchymal stem cells. Studies have indicated that an inappropriate EMT plays a key role in a variety of pathogenic processes such as embryonic development and tumor metastasis. Moreover, recent studies have indicated EMT also plays an important role in renal fibrosis. In the current study, glucose and angiotensin II promoted EMT in podocytes as well as changes in the cellular morphology of podocytes. A high concentration of glucose and angiotensin II also promoted podocyte movement and migration. Moreover, a high concentration of glucose and angiotensin II promoted TCF8 expression. Inhibiting TCF8 expression with siRNA reversed EMT in podocytes in the presence of a high concentration of glucose and angiotensin. Inhibiting TCF8 expression also reversed changes in cellular morphology and podocyte movement and migration. Therefore, glucose and angiotensin II may promote EMT in podocytes via TCF8.
Collapse
Affiliation(s)
- Changhuan Bai
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University
| | | | | | | |
Collapse
|