1
|
He P, Liu Z, Qi J, Shan J, Sheng J. Long noncoding RNA LINC00885 upregulates NCK1 to promote cell viability and migration of triple-negative breast cancer cells through sponging miR-654-3p. Cancer Biomark 2024; 39:63-78. [PMID: 37694355 DOI: 10.3233/cbm-230143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
BACKGROUND LINC00885 is a novel oncogenic long noncoding RNA (LncRNA) which is upregulated in various types of cancer, but its function in triple-negative breast cancer (TNBC) remains unknown. OBJECTIVE This study aimed to determine the role of LINC00885 on TNBC development. METHODS Clinical interrelation and survival analysis were determined using online database. The CCK-8 and Transwell assays were used to detect the proliferation and migration behaviors in TNBC cell lines. The interaction among genes was detected by RNA pull down assay. RESULTS LncRNA LINC00885 was highly expressed in TNBC compared to normal breast like. Low levels of LINC00885 was related to good prognosis in TNBC patients compared to TNBC patients with high LINC00885. LINC00885-downregulation inhibited, whereas LINC00885-overexpression promoted the proliferation and migration capability of TNBC cell lines. In TNBC cell lines, noncatalytic region of tyrosine kinase 1 (NCK1) expression was positively associated with LINC00885 expression, and shRNA-mediated the depletion of NCK1 significantly abolished LINC00885 upregulation-mediated pro-tumor effects. Combined with online databases, miR-654-3p was screened as the direct target gene of LINC00885, which could directly bind to 3'-untranslated regions (3'-UTR) of NCK1, resulting in the decreased expression of NCK1 in TNBC cell lines. LINC00885 overexpression-mediated the upregulation of NCK1 was abrogated by miR-654-3p mimics. MiR-654-3p mimics significantly rescued the tumor promotive role caused by LINC00885-overexpression. However, exogenous NCK1 notably eliminated the anti-tumor effects caused by miR-654-3p mimics in LINC00885-overexpressed cells. CONCLUSIONS LINC00885 is expressed at a high level in TNBC. LINC00885 promoted proliferation and migration by regulating the miR-654-3p/NCK1 axis in TNBC cell lines. Possibly, LINC00885 can be served as a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Peina He
- Department of Medicine, Pingdingshan University, Pingdingshan, Henan, China
| | - Zhi Liu
- Department of Medicine, Pingdingshan University, Pingdingshan, Henan, China
| | - Jinxu Qi
- Department of Medicine, Pingdingshan University, Pingdingshan, Henan, China
| | - Junrao Shan
- Department of Medicine, Pingdingshan University, Pingdingshan, Henan, China
| | - Jianyun Sheng
- Department of Gynecotokology, The First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| |
Collapse
|
2
|
Szigeti K, Ihnatovych I, Rosas N, Dorn RP, Notari E, Cortes Gomez E, He M, Maly I, Prasad S, Nimmer E, Heo Y, Fuchsova B, Bennett DA, Hofmann WA, Pralle A, Bae Y, Wang J. Neuronal actin cytoskeleton gain of function in the human brain. EBioMedicine 2023; 95:104725. [PMID: 37517100 PMCID: PMC10404607 DOI: 10.1016/j.ebiom.2023.104725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/21/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND While advancements in imaging techniques have led to major strides in deciphering the human brain, successful interventions are elusive and represent some of the most persistent translational gaps in medicine. Human restricted CHRFAM7A has been associated with neuropsychiatric disorders. METHODS The physiological role of CHRFAM7A in human brain is explored using multiomics approach on 600 post mortem human brain tissue samples. The emerging pathways and mechanistic hypotheses are tested and validated in an isogenic hiPSC model of CHRFAM7A knock-in medial ganglionic eminence progenitors and neurons. FINDINGS CHRFAM7A is identified as a modulator of intracellular calcium dynamics and an upstream regulator of Rac1. Rac1 activation re-designs the actin cytoskeleton leading to dynamic actin driven remodeling of membrane protrusion and a switch from filopodia to lamellipodia. The reinforced cytoskeleton leads to an advantage to tolerate stiffer mechanical properties of the extracellular environment. INTERPRETATION CHRFAM7A modifies the actin cytoskeleton to a more dynamic and stiffness resistant state in an α7nAChR dependent manner. CHRFAM7A may facilitate neuronal adaptation to changes in the brain environment in physiological and pathological conditions contributing to risk or recovery. Understanding how CHRFAM7A affects human brain requires human studies in the areas of memory formation and erasure, cognitive reserve, and neuronal plasticity. FUNDING This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti). Also, in part by the International Society for Neurochemistry (ISN) and The Company of Biologists (Nicolas Rosas). ROSMAP is supported by NIA grants P30AG10161, P30AG72975, R01AG15819, R01AG17917. U01AG46152, and U01AG61356.
Collapse
Affiliation(s)
- Kinga Szigeti
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA.
| | - Ivanna Ihnatovych
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Nicolás Rosas
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA; Instituto de Investigaciones Biotecnológicas, Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de, Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Ryu P Dorn
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Emily Notari
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | | | - Muye He
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Ivan Maly
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Shreyas Prasad
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Erik Nimmer
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yuna Heo
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Beata Fuchsova
- Instituto de Investigaciones Biotecnológicas, Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de, Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Wilma A Hofmann
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Arnd Pralle
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Yongho Bae
- State University of New York at Buffalo, 875 Ellicott St., Buffalo, NY, 14203, USA
| | - Jianmin Wang
- Roswell Park Comprehensive Cancer Center, 665 Elm St, Buffalo, NY 14203, USA
| |
Collapse
|
3
|
Tan S, Chen Y, Du S, Li W, Liu P, Zhao J, Yang P, Cai J, Gao R, Wang Z. TIE2-high cervical cancer cells promote tumor angiogenesis by upregulating TIE2 and VEGFR2 in endothelial cells. Transl Oncol 2022; 26:101539. [PMID: 36116242 PMCID: PMC9483782 DOI: 10.1016/j.tranon.2022.101539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/21/2022] [Accepted: 09/08/2022] [Indexed: 10/25/2022] Open
Abstract
Tyrosine kinase with immunoglobulin and epidermal growth factor homology domains 2 (TIE2), the receptor for angiopoietins, has been found highly expressed in cervical cancer and associated with poor prognosis. However, the potential role of tumoral TIE2 in cervical cancer angiogenesis and the underlying mechanisms remain unexplored. Here, based on multicolor immunofluorescence of 64 cervical cancer tissues, we found that TIE2 level in cervical cancer cells was positively related to shorter survival and higher microvessel density in tumor. In vitro and in vivo experiments verified that TIE2-high cervical cancer cells could promote tumor angiogenesis. TIE2-high tumor cells induced an amplified expression of TIE2 and vascular endothelial growth factor receptor 2(VEGFR2) in HUVECs to promote angiogenesis via TIE2 -AKT/MAPK signals, which could be reversed or partially reversed by TIE2, AKT or MAPK inhibitors and activated by angiopoietin-1 and angiopoietin-2. In conclusion, TIE2-high cervical cancer cells promote tumor angiogenesis by upregulating TIE2 and VEGFR2 in endothelial cells via TIE2-AKT/MAPK axis inside tumor cells.
Collapse
Affiliation(s)
- Shuran Tan
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Yuanyuan Chen
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Shi Du
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Wenhan Li
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Pan Liu
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Jing Zhao
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Ping Yang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China
| | - Rui Gao
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China.
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei 430022, China.
| |
Collapse
|
4
|
Exogenous Klotho ameliorates extracellular matrix degradation and angiogenesis in intervertebral disc degeneration via inhibition of the Rac1/PAK1/MMP-2 signaling axis. Mech Ageing Dev 2022; 207:111715. [PMID: 35952859 DOI: 10.1016/j.mad.2022.111715] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/21/2022]
Abstract
Intervertebral disc degeneration (IDD) is highly ubiquitous in the aged population and is an essential factor for low back pain and spinal disability. Because of the association between IDD and senescence, we investigated the ability of the anti-aging drug Klotho to inhibit age-dependent advancement of nucleus pulposus cell (NPC) degeneration. The results indicated that 400 pM exogenous Klotho significantly ameliorated extracellular matrix degradation and angiogenesis. Moreover, we demonstrated that the suppression of angiogenesis and extracellular matrix catabolism was related to inhibition of the Ras-related C3 botulinum toxin substrate 1 (Rac1)/PAK1 axis and matrix metalloproteinase 2 protein expression by exogenous Klotho cotreatment with a Rac1 inhibitor, gene overexpression in NPCs, and stimulation of human umbilical vein endothelial cells with conditioned medium from NPCs. The treatment also preserved the NPC phenotype, viability, and matrix content. In conclusion, these results suggest that the new anti-aging drug Klotho is a potential treatment strategy to mitigate IDD, and thus, provides an innovative understanding of the molecular mechanism of IDD. DATA AVAILABILITY: All data supporting the findings of this study are available from the corresponding authors upon reasonable request.
Collapse
|
5
|
Liu M, Wei D, Nie Q, Peng L, He L, Cui Y, Ye Y. Uncovering of potential molecular markers for cervical squamous cell carcinoma (CESC) based on analysis of methylated-differentially expressed genes. Taiwan J Obstet Gynecol 2022; 61:663-671. [PMID: 35779918 DOI: 10.1016/j.tjog.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Cervical squamous cell carcinoma (CESC) is a cancer with high mortality caused by human papillomavirus. The aim of this study was to uncover potential CESC biomarkers to help early diagnosis and treatment. MATERIALS AND METHODS The mRNA transcriptome data and DNA methylation data were downloaded from database for the identification of differentially expressed mRNAs (DEmRNAs) and DNA methylation analysis. Functional analysis was used to reveal the molecular functions. Then, the association between differential methylation and DEmRNA was analyzed. Protein-protein interaction (PPI) network analysis was performed on the selected differentially methylated genes (DEGs). Subsequently, we analyzed the prognosis and constructed a prognostic risk model. We also performed diagnostic analyses of risk model genes. In addition, we verified the protein expression level of identified DEGs. RESULTS 1438 DEmRNAs, 1669 differentially methylated sites (DMSs), 46 differentially methylated CpG islands and 53 differential methylation genes (DMGs) were obtained. In PPI, the highest interaction scores were MX2 and IRF8, and their interaction score was 0.928. Interestingly, 5 DMGs were found to be associated with CESC prognosis. In addition, our results demonstrated that high risk score was associated with poor prognosis of CESC. Furthermore, it was found that ZIK1, ZNRF2, HHEX, VCAM1 could be diagnostic molecular markers for CESC. CONCLUSION Analysis of methylated-differentially expressed genes may contribute to the identification of early diagnosis and therapeutic targets of CESC. In addition, a prognostic model based on 5 DMGs can be used to predict the prognosis of CESC.
Collapse
Affiliation(s)
- Miaomiao Liu
- Department of Medical Imaging, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, China; The Fifth Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Dong Wei
- Department of Urology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Qian Nie
- China Physical Examination Center of Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Lili Peng
- The Fifth Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Liya He
- The Fifth Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Yujie Cui
- The Fifth Department of Oncology, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China
| | - Yuquan Ye
- Department of Medical Imaging, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, China; Department of Ultrasound, Hebei General Hospital, 348 Heping West Road, Shijiazhuang, China.
| |
Collapse
|
6
|
Vignet P, Coquet J, Auber S, Boudet M, Siegel A, Théret N. Discrete modeling for integration and analysis of large-scale signaling networks. PLoS Comput Biol 2022; 18:e1010175. [PMID: 35696426 PMCID: PMC9232147 DOI: 10.1371/journal.pcbi.1010175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/24/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022] Open
Abstract
Most biological processes are orchestrated by large-scale molecular networks which are described in large-scale model repositories and whose dynamics are extremely complex. An observed phenotype is a state of this system that results from control mechanisms whose identification is key to its understanding. The Biological Pathway Exchange (BioPAX) format is widely used to standardize the biological information relative to regulatory processes. However, few modeling approaches developed so far enable for computing the events that control a phenotype in large-scale networks. Here we developed an integrated approach to build large-scale dynamic networks from BioPAX knowledge databases in order to analyse trajectories and to identify sets of biological entities that control a phenotype. The Cadbiom approach relies on the guarded transitions formalism, a discrete modeling approach which models a system dynamics by taking into account competition and cooperation events in chains of reactions. The method can be applied to every BioPAX (large-scale) model thanks to a specific package which automatically generates Cadbiom models from BioPAX files. The Cadbiom framework was applied to the BioPAX version of two resources (PID, KEGG) of the Pathway Commons database and to the Atlas of Cancer Signalling Network (ACSN). As a case-study, it was used to characterize sets of biological entities implicated in the epithelial-mesenchymal transition. Our results highlight the similarities between the PID and ACSN resources in terms of biological content, and underline the heterogeneity of usage of the BioPAX semantics limiting the fusion of models that require curation. Causality analyses demonstrate the smart complementarity of the databases in terms of combinatorics of controllers that explain a phenotype. From a biological perspective, our results show the specificity of controllers for epithelial and mesenchymal phenotypes that are consistent with the literature and identify a novel signature for intermediate states. The computation of sets of biological entities implicated in phenotypes is hampered by the complex nature of controllers acting in competitive or cooperative combinations. These biological mechanisms are underlied by chains of reactions involving interactions between biomolecules (DNA, RNA, proteins, lipids, complexes, etc.), all of which form complex networks. Hence, the identification of controllers relies on computational methods for dynamical systems, which require the biological information about the interactions to be translated into a formal language. The BioPAX standard is a reference ontology associated with a description language to describe biological mechanisms, which satisfies the Linked Open Data initiative recommendations for data interoperability. Although it has been widely adopted by the community to describe biological pathways, no computational method is able of studying the dynamics of the networks described in the BioPAX large-scale resources. To solve this issue, our Cadbiom framework was designed to automatically transcribe the biological systems knowledge of large-scale BioPAX networks into discrete models. The framework then identifies the trajectories that explain a biological phenotype (e.g., all the biomolecules that are activated to induce the expression of a gene). Here, we created Cadbiom models from three biological pathway databases (KEGG, PID and ACSN). The comparative analysis of these models highlighted the diversity of molecules in sets of biological entities that can explain a same phenotype. The application of our framework to the search of biomolecules regulating the epithelial-mesenchymal transition not only confirmed known pathways in the control of epithelial or mesenchymal cell markers but also highlighted new pathways for transient states.
Collapse
Affiliation(s)
- Pierre Vignet
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
| | - Jean Coquet
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
| | - Sébastien Auber
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
| | - Matéo Boudet
- IGEPP, Agrocampus Ouest, INRAE, Université de Rennes 1, Le Rheu, France
| | - Anne Siegel
- Univ Rennes, Inria, CNRS, IRISA, UMR 6074, Rennes, France
- * E-mail: (AS); (NT)
| | - Nathalie Théret
- Univ Rennes, Inserm, EHESP, Irset, UMR S1085, Rennes, France
- * E-mail: (AS); (NT)
| |
Collapse
|
7
|
He P, Sheng J, Qi J, Bai X, Li J, Wang F, Yuan Y, Zheng X. STAT3-induced NCK1 elevation promotes migration of triple-negative breast cancer cells via regulating ERK1/2 signaling. Mol Biol Rep 2021; 49:267-278. [PMID: 34846647 DOI: 10.1007/s11033-021-06868-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Noncatalytic region of tyrosine kinase 1 (NCK1) plays a key role in extracellular matrix degradation, which is required for the metastasis of triple-negative breast cancer (TNBC). However, the role NCK1 plays in the metastatic progression of TNBC is unknown. METHODS AND RESULTS Based on online databases, NCK1 was found to be highly expressed in TNBC as compared to normal breast-like subjects, which was also confirmed using TNBC cells and a tissue microarray. NCK1 expression gradually decreased with increased tumor stage. High NCK1 expression displayed a poor prognosis in lymph node-positive metastatic TNBC patients, but not in lymph node-negative patients. Using transwell assays and immunoblotting, we confirmed that NCK1 overexpression promoted, while NCK1 downregulation inhibited migration capabilities, as well as the expression of matrix metalloproteinases (MMP2/9), uridylyl phosphate adenosine, and plasminogen activator inhibitor-1 in TNBC cells. Mechanistically, NCK1 upregulation mediated the activation of MMP2/9 through ERK1/2 activity. Signal transducer and activator of transcription 3 (STAT3) was positively correlated with NCK1. STAT3 could directly bind to the promoter region of NCK1 to promote its expression and was accompanied by the elevation of MMP2/9 and ERK1/2 signaling, which were partially abolished by the knockdown of NCK1 in TNBC cells. CONCLUSIONS NCK1 may serve as a diagnostic and prognostic marker of metastatic TNBC. STAT3 upregulation promoted the expression of NCK1, which subsequently induced the migration and activity of MMPs in a ERK1/2 signaling-dependent manner in TNBC cells. NCK1 is a promising target for improving TNBC migration.
Collapse
Affiliation(s)
- Peina He
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Jianyun Sheng
- Department of Gynecotokology, Pingdingshan First People's Hospital, Pingdingshan, 410402, China
| | - Jinxu Qi
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Xianguang Bai
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Jiaxin Li
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Fubao Wang
- Department of Gynecotokology, Pingdingshan First People's Hospital, Pingdingshan, 410402, China
| | - Yamin Yuan
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China
| | - Xinhua Zheng
- Department of Medicine, Pingdingshan University, Chongwen Rd., Xincheng District, Pingdingshan, 467092, China.
| |
Collapse
|
8
|
Guo Q, Wang H, Xu Y, Wang M, Tian Z. miR-374a-5p inhibits non-small cell lung cancer cell proliferation and migration via targeting NCK1. Exp Ther Med 2021; 22:943. [PMID: 34306207 PMCID: PMC8281440 DOI: 10.3892/etm.2021.10375] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 04/16/2021] [Indexed: 12/24/2022] Open
Abstract
Emerging studies have indicated that microRNAs (miRNAs/miRs) are involved in regulating non-small cell lung cancer (NSCLC)-associated processes. The present study aimed to evaluate the biological roles of miR-374a-5p in NSCLC. Using reverse transcription-quantitative PCR, the expression levels of miR-374a-5p were determined in NSCLC cells and a normal cell line. Functional experiments were performed to investigate the functions of miR-374a-5p in NSCLC. A luciferase activity reporter assay and rescue experiments were performed to validate NCK adaptor protein 1 (NCK1) as a functional target of miR-374a-5p. It was demonstrated that miR-374a-5p levels were decreased in NSCLC cell lines compared with those in a normal cell line. Furthermore, overexpression of miR-374a-5p inhibited NSCLC cell proliferation and migration in vitro. Of note, NCK1 overexpression reversed the effects of miR-375a-5p on NSCLC cell proliferation and migration. The present results confirmed the tumor suppressor role of miR-374a-5p via targeting NCK1 in NSCLC, indicating the importance of the miR-374a-5p/NCK1 axis in NSCLC.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China.,Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Haijun Wang
- Department of Thoracic Surgery, Xingtai People's Hospital, Xingtai, Hebei 054000, P.R. China
| | - Yanzhao Xu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Mingbo Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
9
|
Sundaram MK, Almutary AG, Alsulimani A, Ahmad SR, Somvanshi P, Bhardwaj T, Pellicano R, Fagoonee S, Hussain A, Haque S. Antineoplastic action of sulforaphane on HeLa cells by modulation of signaling pathways and epigenetic pathways. Minerva Med 2021; 112:792-803. [PMID: 34114450 DOI: 10.23736/s0026-4806.21.07656-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Epigenetic modifications alter signaling and molecular pathways and are an important therapeutic target. This study examined the effect of sulforaphane on molecular targets in HeLa cells. METHODS Quantitative PCR of various molecular targets was performed. Activity of epigenetic enzymes was measured by ELISA and molecular docking analysis was conducted. Promoter methylation of some tumour suppressor genes was quantified using PCR based methylation array. In-silico protein-protein interaction network analysis was performed to understand the effect of transcriptional changes. RESULTS Quantitative PCR demonstrated the transcriptional modulation of genes involved in proliferation, metastasis, inflammation, signal transduction pathways and chromatin modifiers. Sulforaphane reduced the enzymatic activity of DNA methyl transferases, histone deacetylases and histone methyltransferases. Molecular docking results suggest that sulforaphane competitively inhibited several DNA methyl transferases and histone deacetylases. Promoter 5'CpG methylation levels of selected tumour suppressor genes was found to be reduced which correlated with their transcriptional increase as well modulation of epigenetic enzymes. Further, protein-protein interaction network analysis discerned the participation of genes towards cancer pathways. Functional enrichment and pathway-based analysis represented the modulation of epigenetic and signaling pathways on sulforaphane treatment. CONCLUSIONS The modulation in transcriptional status of epigenetic regulators, genes involved in tumorigenesis resulting in tumour suppressor genes demethylation and re-expression underscores the mechanism behind the anti-cancer effect of sulforaphane on HeLa cells.
Collapse
Affiliation(s)
- Madhumitha K Sundaram
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Abdulmajeed G Almutary
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Alsulimani
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - S Rehan Ahmad
- Hiralal Mazumdar Memorial College of Women, Kolkata, India
| | - Pallavi Somvanshi
- School of Computational & Integrative Sciences (SC&IS), Jawaharlal Nehru University, JNU Campus, New Delhi, India
| | - Tulika Bhardwaj
- School of Computational & Integrative Sciences (SC&IS), Jawaharlal Nehru University, JNU Campus, New Delhi, India
| | | | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates -
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
10
|
Liang J, Oyang L, Rao S, Han Y, Luo X, Yi P, Lin J, Xia L, Hu J, Tan S, Tang L, Pan Q, Tang Y, Zhou Y, Liao Q. Rac1, A Potential Target for Tumor Therapy. Front Oncol 2021; 11:674426. [PMID: 34079763 PMCID: PMC8165220 DOI: 10.3389/fonc.2021.674426] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
RAS-related C3 botulinum toxin substrate 1 (Rac.1) is one of the important members of Rho GTPases. It is well known that Rac1 is a cytoskeleton regulation protein that regulates cell adhesion, morphology, and movement. Rac1 is highly expressed in different types of tumors, which is related to poor prognosis. Studies have shown that Rac1 not only participates in the tumor cell cycle, apoptosis, proliferation, invasion, migration and angiogenesis, but also participates in the regulation of tumor stem cell, thus promoting the occurrence of tumors. Rac1 also plays a key role in anti-tumor therapy and participates in immune escape mediated by the tumor microenvironment. In addition, the good prospects of Rac1 inhibitors in cancer prevention and treatment are exciting. Therefore, Rac1 is considered as a potential target for the prevention and treatment of cancer. The necessity and importance of Rac1 are obvious, but it still needs further study.
Collapse
Affiliation(s)
- Jiaxin Liang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shan Rao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Pin Yi
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiaqi Hu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Lu Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,University of South China, Hengyang, China
| | - Qing Pan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,University of South China, Hengyang, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Clinical Research Center for Wound Healing in Hunan Province, Changsha, China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Clinical Research Center for Wound Healing in Hunan Province, Changsha, China
| |
Collapse
|
11
|
Xu J, Ma X, Yang H, Zhang J, Cai G, Yao N. MiR-509-3p Induces Apoptosis and Affects the Chemosensitivity of Cervical Cancer Cells by Targeting the RAC1/PAK1/LIMK1/Cofilin Pathway. Chem Pharm Bull (Tokyo) 2021; 69:325-332. [PMID: 33790078 DOI: 10.1248/cpb.c20-00600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chemoresistance is one of the main factors of treatment failure of cervical cancer (CC). Here, we intended to discover the role and mechanism of miR-509-5p in the paclitaxel chemoresistance of CC cells. RT-PCR was conducted to verify miR-509-3p expression. HCC94 and C-33A paclitaxel-resistant CC cell models were constructed. Additionally, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry were performed to verify the viability and apoptosis of HCC94 and C-33A cells after upregulating miR-509-3p. Besides, the downstream target of miR-509-3p was analyzed by bioinformatics, and the targeted relationship between miR-509-3p and RAC1 was identified by the dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. Further, the expression of apoptotic proteins (Bcl2, Bax, and Caspase3) and the RAC1/PAK1/LIMK1/Cofilin pathway was monitored by Western blot. The result showed that upregulating miR-509-3p markedly inhibited the viability and promoted the apoptosis of CC cells. On the other hand, miR-509-3p was distinctly downregulated in paclitaxel-resistant HCC94 and C-33A cells (vs. normal cells). The transfection of miR-509-3p mimics notably increased their sensitivity to paclitaxel. Meanwhile, RAC1 was found as the potential target of miR-509-3p in bioinformatics analysis. Moreover, the RAC1/p21 (RAC1) activated kinase 1 (PAK1)/LIM kinase 1 (LIMK1)/Cofilin pathway was significantly activated in paclitaxel-resistant HCC94 and C-33A cells, while miR-509-3p overexpression significantly inactivated this pathway. Additionally, downregulation of RAC1 also partly reversed the paclitaxel-resistance of CC cells and inhibited PAK1/LIMK1/Cofilin. All in all, miR-509-3p enhances the apoptosis and chemosensitivity of CC cells by regulating the RAC1/PAK1/LIMK1/Cofilin pathway.
Collapse
Affiliation(s)
- Jia Xu
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University
| | - Xiangdong Ma
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University
| | - Hong Yang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University
| | - Junru Zhang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University
| | - Guoqing Cai
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University
| | - Nianling Yao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University
| |
Collapse
|
12
|
Qiu JJ, Sun SG, Tang XY, Lin YY, Hua KQ. Extracellular vesicular Wnt7b mediates HPV E6-induced cervical cancer angiogenesis by activating the β-catenin signaling pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:260. [PMID: 33234148 PMCID: PMC7687741 DOI: 10.1186/s13046-020-01745-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/20/2020] [Indexed: 02/08/2023]
Abstract
Background The E6 oncoproteins of human papillomavirus (HPV) 16/18 are the critical drivers of cervical cancer (CC) progression. Extracellular vesicles (EVs) are emerging as critical mediators of cancer-tumor microenvironment (TME) communication. However, whether EVs contribute to HPV 16/18 E6-mediated impacts on CC progression remains unclear. Methods A series of in vitro and in vivo assays were performed to elucidate the roles and mechanism of EV-Wnt7b in HPV E6-induced CC angiogenesis. The prognostic value of serum EV-Wnt7b was determined and a predictive nomogram model was established. Results HPV 16/18 E6 upregulated Wnt7b mRNA expression in four HPV 16/18-positive CC cell lines and their EVs. In vitro and in vivo experiments demonstrated that EV-Wnt7b mRNA was transferred to and modulated human umbilical vein endothelial cells (HUVECs) toward more proliferative and proangiogenic behaviors by impacting β-catenin signaling. Clinically, serum EV-Wnt7b levels were elevated in CC patients and significantly correlated with an aggressive phenotype. Serum EV-Wnt7b was determined to be an independent prognostic factor for CC overall survival (OS) and recurrence-free survival (RFS). Notably, we successfully established a novel predictive nomogram model using serum EV-Wnt7b, which showed good prediction of 1- and 3-year OS and RFS. Conclusions Our results illustrate a potential crosstalk between HPV 16/18-positive CC cells and HUVECs via EVs in the TME and highlight the potential of circulating EV-Wnt7b as a novel predictive biomarker for CC prognosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-020-01745-1.
Collapse
Affiliation(s)
- Jun-Jun Qiu
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Shu-Gen Sun
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Xiao-Yan Tang
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China
| | - Ying-Ying Lin
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Ke-Qin Hua
- Department of Gynaecology, Obstetrics and Gynaecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China. .,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, 413 Zhaozhou Road, Shanghai, 200011, China.
| |
Collapse
|
13
|
Jiang X, Wang J, Deng X, Xiong F, Zhang S, Gong Z, Li X, Cao K, Deng H, He Y, Liao Q, Xiang B, Zhou M, Guo C, Zeng Z, Li G, Li X, Xiong W. The role of microenvironment in tumor angiogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:204. [PMID: 32993787 PMCID: PMC7526376 DOI: 10.1186/s13046-020-01709-5] [Citation(s) in RCA: 367] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Tumor angiogenesis is necessary for the continued survival and development of tumor cells, and plays an important role in their growth, invasion, and metastasis. The tumor microenvironment—composed of tumor cells, surrounding cells, and secreted cytokines—provides a conducive environment for the growth and survival of tumors. Different components of the tumor microenvironment can regulate tumor development. In this review, we have discussed the regulatory role of the microenvironment in tumor angiogenesis. High expression of angiogenic factors and inflammatory cytokines in the tumor microenvironment, as well as hypoxia, are presumed to be the reasons for poor therapeutic efficacy of current anti-angiogenic drugs. A combination of anti-angiogenic drugs and antitumor inflammatory drugs or hypoxia inhibitors might improve the therapeutic outcome.
Collapse
Affiliation(s)
- Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Jie Wang
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiangying Deng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Fang Xiong
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanshan Zhang
- Department of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ke Cao
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hao Deng
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yi He
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Ming Zhou
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Can Guo
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China. .,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China.
| |
Collapse
|
14
|
李 娴, 李 姗, 庞 金, 黄 富, 郭 滨, 刘 浩. [Alkaline processing of cantharidin can significanty improve the antitumor activity of cantharidin]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1332-1339. [PMID: 32990225 PMCID: PMC7544588 DOI: 10.12122/j.issn.1673-4254.2020.09.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To assess the changes in the effects of cantharides after alkaline processing on proliferation, migration, invasion, and apoptosis of human lung cancer A549 cells. METHODS Human non-small cell lung cancer A549 cells were treated with cantharis extract (CTE) from raw cantharides and alkali processed cantharis extract (ACE). The proliferation of the cells was detected with CCK-8 assay, and the cell migration and invasion were assessed using wound healing assay and Transwell assay, respectively. The expressions of MMP1 and MMP2 in the cells were detected using Western blotting, the contents of IFN-γ, IL-1β and TNF-α were measured with ELISA, and cell apoptosis was analyzed with annexinV/PI fluorescent staining. RESULTS Both CTE and ACE significantly reduced the viability and inhibited the migration of A549 cells, and high-dose ACE produced a significantly stronger inhibitory effect on cell migration than high- dose CTE (P < 0.01). ACE showed more potent inhibitory effect than CTE on the invasion of A549 cells (P < 0.01). Both CTE and ACE inhibited the expressions of MMP1 and MMP2 and up-regulated the level of IFN-γ without significantly affecting the levels of IL-1β and TNF-α. Annexin V/PI staining showed that both CTE and ACE caused apoptosis of A549 cells, but ACE had a stronger proapoptotic effect. CONCLUSIONS Processing with sodium hydroxide can significantly improve the antitumor activity of cantharides, which inhibits the proliferation, migration and invasion of A549 cells possibly by down-regulating the expressions of MMP1 and MMP2, promoting apoptosis and increasing the level of IFN-γ.
Collapse
Affiliation(s)
- 娴 李
- 蚌埠医学院药学院,安徽 蚌埠 2330302School of Pharmacy, Bengbu Medical College, Bengbu 2330302, China
- 中药饮片制造新技术安徽省重点实验室,安徽 亳州 236800Anhui Provincial Key Laboratory of New Technology of Chinese Herbal Pieces Manufacturing, Bozhou 236800, China
- 安徽协和成药业饮片有限公司博士后工作站,安徽 亳州 236800Postdoctoral Workstation of Anhui Xiehecheng Pharmaceutical Yinpian Co Ltd, Bozhou 236800, China
| | - 姗姗 李
- 蚌埠医学院药学院,安徽 蚌埠 2330302School of Pharmacy, Bengbu Medical College, Bengbu 2330302, China
| | - 金龙 庞
- 蚌埠医学院药学院,安徽 蚌埠 2330302School of Pharmacy, Bengbu Medical College, Bengbu 2330302, China
| | - 富豪 黄
- 蚌埠医学院药学院,安徽 蚌埠 2330302School of Pharmacy, Bengbu Medical College, Bengbu 2330302, China
| | - 滨 郭
- 蚌埠医学院药学院,安徽 蚌埠 2330302School of Pharmacy, Bengbu Medical College, Bengbu 2330302, China
| | - 浩 刘
- 蚌埠医学院药学院,安徽 蚌埠 2330302School of Pharmacy, Bengbu Medical College, Bengbu 2330302, China
| |
Collapse
|
15
|
Yao D, Li C, Rajoka MSR, He Z, Huang J, Wang J, Zhang J. P21-Activated Kinase 1: Emerging biological functions and potential therapeutic targets in Cancer. Am J Cancer Res 2020; 10:9741-9766. [PMID: 32863957 PMCID: PMC7449905 DOI: 10.7150/thno.46913] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
The p21-Activated kinase 1 (PAK1), a member of serine-threonine kinases family, was initially identified as an interactor of the Rho GTPases RAC1 and CDC42, which affect a wide range of processes associated with cell motility, survival, metabolism, cell cycle, proliferation, transformation, stress, inflammation, and gene expression. Recently, the PAK1 has emerged as a potential therapeutic target in cancer due to its role in many oncogenic signaling pathways. Many PAK1 inhibitors have been developed as potential preclinical agents for cancer therapy. Here, we provide an overview of essential roles that PAK1 plays in cancer, including its structure and autoactivation mechanism, its crucial function from onset to progression to metastasis, metabolism, immune escape and even drug resistance in cancer; endogenous regulators; and cancer-related pathways. We also summarize the reported PAK1 small-molecule inhibitors based on their structure types and their potential application in cancer. In addition, we provide overviews on current progress and future challenges of PAK1 in cancer, hoping to provide new ideas for the diagnosis and treatment of cancer.
Collapse
|
16
|
Active RAC1 Promotes Tumorigenic Phenotypes and Therapy Resistance in Solid Tumors. Cancers (Basel) 2020; 12:cancers12061541. [PMID: 32545340 PMCID: PMC7352592 DOI: 10.3390/cancers12061541] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Acting as molecular switches, all three members of the Guanosine triphosphate (GTP)-ase-family, Ras-related C3 botulinum toxin substrate (RAC), Rho, and Cdc42 contribute to various processes of oncogenic transformations in several solid tumors. We have reviewed the distribution of patterns regarding the frequency of Ras-related C3 botulinum toxin substrate 1 (RAC1)-alteration(s) and their modes of actions in various cancers. The RAC1 hyperactivation/copy-number gain is one of the frequently observed features in various solid tumors. We argued that RAC1 plays a critical role in the progression of tumors and the development of resistance to various therapeutic modalities applied in the clinic. With this perspective, here we interrogated multiple functions of RAC1 in solid tumors pertaining to the progression of tumors and the development of resistance with a special emphasis on different tumor cell phenotypes, including the inhibition of apoptosis and increase in the proliferation, epithelial-to-mesenchymal transition (EMT), stemness, pro-angiogenic, and metastatic phenotypes. Our review focuses on the role of RAC1 in adult solid-tumors and summarizes the contextual mechanisms of RAC1 involvement in the development of resistance to cancer therapies.
Collapse
|
17
|
Nck1 promotes the progression of ovarian carcinoma by enhancing the PI3K/AKT/p70S6K signaling. Hum Cell 2020; 33:768-779. [PMID: 32166565 DOI: 10.1007/s13577-020-00344-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/02/2020] [Indexed: 02/05/2023]
Abstract
Non-catalytic region of tyrosine kinase adaptor protein 1 (Nck1) is crucial for the progression of cancers. However, little is known on the role of Nck1 in the progression of ovarian carcinoma (OC). Here, we show that Nck1 expression is up-regulated in 176 OC tissues, compared with non-carcinoma ovarian tissues, and the up-regulated Nck1 expression is associated with the aggressiveness of OC and shorter overall and disease-free survival in this population. Higher Nck1 expression was an independent risk factor for poor prognosis of OC. Furthermore, Nck1 silencing by short hairpin RNA (shRNA) technology significantly inhibited the proliferation, migration and invasion of OC cells in vitro and the growth and metastasis of implanted OC tumors in vivo. Human kinase phosphorylation array indicated that Nck1 silencing significantly reduced the relative levels of 11 kinase expression and phosphorylation in OC cells, particularly for decreased levels of p70S6 kinase (p70S6K) and protein kinase B (AKT) expression in SKOV3 cells. Actually, Nck1 silencing significantly decreased PI3K and AKT expression, and reduced AKT and p70S6K phosphorylation while Nck1 over-expression had opposite effects in OC cells. Therefore, our data indicate that Nck1 promotes the progression of OC by enhancing the PI3k/AKT/p70S6K signaling in OC. Our findings suggest that Nck1 expression may be valuable for evaluating the prognosis of OC and as a target for design of new therapies for OC.
Collapse
|
18
|
Sundaram MK, Unni S, Somvanshi P, Bhardwaj T, Mandal RK, Hussain A, Haque S. Genistein Modulates Signaling Pathways and Targets Several Epigenetic Markers in HeLa Cells. Genes (Basel) 2019; 10:E955. [PMID: 31766427 PMCID: PMC6947182 DOI: 10.3390/genes10120955] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Several epigenetic changes are responsible for transcriptional alterations of signaling pathways and tumour suppressor genes (TSGs) contributing to carcinogenesis. This study was aimed to examine the effect of the phytochemical, genistein on various molecular targets in HeLa cells. METHODS Quantitative PCR was used to analyze the expression of various molecular targets. Biochemical assays were employed to study the epigenetic enzymes. To correlate the transcriptional status of the selected TSGs and epigenetic modulation, their promoter 5'CpG methylation levels were evaluated by quantitative methylation array followed by methylation specific restriction digestion. RESULTS The expression of several genes involved in the cell cycle regulation, migration, inflammation, phosphatidylinositol 3-kinase (PI3K) and mitogen activated kinase-like protein (MAPK) pathway were found to be modulated including CCNB1, TWIST1, MMP14, TERT, AKT1, PTPRR, FOS and IL1A. Genistein modulated the expression of DNA methyltransferases (DNMTs), histone deacetylases (HDACs), histone methyltransferases (HMTs), demethylases, and histone phosphorylases. Furthermore, genistein decreased the activity of DNMTs, HDACs, and HMTs and reduced global DNA methylation levels. Promoter methylation of several TSGs, including FHIT, RUNX3, CDH1, PTEN, and SOC51, was lowered with corresponding transcriptional increase. Network analysis indicated similar effect of genistein. CONCLUSION This study presents a comprehensive mechanism of action of genistein showcasing effective epigenetic modulation and widespread transcriptional changes resulting in restoration of tumour suppressor gene expression. This study corroborates the development of genistein as a candidate for anti-cancer therapy.
Collapse
Affiliation(s)
| | - Sreepoorna Unni
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, P.O. Box 19282 Dubai, UAE;
| | - Pallavi Somvanshi
- Department of Biotechnology, TERI School of Advanced Studies, 10, Institutional Area, Vasant Kunj, New Delhi-110070, India; (P.S.); (T.B.)
| | - Tulika Bhardwaj
- Department of Biotechnology, TERI School of Advanced Studies, 10, Institutional Area, Vasant Kunj, New Delhi-110070, India; (P.S.); (T.B.)
| | - Raju K. Mandal
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia;
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, P.O. Box 345050 Dubai, UAE;
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan-45142, Saudi Arabia;
| |
Collapse
|
19
|
Guan Z, Song Y, Ma J, Li F, Zhao X, Liang G, An H, Pu J. Altered expression of lncRNA NCK1-AS1 distinguished patients with prostate cancer from those with benign prostatic hyperplasia. Oncol Lett 2019; 18:6379-6384. [PMID: 31807161 PMCID: PMC6876338 DOI: 10.3892/ol.2019.11039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022] Open
Abstract
Long non-coding (lnc)RNA NCK1 antisense RNA 1 (NCK1-AS1) has been characterized as an oncogene in cervical cancer, while its role in prostate cancer (PC) remains unknown. It was revealed in the present study that plasma NCK1-AS1 was upregulated in patients with PC when compared with patients with benign prostatic hyperplasia (BPH) and healthy controls. Upregulation of NCK1-AS1 distinguished patients with PC from patients with BPH and healthy controls. Overexpression of NCK1-AS1 led to significantly upregulated transforming growth factor (TGF)-β1, while TGF-β1 overexpression failed to significantly affect NCK1-AS1 in PC cells. NCK1-AS1 overexpression led to promoted migration and invasion. TGF-β inhibitor played an opposite role and attenuated the effects of NCK1-AS1 overexpression. Therefore, NCK1-AS1 may upregulate TGF-β1 to promote PC.
Collapse
Affiliation(s)
- Zhihui Guan
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Department of Urology, Hulunbuir People's Hospital, Hulunbuir, Inner Mongolia 021008, P.R. China
| | - Yuwei Song
- Department of Pediatrics, Hulunbuir People's Hospital, Hulunbuir, Inner Mongolia 021008, P.R. China
| | - Jinguo Ma
- Department of Oncology, Hulunbuir People's Hospital, Hulunbuir, Inner Mongolia 021008, P.R. China
| | - Feng Li
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiaojun Zhao
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Guodong Liang
- Department of Pathology, Hulunbuir People's Hospital, Hulunbuir, Inner Mongolia 021008, P.R. China
| | - Haiquan An
- Department of Urology, Hulunbuir People's Hospital, Hulunbuir, Inner Mongolia 021008, P.R. China
| | - Jinxian Pu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
20
|
Ji Z, Pan X, Shang Y, Ni DT, Wu FL. KIF18B as a regulator in microtubule movement accelerates tumor progression and triggers poor outcome in lung adenocarcinoma. Tissue Cell 2019; 61:44-50. [PMID: 31759406 DOI: 10.1016/j.tice.2019.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/01/2019] [Accepted: 09/01/2019] [Indexed: 12/24/2022]
Abstract
KIF18B is involved in several tumor progression and exerts critical effects on microtubule growth during mitosis, but its role in lung adenocarcinoma still remains rare. Hence, we attempted to explore the biological function of KIF18B in lung adenocarcinoma. We first analyzed the expressional pattern of KIF18B in lung adenocarcinoma, and detected the correlation between KIF18B expression and clinical characteristics in lung adenocarcinoma based on The Cancer Genome Atlas (TCGA) database and Oncomine dataset. Subsequently, cell counting kit-8 (CCK-8) assay, wound-healing analysis, and transwell method were performed to assess the effects of KIF18B in lung adenocarcinoma cells. Quantitative real-time reverse transcription-PCR (qRT-PCR) and western blotting were utilized to measure the mRNA and protein expression levels. Our results illustrated that KIF18B expression was significantly up-regulated in lung adenocarcinoma samples compared to normal specimens. High levels of KIF18B were associated with unfavorable prognosis of lung adenocarcinoma patients. Down-regulation of KIF18B in lung adenocarcinoma cells inhibited cell prolifartion, migration, and invasion. Western blot assay demonstrated that KIF18B knockdown markedly decreased Rac1-GTP expression, an important marker of migration and invasion in tumors. Moreover, the phosphorylation of AKT and mTOR expression levels were attenuated after KIF18B knockdown. Taken together, these data enhanced the point that KIF18B might promote lung adenocarcinoma cell proliferation, migration, and invasion by activating Rac1 and mediating the AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Ze Ji
- Department of Respiratory Medicine, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, 215028, PR China
| | - Xing Pan
- Department of Nursing, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, 215028, PR China
| | - Yan Shang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Second Military Medical University, Shanghai, 200433, PR China
| | - Dian-Tao Ni
- Department of Respiratory Medicine, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, Jiangsu, 215028, PR China.
| | - Feng-Lei Wu
- Department of Oncology, First People Hospital of Lianyungang, Lianyungang, Jiangsu, 222002, PR China.
| |
Collapse
|
21
|
Yin J, Wu K, Ma Q, Dong H, Zhu Y, Hu L, Kong X. Revisiting Non-BRCA1/2 Familial Whole Exome Sequencing Datasets Implicates NCK1 as a Cancer Gene. Front Genet 2019; 10:527. [PMID: 31214250 PMCID: PMC6557995 DOI: 10.3389/fgene.2019.00527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Through linkage and candidate gene screening, many breast cancer (BC) predisposition genes have been identified in the past 20 years. However, the majority of genetic risks that contribute to familial BC remains undetermined. In this study, we revisited whole exome sequencing datasets from non-BRCA1/2 familial BC patients, to search for novel BC predisposition genes. Based on the infinite mutation model, we supposed that rare non-silent variants that cooccurred between familial and TCGA-germline datasets, might play a predisposition contributing role. In our analysis, we not only identified novel potential pathogenic variants from known cancer predisposition genes, such as MRE11, CTR9 but also identified novel candidate predisposition genes, such as NCK1. According to the TCGA mRNA expression dataset of BC, NCK1 was significantly upregulated in basal-like subtypes and downregulated in luminal subtypes. In vitro, NCK1 mutants (D73H and R42Q) transfected MCF7 cell lines, which attributed to the luminal subtype, were much more viable and invasive than the wild type. On the other side, our results also showed that overall survival and disease-free survival of patients with NCK1 variations might be dependent on the genomic context. In conclusion, genetic heterogeneity exists among non-BRCA1/2 BC pedigrees and NCK1 could be a novel BC predisposition gene.
Collapse
Affiliation(s)
- Jie Yin
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kai Wu
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qingyang Ma
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hang Dong
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yufei Zhu
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Landian Hu
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiangyin Kong
- State Key Laboratory of Medical Genomics, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|