1
|
Sumorek-Wiadro J, Kapral-Piotrowska J, Zając A, Maciejczyk A, Hułas-Stasiak M, Skalicka-Woźniak K, Rzeski W, Pawlikowska-Pawlęga B, Jakubowicz-Gil J. Proapoptotic and antimigration properties of osthole in combination with LY294002 against human glioma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3147-3161. [PMID: 39352533 PMCID: PMC11919984 DOI: 10.1007/s00210-024-03424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 08/30/2024] [Indexed: 03/19/2025]
Abstract
Anaplastic astrocytoma and glioblastoma multiforme are infiltrating and vascularized gliomas with a high degree of chemoresistance and metastasis. Our previous studies have shown that osthole may be of great importance in the treatment of gliomas. Therefore, in this work, for the first time, coumarin was used in combination with LY294002-an inhibitor of the PI3K-Akt/PKB-mTOR pathway, which is overly active in gliomas. MOGGCCM and T98G cells were incubated with osthole and LY294002, alone and in combination. Staining with specific fluorochromes was used to visualize cell death and the scratch test to assess the migration. The level of proteins was estimated by immunoblotting. Forming protrusions were visualized by SEM, and immunocytochemistry was used to determine the localization of proteins. Additionally, the expression of Bcl-2, beclin 1 and Raf kinase was silenced using specific siRNA. The obtained results showed that osthole in combination with LY294092 effectively inhibited the migration of glioma cells by reducing the level of metaloproteinases and Rho family proteins, as well as decreasing the level of N-cadherin. In addition, the combination of compounds induced apoptosis. New combination of compounds shows a high pro-apoptotic potential and also inhibits the migration of gliomas cells.
Collapse
Affiliation(s)
- Joanna Sumorek-Wiadro
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Justyna Kapral-Piotrowska
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Adrian Zając
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Aleksandra Maciejczyk
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Monika Hułas-Stasiak
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Krystyna Skalicka-Woźniak
- Independent Laboratory of Natural Products, Medical University of Lublin, Chodzki 1, 20-093, Lublin, Poland
| | - Wojciech Rzeski
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
- Department of Medical Biology, Institute of Rural Health, Institute of Agricultural Medicine, Jaczewskiego 2, 20-950, Lublin, Poland
| | - Bożena Pawlikowska-Pawlęga
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
2
|
Santa S, Kwofie SK, Agyenkwa-Mawuli K, Quaye O, Brown CA, Tagoe EA. Prediction of Human Papillomavirus-Host Oncoprotein Interactions Using Deep Learning. Bioinform Biol Insights 2024; 18:11779322241304666. [PMID: 39664297 PMCID: PMC11632871 DOI: 10.1177/11779322241304666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 11/16/2024] [Indexed: 12/13/2024] Open
Abstract
Background Human papillomavirus (HPV) causes disease through complex interactions between viral and host proteins, with the PI3K signaling pathway playing a key role. Proteins like AKT, IQGAP1, and MMP16 are involved in HPV-related cancer development. Traditional methods for studying protein-protein interactions (PPIs) are labor-intensive and time-consuming. Computational models are becoming more popular as they are less labor-intensive and often more efficient. This study aimed to develop a deep learning model to predict interactions between HPV and host proteins. Method To achieve this, available HPV and host protein interaction data was retrieved from the protocol of Eckhardt et al and used to train a Recurrent Neural Network algorithm. Training of the model was performed on the SPYDER (scientific python development environment) platform using python libraries; Scikit-learn, Pandas, NumPy, and TensorFlow. The data was split into training, validation, and testing sets in the ratio 7:1:2, respectively. After the training and validation, the model was then used to predict the possible interactions between HPV 31 and 18 E6 and E7, and host oncoproteins AKT, IQGAP1 and MMP16. Results The model showed good performance, with an MCC score of 0.7937 and all other metrics above 88%. The model predicted an interaction between E6 and E7 of both HPV types with AKT, while only HPV31 E7 was shown to interact with IQGAP1 and MMP16 with confidence scores of 0.9638 and 0.5793, respectively. Conclusion The current model strongly predicted HPVs E6 and E7 interactions with PI3K pathway, and the viral proteins may be involved in AKT activation, driving HPV-associated cancers. This model supports the robust prediction of interactomes for experimental validation.
Collapse
Affiliation(s)
- Sheila Santa
- Department of Biochemistry, Cell & Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Samuel Kojo Kwofie
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kwasi Agyenkwa-Mawuli
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Osbourne Quaye
- Department of Biochemistry, Cell & Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Charles A Brown
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Emmanuel A Tagoe
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
3
|
Xu H, Cao Y, Ruan J, Wang F, He Y, Yang L, Yu T, Du F, Zhang N, Cao X. The effects of BMP2 and the mechanisms involved in the invasion and angiogenesis of IDH1 mutant glioma cells. J Neurooncol 2024; 170:161-171. [PMID: 39117967 PMCID: PMC11447149 DOI: 10.1007/s11060-024-04789-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE This study investigated the effect of an isocitrate dehydrogenase 1 (IDH1) mutation (mutIDH1) on the invasion and angiogenesis of human glioma cells. METHODS Doxycycline was used to induce the expression of mutIDH1 in glioma cells. Transwell and wound healing assays were conducted to assess glioma cell migration and invasion. Western blotting and cell immunofluorescence were used to measure the expression levels of various proteins. The influence of bone morphogenetic protein 2 (BMP2) on invasion, angiogenesis-related factors, BMP2-related receptor expression, and changes in Smad signaling pathway-related proteins were evaluated after treatment with BMP2. Differential gene expression and reference transcription analysis were performed. RESULTS Successful infection with recombinant lentivirus expressing mutIDH1 was demonstrated. The IDH1 mutation promoted glioma cell migration and invasion while positively regulating the expression of vascularization-related factors and BMP2-related receptors. BMP2 exhibited a positive regulatory effect on the migration, invasion, and angiogenesis of mutIDH1-glioma cells, possibly mediated by BMP2-induced alterations in Smad signaling pathway-related factors.After BMP2 treatment, the differential genes of MutIDH1-glioma cells are closely related to the regulation of cell migration and cell adhesion, especially the regulation of Smad-related proteins. KEGG analysis confirmed that it was related to BMP signaling pathway and TGF-β signaling pathway and cell adhesion. Enrichment analysis of gene ontology and genome encyclopedia further confirmed the correlation of these pathways. CONCLUSION Mutation of isocitrate dehydrogenase 1 promotes the migration, invasion, and angiogenesis of glioma cells, through its effects on the BMP2-driven Smad signaling pathway. In addition, BMP2 altered the transcriptional patterns of mutIDH1 glioma cells, enriching different gene loci in pathways associated with invasion, migration, and angiogenesis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China
- Department of Pathology, The First People's Hospital of Yinchuan, Yinchuan, 750001, Ningxia, China
| | - Yu Cao
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China
| | - Jianqiao Ruan
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China
| | - Fei Wang
- Department of Pathology, The First People's Hospital of Yinchuan, Yinchuan, 750001, Ningxia, China
| | - Yuhong He
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China
| | - Lina Yang
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China
| | - Tian Yu
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China
| | - Fang Du
- School of Information Engineering, Ningxia University, Yinchuan, 750021, Ningxia, China
| | - Ningmei Zhang
- Department of Pathology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Yinchuan, 750004, Ningxia Hui Autonomous Region, P.R. China.
| | - Xiangmei Cao
- Department of Pathology, Basic Medical School of Ningxia Medical University, 1160 Shengli South Street, Yinchuan, 750004, Ningxia, Hui Autonomous Region, P.R. China.
| |
Collapse
|
4
|
Sheila S, Adoquaye BC, Kafui AP, Lawrence E, Richard HA, Osbourne Q, Ayitey TE. Differential expression of host oncogenes in human papillomavirus-associated nasopharyngeal and cervical epithelial cancers. Kaohsiung J Med Sci 2024; 40:830-836. [PMID: 39073693 PMCID: PMC11895668 DOI: 10.1002/kjm2.12880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Human papillomavirus (HPV)-related cervical and nasopharyngeal cancers differ in molecular mechanisms underlying the oncogenic processes. The disparity may be attributed to differential expression of oncoproteins. The current study investigated the host oncogenes expression pattern in HPV-associated cervical and nasopharyngeal cancer. Formalin-fixed paraffin-embedded tissues originating from the nasopharyngeal and cervical regions were screened using Hematoxylin and Eosin staining. Genomic DNA and total RNA were extracted from confirmed cancer biopsies and non-cancer tissues (NC). HPV was detected by PCR using MY09/GP5+/6+ primers. Protein expression levels of AKT, IQGAP1, and MMP16 in HPV-infected cancers and controls were determined by immunohistochemistry. RT-qPCR was used to profile mRNAs of the oncogenes. AKT and IQGAP1 proteins were highly expressed in the epithelial cancers compared with the non-cancer tissues (p < 0.05). IQGAP1 and MMP16 mRNAs level was significantly higher in the cancers than in the NC (p < 0.05), but not AKT mRNA levels. MMP16 protein was ubiquitously expressed in all tissues. AKT mRNA level was significantly elevated in CC compared with NPC (p < 0.001). However, the difference in AKT, IQGAP1 and MMP16 proteins level between CC and NPC was not significant (p > 0.05). The oncoproteins expression level between the HPV-positive and HPV-negative cancer biopsies showed no significant difference (p < 0.05). Current study reports AKT but not IQGAP1 and MMP16 mRNAs differentially expression in cervical and nasopharyngeal cancers, independent of HPV infection status.
Collapse
Affiliation(s)
- Santa Sheila
- Department of Biochemistry, Cell & Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP)University of GhanaAccraGhana
- Department of Medical Laboratory SciencesUniversity of GhanaAccraGhana
| | | | - Akakpo Patrick Kafui
- Department of Pathology, School of Medical SciencesUniversity of Cape CoastCape CoastGhana
- Pathologists Without BordersAccraGhana
| | | | - Hooper Andrew Richard
- Department of PathologyUniversity of Ghana Medical School, University of GhanaAccraGhana
- Korle Bu Teaching HospitalAccraGhana
| | - Quaye Osbourne
- Department of Biochemistry, Cell & Molecular Biology/West African Centre for Cell Biology of Infectious Pathogens (WACCBIP)University of GhanaAccraGhana
| | | |
Collapse
|
5
|
Hu L, Liu H, Ma H, Zeng X, Cao Y, Liu B, Li H, Zhang X. TRAF6-mediated ubiquitination of AKT1 in the nucleus occurs in a β-arrestin2-dependent manner upon insulin stimulation. Biochem Pharmacol 2024; 226:116362. [PMID: 38871335 DOI: 10.1016/j.bcp.2024.116362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
AKT, also known as protein kinase B (PKB), serves as a crucial regulator of numerous biological functions, including cell growth, metabolism, and tumorigenesis. Increasing evidence suggests that the kinase activity of AKT is regulated via ubiquitination by various E3 ligase enzymes in response to different stimuli. However, the molecular mechanisms underlying insulin-induced AKT ubiquitination are not yet fully understood. Here, we show that activation of the insulin receptor (IR) leads to enhanced ubiquitination of AKT1 at K8 and K14 residues, facilitated by the cytosolic E3 ubiquitin ligase enzyme, TRAF6. Further investigation using AKT1 mutants with modified nucleocytoplasmic shuttling properties reveals that TRAF6-mediated AKT1 ubiquitination occurs within the nucleus in a β-Arr2-dependent manner. The nuclear entry of TRAF6 depends on importin β1, while β-Arr2 regulates this process by facilitating the interaction between TRAF6 and importin β1. Additionally, the ubiquitination of AKT1 is essential for its translocation to the activated IR on the plasma membrane, where it plays a functional role in recruiting Glut4 and facilitating glucose uptake. This study uncovers the cellular components and processes involved in insulin-induced ubiquitination and activation of AKT1, providing insights and detailed strategies for manipulating AKT1.
Collapse
Affiliation(s)
- Li Hu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Haiping Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Haixiang Ma
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Xingyue Zeng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Yongkai Cao
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Bing Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Huijun Li
- Department of Pharmaceuticals, People's Hospital of Zunyi City Bo Zhou District, Zunyi 563000, China
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
6
|
Salmaninejad A, Layeghi SM, Falakian Z, Golestani S, Kobravi S, Talebi S, Yousefi M. An update to experimental and clinical aspects of tumor-associated macrophages in cancer development: hopes and pitfalls. Clin Exp Med 2024; 24:156. [PMID: 39003350 PMCID: PMC11246281 DOI: 10.1007/s10238-024-01417-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant tumor-infiltrating stromal cells, and their normal function in tumor microenvironment (TME) is to suppress tumor cells by producing cytokines which trigger both direct cell cytotoxicity and antibody-mediated immune response. However, upon prolonged exposure to TME, the classical function of these so-called M1-type TAMs can be converted to another type, "M2-type," which are recruited by tumor cells so that they promote tumor growth and metastasis. This is the reason why the accumulation of TAMs in TME is correlated with poor prognosis in cancer patients. Both M1- and M2-types have high degree of plasticity, and M2-type cells can be reprogrammed to M1-type for therapeutic purposes. This characteristic introduces TAMs as promising target for developing novel cancer treatments. In addition, inhibition of M2-type cells and blocking their recruitment in TME, as well as their depletion by inducing apoptosis, are other approaches for effective immunotherapy of cancer. In this review, we summarize the potential of TAMs to be targeted for cancer immunotherapy and provide an up-to-date about novel strategies for targeting TAMs.
Collapse
Affiliation(s)
- Arash Salmaninejad
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Pediatric Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sepideh Mehrpour Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Falakian
- Department of Laboratory Science, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Shahin Golestani
- Department of Ophthalmology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Tehran Azad University, Tehran, Iran
| | - Samaneh Talebi
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Yousefi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
7
|
Chang R, Su Y, Kong H, Wang F, Xing Y, Jiang L, Xin H. Upregulation of SEMP1 Contributes to Improving the Biological Functions of Trophoblast via the PI3K/AKT Pathway in Preeclampsia. Mol Biotechnol 2024; 66:531-543. [PMID: 37277581 DOI: 10.1007/s12033-023-00774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023]
Abstract
Disturbance of extravillous trophoblast infiltration is associated with preeclampsia (PE), a severe condition of pregnancy characterized by hypertension and proteinuria. Senescence-associated epithelial membrane protein 1 (SEMP1), an integral membrane protein, is a vital component of tight junction strands in epithelial or endothelial cells, with no clear function reported in PE. Gene Expression Omnibus (GEO) datasets showed that SEMP1 expression was downregulated in the placental tissues of PE patients, which was confirmed by assessing SEMP1 levels in placental samples collected in our hospital. Furthermore, less SEMP1 was detected in cytokeratin 7 positive trophoblast cells in the spiral arteries of rat placentas post L-arginine methyl ester hydrochloride (L-NAME) treatment. Trophoblast cells acquired robust ability of proliferation, migration, and invasion when SEMP1 was overexpressed. Such capability was weakened in SEMP1-silenced cells. Trophoblast cells overexpressing SEMP1 secreted more vascular endothelial growth factor A (VEGFA), which facilitated the tube formation of human umbilical vein endothelial cells. Blockade of PI3K/AKT signaling transduction with LY294002 dampened the effects of SEMP1 on trophoblast cells. Collectively, we firstly indicated that SEMP1 inhibition is a potential driver for PE, which may be associated with the deactivation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Ruijing Chang
- Department of Obstetrics, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Yuan Su
- Department of Obstetrics, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Hongfang Kong
- Department of Obstetrics, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Fang Wang
- Department of Obstetrics, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Ying Xing
- Department of Obstetrics, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Lei Jiang
- Department of Obstetrics, Shijiazhuang Maternity & Child Healthcare Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Hong Xin
- Department of Obstetrics, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
8
|
Han I, Song IS, Choi SA, Lee T, Yusupov M, Shaw P, Bogaerts A, Choi EH, Ryu JJ. Bioactive Nonthermal Biocompatible Plasma Enhances Migration on Human Gingival Fibroblasts. Adv Healthc Mater 2023; 12:e2200527. [PMID: 36373222 DOI: 10.1002/adhm.202200527] [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: 03/07/2022] [Revised: 10/18/2022] [Indexed: 11/16/2022]
Abstract
This study hypothesizes that the application of low-dose nonthermal biocompatible dielectric barrier discharge plasma (DBD-NBP) to human gingival fibroblasts (HGFs) will inhibit colony formation but not cell death and induce matrix metalloproteinase (MMP) expression, extracellular matrix (ECM) degradation, and subsequent cell migration, which can result in enhanced wound healing. HGFs treated with plasma for 3 min migrate to each other across the gap faster than those in the control and 5-min treatment groups on days 1 and 3. The plasma-treated HGFs show significantly high expression levels of the cell cycle arrest-related p21 gene and enhanced MMP activity. Focal adhesion kinase (FAK) mediated attenuation of wound healing or actin cytoskeleton rearrangement, and plasma-mediated reversal of this attenuation support the migratory effect of DBD-NBP. Further, this work performs computer simulations to investigate the effect of oxidation on the stability and conformation of the catalytic kinase domain (KD) of FAK. It is found that the oxidation of highly reactive amino acids (AAs) Cys427, Met442, Cys559, Met571, Met617, and Met643 changes the conformation and increases the structural flexibility of the FAK protein and thus modulates its function and activity. Low-dose DBD-NBP-induces host cell cycle arrest, ECM breakdown, and subsequent migration, thus contributing to the enhanced wound healing process.
Collapse
Affiliation(s)
- Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Kwangwoon University, Seoul, 01897, Republic of Korea.,Department of Plasma Bio-Display, Kwangwoon University, Seoul, 01897, Korea
| | - In-Seok Song
- Department of Dentistry, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul, 03080, Republic of Korea
| | - Taebok Lee
- Confocal Core Facility, Center for Medical Innovation, Seoul National University Hospital, Seoul, 03082, Korea
| | - Maksudbek Yusupov
- Research group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Priyanka Shaw
- Research group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Annemie Bogaerts
- Research group PLASMANT, Department of Chemistry, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Kwangwoon University, Seoul, 01897, Republic of Korea
| | - Jae Jun Ryu
- Department of Dentistry, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| |
Collapse
|
9
|
RAB11A Promotes Cell Malignant Progression and Tumor Formation of Prostate Cancer via Activating FAK/AKT Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:5885387. [PMID: 36760469 PMCID: PMC9904921 DOI: 10.1155/2023/5885387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/25/2022] [Accepted: 11/24/2022] [Indexed: 02/04/2023]
Abstract
Background RAB11A, a member of the GTPase family, acts as a regulator in diverse cancers development. The dysregulation of the FAK/AKT signaling pathway is mainly related to tumorigenesis. This study aimed to investigate the possible effect of RAB11A in prostate cancer and further explore the potential mechanisms. Results In this study, we illustrated the tumor-promoting effects of RAB11A based on in vivo and in vitro experiments. RAB11A expression was upregulated in prostate cancer cells. RAB11A knockdown decreased the prostate cancer cell proliferation, migration, and invasion. RAB11A also induced the epithelial-mesenchymal transition. PF562271 suppressed the malignant characteristics of prostate cancer cells caused by RAB11A knockdown. Furthermore, the interference of RAB11A reduced the tumor growth and the protein levels of p-FAK/FAK and p-AKT/AKT in vivo. Conclusion RAB11A promotes cell malignant progression and tumor formation in prostate cancer via activating FAK/AKT signaling pathway.
Collapse
|
10
|
Zangouei AS, Zangoue M, Taghehchian N, Zangooie A, Rahimi HR, Saburi E, Alavi MS, Moghbeli M. Cell cycle related long non-coding RNAs as the critical regulators of breast cancer progression and metastasis. Biol Res 2023; 56:1. [PMID: 36597150 PMCID: PMC9808980 DOI: 10.1186/s40659-022-00411-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Cell cycle is one of the main cellular mechanisms involved in tumor progression. Almost all of the active molecular pathways in tumor cells directly or indirectly target the cell cycle progression. Therefore, it is necessary to assess the molecular mechanisms involved in cell cycle regulation in tumor cells. Since, early diagnosis has pivotal role in better cancer management and treatment, it is required to introduce the non-invasive diagnostic markers. Long non-coding RNAs (LncRNAs) have higher stability in body fluids in comparison with mRNAs. Therefore, they can be used as efficient non-invasive markers for the early detection of breast cancer (BCa). In the present review we have summarized all of the reported lncRNAs involved in cell cycle regulation in BCa. It has been reported that lncRNAs mainly affect the cell cycle in G1/S transition through the CCND1/CDK4-6 complex. Present review paves the way of introducing the cell cycle related lncRNAs as efficient markers for the early detection of BCa.
Collapse
Affiliation(s)
- Amir Sadra Zangouei
- grid.411583.a0000 0001 2198 6209Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Zangoue
- grid.411701.20000 0004 0417 4622Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran ,grid.411701.20000 0004 0417 4622Department of Anesthesiology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Negin Taghehchian
- grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Zangooie
- grid.411701.20000 0004 0417 4622Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran ,grid.411701.20000 0004 0417 4622Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Hamid Reza Rahimi
- grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahya Sadat Alavi
- grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- grid.411583.a0000 0001 2198 6209Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Lin L, Gao Y, Hu X, Ouyang J, Liu C. Pentamidine inhibits proliferation, migration and invasion in endometrial cancer via the PI3K/AKT signaling pathway. BMC Womens Health 2022; 22:470. [PMID: 36434592 PMCID: PMC9700983 DOI: 10.1186/s12905-022-02078-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/16/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Pentamidine has been reported to have many pharmacological effects including anti- protozoal, anti-inflammatory, and anti-tumor activities. The aim of this study is to investigate the potential therapeutic role of Pentamidine and molecular mechanisms of Pentamidine on PI3K/AKT signaling pathway underlying the anti-tumor properties in endometrial cancer. METHODS Our study was carried out in the central laboratory of Harbin Medical University from 2019 to 2021. Human endometrial cancer cell lines Ishikawa and HEC-1A were treated with Pentamidine. The proliferation ability of cells was investigated by MTS and colony formation assays. The cell cycle distribution was detected by flow cytometry. Cell migration and invasion were analyzed by using the wound healing assay and Transwell assay. Western blotting was performed to measure the levels of AKT, p-AKT, MMP-2, and MMP-9. RESULTS Our results revealed that treatment of Pentamidine inhibited proliferation, migration and invasion of Ishikawa and HEC-1A endometrial cancer cells. Mechanistic investigation showed that Pentamidine inhibited PI3K/AKT signaling pathway and also reduced the expression of MMP-2 and MMP-9. In addition, co-treatment with PI3K kinase inhibitor LY294002 and Pentamidine leaded to increased repression of cell viability and the protein expression of p-AKT in Ishikawa cells. CONCLUSIONS Pentamidine suppresses PI3K/AKT signaling pathway, and inhibits proliferation, migration and invasion of EC cells. These findings suggested that Pentamidine might be a potential candidate for treating EC through PI3K/AKT pathway.
Collapse
Affiliation(s)
- Lin Lin
- grid.412596.d0000 0004 1797 9737Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin City, 150001 Heilongjiang Province People’s Republic of China
| | - Yunan Gao
- grid.411491.8Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin City, 150001 Heilongjiang Province People’s Republic of China
| | - Xiaochen Hu
- grid.412596.d0000 0004 1797 9737Department of Respiratory Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin city, 150001 Heilongjiang Province People’s Republic of China
| | - Jiabao Ouyang
- grid.412596.d0000 0004 1797 9737Ultrasound Department, The First Affiliated Hospital of Harbin Medical University, Harbin city, 150001 Heilongjiang Province People’s Republic of China
| | - Chunbo Liu
- grid.412596.d0000 0004 1797 9737Ultrasound Department, The First Affiliated Hospital of Harbin Medical University, Harbin city, 150001 Heilongjiang Province People’s Republic of China
| |
Collapse
|
12
|
Tungsukruthai S, Sritularak B, Chanvorachote P. Cycloartocarpin Inhibits Migration through the Suppression of Epithelial-to-Mesenchymal Transition and FAK/AKT Signaling in Non-Small-Cell Lung Cancer Cells. Molecules 2022; 27:8121. [PMID: 36500213 PMCID: PMC9737129 DOI: 10.3390/molecules27238121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Lung cancer metastasis is a multifaceted process that accounts for 90% of cancer deaths. According to several studies, the epithelial-mesenchymal transition (EMT) plays an essential role in lung cancer metastasis. Therefore, this study aimed to investigate the potential pharmacological effect of cycloartocarpin on the suppression of metastasis-related behaviors and EMT. An MTT assay was used to examine cell viability. Cell migration was determined using a wound healing assay. Anchorage-independent cell growth was also performed. Western blot analysis was used to identify the key signaling proteins involved in the regulation of EMT and migration. The results found that non-toxic concentrations of cycloartocarpin (10-20 μM) effectively suppressed cell migration and attenuated anchorage-independent growth in H292, A549, and H460 cells. Interestingly, these effects were consistent with the findings of Western blot analysis, which revealed that the level of phosphorylated focal adhesion kinase (p-FAK), phosphorylated ATP-dependent tyrosine kinase (p-AKT), and cell division cycle 42 (Cdc42) were significantly reduced, resulting in the inhibition of the EMT process, as evidenced by decreased N-cadherin, vimentin, and slug expression. Taken together, the results suggest that cycloartocarpin inhibits EMT by suppressing the FAK/AKT signaling pathway, which is involved in Cdc42 attenuation. Our findings demonstrated that cycloartocarpin has antimetastatic potential for further research and development in lung cancer therapy.
Collapse
Affiliation(s)
- Sucharat Tungsukruthai
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai 90110, Thailand
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pithi Chanvorachote
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
13
|
Jin Z, Tao S, Zhang C, Xu D, Zhu Z. KIF20A promotes the development of fibrosarcoma via PI3K-Akt signaling pathway. Exp Cell Res 2022; 420:113322. [PMID: 36037925 DOI: 10.1016/j.yexcr.2022.113322] [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: 02/28/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 11/04/2022]
Abstract
Adult fibrosarcoma is an aggressive subtype of soft tissue sarcoma (STS), in which high expression of KIF20A indicates a poor prognosis. However, the precise role of KIF20A in fibrosarcoma progression remains unknown. In this study, we initially examined KIF20A expression and function in the human fibrosarcoma cell line HT-1080. The results showed that KIF20A was highly expressed in HT-1080, knockdown of KIF20A impaired cell proliferation, migration, invasion and induced G2/M arrest and cell apoptosis. Transcriptome study suggested that PI3K-Akt signal pathway was involved in these biological changes. We confirmed that PI3K-Akt and NF-κB signaling pathways were impaired after the down-regulation of KIF20A, which can be reversed by the Akt activator SC79 in HT-1080 in vitro. In a xenograft mouse model, knockdown of KIF20A inhibited tumor growth, Ki67 expression and liver metastasis. Taken together, our results suggested that KIF20A promoted fibrosarcoma progression via PI3K-Akt signaling pathway and might be a potential therapeutic target for fibrosarcoma.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University. Shenzhen, Guangdong Province, China
| | - Shuang Tao
- Department of Otorhinolaryngology Head and Neck Surgery, Longgang Central Hospital of Shenzhen, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Chao Zhang
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Damo Xu
- Department of Respirology & Allergy, The Third Affiliated Hospital of Shenzhen University. Shenzhen, Guangdong Province, China; State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong Province, China.
| | - Zhenhua Zhu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
14
|
Ng EFY, Kaida A, Nojima H, Miura M. Roles of IGFBP-3 in cell migration and growth in an endophytic tongue squamous cell carcinoma cell line. Sci Rep 2022; 12:11503. [PMID: 35798794 PMCID: PMC9262895 DOI: 10.1038/s41598-022-15737-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 06/28/2022] [Indexed: 11/20/2022] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP-3) is a member of the IGFBP family that has high affinity for IGFs and functions as either an oncogene or tumor suppressor in various types of cancer. We previously found that IGFBP3 mRNA levels are higher in endophytic-type human tongue squamous cell carcinoma (TSCC) that is more invasive and more prone to metastasis than exophytic and superficial types. This finding prompted us to investigate the roles of IGFBP-3 in TSCC using SAS cells, which were originally derived from endophytic-type TSCC. Specifically, we used SAS cells that express a fluorescent ubiquitination-based cell-cycle indicator (Fucci). RNA-sequencing analysis indicated that IGFBP-3 is associated with cell migration and cell growth. In fact, IGFBP-3 knockdown downregulates cell migration and causes cells to arrest in G1. This migratory potential appears to be cell cycle–independent. IGFBP-3 knockdown also reduced levels of secreted IGFBP-3; however, decreased migratory potential was not rescued by exogenous recombinant human IGFBP-3. Furthermore, ERK activity was downregulated by IGFBP-3 depletion, which suggests that MEK/ERK signaling may be involved in IGFBP-3-mediated cell migration. We therefore conclude that intracellular IGFBP-3 enhances cell migration independently of the cell cycle in TSCC with a higher metastatic potential.
Collapse
Affiliation(s)
- Esther Feng Ying Ng
- Department of Oral Radiation Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical & Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Atsushi Kaida
- Department of Oral Radiation Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical & Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.
| | - Hitomi Nojima
- Department of Oral Radiation Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical & Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Masahiko Miura
- Department of Oral Radiation Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical & Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.
| |
Collapse
|
15
|
Hawkins CC, Jones AB, Gordon ER, Williford SE, Harsh Y, Ziebro JK, Landis CJ, Gc S, Crossman DK, Cooper SJ, Ramanadham S, Doan N, Hjelmeland AB. Targeting Acid Ceramidase Inhibits Glioblastoma Cell Migration through Decreased AKT Signaling. Cells 2022; 11:1873. [PMID: 35741006 PMCID: PMC9221433 DOI: 10.3390/cells11121873] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 01/12/2023] Open
Abstract
Glioblastoma (GBM) remains one of the most aggressive cancers, partially due to its ability to migrate into the surrounding brain. The sphingolipid balance, or the balance between ceramides and sphingosine-1-phosphate, contributes to the ability of GBM cells to migrate or invade. Of the ceramidases which hydrolyze ceramides, acid ceramidase (ASAH1) is highly expressed in GBM samples compared to non-tumor brain. ASAH1 expression also correlates with genes associated with migration and focal adhesion. To understand the role of ASAH1 in GBM migration, we utilized shRNA knockdown and observed decreased migration that did not depend upon changes in growth. Next, we inhibited ASAH1 using carmofur, a clinically utilized small molecule inhibitor. Inhibition of ASAH1 by carmofur blocks in vitro migration of U251 (GBM cell line) and GBM cells derived from patient-derived xenografts (PDXs). RNA-sequencing suggested roles for carmofur in MAPK and AKT signaling. We found that carmofur treatment decreases phosphorylation of AKT, but not of MAPK. The decrease in AKT phosphorylation was confirmed by shRNA knockdown of ASAH1. Our findings substantiate ASAH1 inhibition using carmofur as a potential clinically relevant treatment to advance GBM therapeutics, particularly due to its impact on migration.
Collapse
Affiliation(s)
- Cyntanna C. Hawkins
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| | - Amber B. Jones
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| | - Emily R. Gordon
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; (E.R.G.); (S.J.C.)
| | - Sarah E. Williford
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| | - Yuvika Harsh
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| | - Julia K. Ziebro
- Graduate Biomedical Sciences, Division of Neuropathology, Department of Pathology, O’Neal Comprehensive Cancer Center, University of Alabama School of Medicine, Birmingham, AL 35233, USA;
| | - Catherine J. Landis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| | - Sajina Gc
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Sara J. Cooper
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; (E.R.G.); (S.J.C.)
| | - Sasanka Ramanadham
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ninh Doan
- Baptist South Medical Center, Montgomery, AL 36116, USA;
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (C.C.H.); (A.B.J.); (S.E.W.); (Y.H.); (C.J.L.); (S.G.); (S.R.)
| |
Collapse
|
16
|
Qi Q, Wang Q, Wang Z, Gao W, Gong X, Wang L. Visnagin inhibits cervical cancer cells proliferation through the induction of apoptosis and modulation of PI3K/AKT/mTOR and MAPK signaling pathway. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
17
|
A novel partial duplication in OPHN1, associated with vermis cerebellar hypoplasia, seizures and developmental delay. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
Anti-Cancer Effects of α-Cubebenoate Derived from Schisandra chinensis in CT26 Colon Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030737. [PMID: 35164001 PMCID: PMC8839175 DOI: 10.3390/molecules27030737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/26/2022]
Abstract
α-Cubebenoate derived from Schisandra chinensis has been reported to possess anti-allergic, anti-obesity, and anti-inflammatory effects and to exhibit anti-septic activity, but its anti-cancer effects have not been investigated. To examine the anti-cancer activity of α-cubebenoate, we investigated its effects on the proliferation, apoptosis, and metastasis of CT26 cells. The viabilities of CT26 cells (a murine colorectal carcinoma cell line) and HCT116 cells (a human colon cancer cell line) were remarkably and dose-dependently diminished by α-cubebenoate, whereas the viability of CCD-18Co cells (a normal human fibroblast cell line) were unaffected. Furthermore, α-cubebenoate treatment increased the number of apoptotic CT26 cells as compared with Vehicle-treated cells and increased Bax, Bcl-2, Cas-3, and Cleaved Cas-3 protein levels by activating the MAP kinase signaling pathway. α-Cubebenoate also suppressed CT26 migration by regulating the PI3K/AKT signaling pathway. Furthermore, similar reductions were observed in the expression levels of some migration-related proteins including VEGFA, MMP2, and MMP9. Furthermore, reduced VEGFA expression was found to be accompanied by the phosphorylations of FAK and MLC in the downstream signaling pathway of adhesion protein. The results of the present study provide novel evidence that α-cubebenoate can stimulate apoptosis and inhibit metastasis by regulating the MAPK, PI3K/AKT, and FAK/MLC signaling pathways.
Collapse
|
19
|
Jung HW, Hwang JH. Anticancer Effects of Ursi Fel Extract and Its Active Compound, Ursodeoxycholic Acid, in FRO Anaplastic Thyroid Cancer Cells. Molecules 2021; 26:molecules26175309. [PMID: 34500742 PMCID: PMC8434085 DOI: 10.3390/molecules26175309] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 01/13/2023] Open
Abstract
Anaplastic thyroid cancer (ATC) is one of the most fatal human malignancies. Ursi Fel (UF) is the bile of a brown bear that has been traditionally used for heat clearance and toxin relief in Korean and Chinese medicines. In this study, we determined the anticancer effects of a UF extract and its active compound, ursodeoxycholic acid (UDCA), in FRO human ATC cells. FRO cells were treated with UF extract and UDCA at different concentrations for various durations. Cell viability was measured using an MTT assay. Cell apoptosis was investigated by flow cytometric analysis following Annexin V and propidium iodide (PI) staining, and Hoechst staining was used to observe nuclear fragmentation. The expression of pro-apoptotic (Bax, caspase-3, cytochrome c, and PARP), anti-apoptotic (Bcl-2), and angiogenetic (TGF-β, VEGF, N-cadherin, and sirtuin-1) proteins and the phosphorylation of Akt and mechanistic target of rapamycin (mTOR) were determined by western blot analysis. Treatment with UF extract at 10, 25, and 50 μg/mL and UDCA at 25, 50, and 100 μM/mL significantly inhibited the growth of FRO cells in a dose-dependent manner. Flow cytometry and Hoechst staining revealed an increase in the apoptosis of FRO cells mediated by UF extract and UDCA in a dose-dependent manner. UF extract (25 and 50 μg) and UDCA (50 and 100 μM) significantly increased the expression of Bax, caspase-3, cytochrome c, and PARP and inhibited the expression of Bcl-2, TGF-β, VEGF, N-cadherin, and sirtuin-1 in FRO cells. Furthermore, UF extract and UDCA treatment stimulated Akt phosphorylation and inhibited mTOR phosphorylation in these cells. These results indicate that UF extract and UDCA exert anticancer properties in FRO cells by inducing apoptosis and inhibiting angiogenesis via regulating the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hyo Won Jung
- Department of Herbology, College of Korean Medicine, Dongguk University, Dongdae-ro 123, Gyeongju 38066, Korea;
| | - Ji Hye Hwang
- Department of Acupuncture & Moxibustion Medicine, College of Korean Medicine, Gachon University, Seongnam 13120, Korea
- Correspondence: ; Tel.: +82-32-770-1300
| |
Collapse
|
20
|
Scopetti D, Piobbico D, Brunacci C, Pieroni S, Bellezza G, Castelli M, Ludovini V, Tofanetti FR, Cagini L, Sidoni A, Puxeddu E, Della-Fazia MA, Servillo G. INSL4 as prognostic marker for proliferation and invasiveness in Non-Small-Cell Lung Cancer. J Cancer 2021; 12:3781-3795. [PMID: 34093787 PMCID: PMC8176261 DOI: 10.7150/jca.51332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell-lung cancer accounts for 80-85% of all forms of lung cancer as leading cause of cancer-related death in human. Despite remarkable advances in the diagnosis and therapy of lung cancer, no significant improvements have thus far been achieved in terms of patients' prognosis. Here, we investigated the role of INSL4 - a member of the relaxin-family - in NSCLC. We overexpressed INSL4 in NSCLC cells to analyse in vitro the growth rate and the tumourigenic features. We investigated the signalling pathways engaged in INSL4 overexpressing cells and the tumour growth ability by studying the tumour development in a patient derived tumour xenograft mouse model. We found an INSL4 cell growth promoting effect in vitro in H1299 cells and in vivo in NOD/SCID mice. Surprisingly, in NSCLC-A549 cells, INSL4 overexpression has not similar effect, despite huge basal INSL4-mRNA expression respect to H1299. The INSL4-mRNA analysis of eight different NSCLC-derived cell lines, revealed highly difference in the INSL4-mRNA amount. Transfection of NSCLC lines with INSL4-Myc showed huge level of INSL4-mRNA with a very low amount of protein expressed. Notably, similar discrepancy has been observed in NSCLC patients. However, in a cohort of NSCLC patients analysing a database, we found a significant inverse correlation between INSL4 expression and Overall Survival. By combining the in vitro and in vivo results, suggest that in patients whose NSCLC adenocarcinoma spontaneously expressed high levels of INSL4 post-transcriptional modifications affecting INSL4 do not allow to assess precision therapy in selected patients without consider protein INSL4 amount.
Collapse
Affiliation(s)
- Damiano Scopetti
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Danilo Piobbico
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Cinzia Brunacci
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Stefania Pieroni
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Guido Bellezza
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia- Italy
| | - Marilena Castelli
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Vienna Ludovini
- Medical Oncology, S. Maria Della Misericordia Hospital, Perugia, Italy
| | | | - Lucio Cagini
- Department of Medicine and Surgery, Section of internal medicine, angiology and atherosclerosis diseases
| | - Angelo Sidoni
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, University of Perugia, Perugia- Italy
| | - Efisio Puxeddu
- Department of Medicine and Surgery, Section of internal medicine and endocrine and metabolic sciences
| | - Maria Agnese Della-Fazia
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| | - Giuseppe Servillo
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, Perugia- Italy
| |
Collapse
|
21
|
Kang MJ, Kim JE, Park JW, Choi HJ, Bae SJ, Choi SI, Hong JT, Hwang DY. Effects of Gallotannin-Enriched Extract of Galla Rhois on the Activation of Apoptosis, Cell Cycle Arrest, and Inhibition of Migration Ability in LLC1 Cells and LLC1 Tumors. Pathol Oncol Res 2021; 27:588084. [PMID: 34257536 PMCID: PMC8262247 DOI: 10.3389/pore.2021.588084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 03/16/2021] [Indexed: 12/19/2022]
Abstract
Gallotannin (GT) and GT-enriched extracts derived from various sources are reported to have anti-tumor activity in esophageal, colon and prostate tumors, although their anti-tumor effects have not been determined in lung carcinomas. To investigate the anti-tumor activity of GT-enriched extract of galla rhois (GEGR) against lung carcinomas, alterations in the cytotoxicity, apoptosis activation, cell cycle progression, migration ability, tumor growth, histopathological structure, and the regulation of signaling pathways were analyzed in Lewis lung carcinoma (LLC1) cells and LLC1 tumor bearing C57BL/6NKorl mice, after exposure to GEGR. A high concentration of GT (69%) and DPPH scavenging activity (IC50=7.922 µg/ml) was obtained in GEGR. GEGR treatment exerted strong cytotoxicity, cell cycle arrest at the G2/M phase and subsequent activation of apoptosis, as well as inhibitory effects on the MAPK pathway and PI3K/AKT mediated cell migration in LLC1 cells. In the in vivo syngeneic model, exposure to GEGR resulted in suppressed growth of the LLC1 tumors, as well as inhibition of NF-κB signaling and their inflammatory cytokines. Taken together, our results provide novel evidence that exposure to GEGR induces activation of apoptosis, cell cycle arrest, and inhibition of cell migration via suppression of the MAPK, NF-κB and PI3K/AKT signaling pathways in LLC1 cells and the LLC1 syngeneic model.
Collapse
Affiliation(s)
- Mi Ju Kang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Ji Eun Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Ji Won Park
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Hyun Jun Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Su Ji Bae
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| | - Sun Il Choi
- Division of Convergence Technology, Research Institute of National Cancer Center, Goyang, South Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Chungju, Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Korea
| |
Collapse
|
22
|
Fraldi M, Cutolo A, Carotenuto AR, Palumbo S, Pugno N. A lesson from earthquake engineering for selectively damaging cancer cell structures. J Mech Behav Biomed Mater 2021; 119:104533. [PMID: 33895664 DOI: 10.1016/j.jmbbm.2021.104533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 01/04/2023]
Abstract
The progressive falling of barriers among disciplines is opening unforeseen scenarios in diagnosis and treatment of cancer diseases. By sharing models and mature knowledge in physics, engineering, computer sciences and molecular biology, synergistic efforts have in fact contributed in the last years to re-think still unsolved problems, shedding light on key roles of mechanobiology in tumors and envisaging new effective strategies for a precise medicine. The use of ultrasounds for altering cancer cells' program is one of the most attracting grounds to be explored in oncophysics, although how to administer mechanical energy to impair selected cell structures and functions simultaneously overcoming the critical trade-off between the impact of the cure and the patient risk still remains an open issue. Within this framework, by starting from the theoretical possibility of selectively attacking malignant cells by exploiting the stiffness discrepancies between tumor and healthy single cells, first proposed by Fraldi et al. (2015), we here investigate the in-frequency response of an overall spherical close-packing of geometrically equal polyhedral cells to gain insights into how mechanical resonance and vibration-induced failure phenomena can be oriented to destroy specific target units when both the cell populations coexist, as it happens for in vivo cases. Inspired by the dynamic action of earthquakes - which fracture only selected elements among adjacent ones in the same structure or damage individual constructions in contiguous buildings - we study the harmonic response of hierarchically architectured cell agglomerates, inhabited by both tumor and healthy cells that interact mutually throughout the extra-cellular matrix and whose cytoskeleton is modeled as a nonlinear soft-tensegrity structure. Numerical Finite Element results show that, at frequencies compatible with low intensity therapeutic ultrasounds, mechanical resonance and possible fatigue cycles of the pre-stressed actin filaments and microtubules can be selectively induced in cancer cells as a function of the global volume fraction of the cell species, paving the way for future engineered treatment protocols.
Collapse
Affiliation(s)
- Massimiliano Fraldi
- Department of Structures for Engineering and Architecture, University of Napoli Federico II, Italy.
| | - Arsenio Cutolo
- Department of Structures for Engineering and Architecture, University of Napoli Federico II, Italy
| | | | - Stefania Palumbo
- Department of Structures for Engineering and Architecture, University of Napoli Federico II, Italy
| | - Nicola Pugno
- Laboratory of Bio-inspired, Bionic, Nano, Meta Materials and Mechanics, Department of Civil, Environmental and Mechanical Engineering, University of Trento, Italy; School of Engineering and Materials Science, Queen Mary University of London, UK.
| |
Collapse
|
23
|
Pellecchia S, De Martino M, Esposito F, Quintavalle C, Fusco A, Pallante P. MPPED2 is downregulated in glioblastoma, and its restoration inhibits proliferation and increases the sensitivity to temozolomide of glioblastoma cells. Cell Cycle 2021; 20:716-729. [PMID: 33734003 DOI: 10.1080/15384101.2021.1901042] [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] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and lethal neoplasia of the central nervous system in adults. Based on the molecular signature genes, GBM has been classified in proneural, neural, mesenchymal and classical subtypes. The Metallophosphoesterase-domain-containing protein 2 (MPPED2) gene encodes a metallophosphodiesterase protein highly conserved throughout the evolution. MPPED2 downregulation, likely due to its promoter hypermethylation, has been found in several malignant neoplasias and correlated with a poor prognosis. In this study, we aimed to investigate the expression and the functional role of MPPED2 in GBM. TCGA and Gravendeel databases were employed to explore the MPPED2 expression levels in this type of tumor. We have found that MPPED2 expression is downregulated in GBM patients, showing a positive correlation with survival. Moreover, TCGA and Gravendeel data also revealed that MPPED2 expression negatively correlates with the most aggressive mesenchymal subtype. Additionally, the restoration of MPPED2 expression in U251 and GLI36 GBM cell lines decreases cell growth, migration and enhanced the sensitivity to the temozolomide, inducing apoptotic cell death, of GBM cells. These findings suggest that the restoration of MPPED2 function can be taken into consideration for an innovative GBM therapy.
Collapse
Affiliation(s)
- Simona Pellecchia
- Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II", Naples, Italy
| | - Marco De Martino
- Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy.,Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Francesco Esposito
- Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II", Naples, Italy
| | - Cristina Quintavalle
- Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| | - Alfredo Fusco
- Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples "Federico II", Naples, Italy
| | - Pierlorenzo Pallante
- Institute for Experimental Endocrinology and Oncology (IEOS) "G. Salvatore", National Research Council (CNR), Naples, Italy
| |
Collapse
|
24
|
Zhang B, Zhang Y, Jiang X, Su H, Wang Q, Wudu M, Jiang J, Ren H, Xu Y, Liu Z, Qiu X. JMJD8 Promotes Malignant Progression of Lung Cancer by Maintaining EGFR Stability and EGFR/PI3K/AKT Pathway Activation. J Cancer 2021; 12:976-987. [PMID: 33442397 PMCID: PMC7797639 DOI: 10.7150/jca.50234] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022] Open
Abstract
JMJD8 is a JmjC domain-containing protein that has not been widely examined, despite its potential role in malignant tumor development. The underlying biological functions and molecular mechanisms of JMJD8 in non-small-cell lung cancer (NSCLC) remain unclear. Herein, we explored the relationship between JMJD8 and the activation of malignancy pathways in NSCLC. Immunohistochemical analyses revealed that high JMJD8 expression significantly correlated with cell differentiation and advanced TNM stages of NSCLC. The overexpression of JMJD8 promoted cell proliferation and invasion in vitro. Upon JMJD8 knockdown in lung cancer cell lines, cyclin B1, RhoA, RhoC, MMP9, and N-cadherin were down-regulated, and p21 and E-cadherin were conversely up-regulated. Key factors in the PI3K/AKT signaling pathway, such as p‑AKT, showed clear decreases in expression; additionally, the expression of epidermal growth factor receptor (EGFR), which functions upstream of PI3K, was altered. Co-immunoprecipitation experiments indicated that JMJD8 interacts with EGFR, and JMJD8 knockdown accelerated EGFR degradation. Our results suggested that JMJD8 functions as an oncogenic regulator in NSCLC. We found that JMJD8 promotes carcinogenic activity in NSCLC cells by facilitating EGFR stability, thereby activating the downstream PI3K/AKT signaling pathway. JMJD8 shows potential as a prognostic marker for lung cancer patients, providing a new target for therapeutic strategies.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yao Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xizi Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hongbo Su
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qiongzi Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Muli Wudu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jun Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hongjiu Ren
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yitong Xu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Zongang Liu
- Department of Thoracic Surgical, Shengjing Hospital Affiliated with China Medical University, Shenyang, China
| | - Xueshan Qiu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
25
|
Noh JJ, Kim MS, Cho YJ, Jeong SY, Lee YY, Ryu JY, Choi JJ, Bae I, Wu Z, Kim BG, Hwang JR, Lee JW. Anti-Cancer Activity of As 4O 6 and its Efficacy in a Series of Patient-Derived Xenografts for Human Cervical Cancer. Pharmaceutics 2020; 12:pharmaceutics12100987. [PMID: 33086573 PMCID: PMC7590205 DOI: 10.3390/pharmaceutics12100987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/12/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022] Open
Abstract
Purpose: To investigate the anti-cancer effects of tetraarsenic hexoxide (TAO, As4O6) in cervical cancer cell lines and in a series of patient-derived xenograft (PDX) mouse models. Methods: Human cervical cancer cell lines, including HeLa, SiHa and CaSki, and human umbilical vein endothelial cells (HUVECs), were used to evaluate the anti-cancer activity of TAO. Cellular proliferation, apoptosis, and enzyme-linked immunosorbent assay (ELISA) for matrix metallopeptidase 2 (MMP-2) and 9 (MMP-9) were assessed. The tumor weights of the PDXs that were given TAO were measured. The PDXs included primary squamous cell carcinoma, primary adenocarcinoma, recurrent squamous cell carcinoma, and recurrent adenocarcinoma. Results: TAO significantly decreased cellular proliferation and increased apoptosis in cervical cancer cell lines and HUVEC. The functional studies on the cytotoxicity of TAO revealed that it inhibited the activation of Akt and vascular endothelial growth factor receptor 2 (VEGFR2). It also decreased the concentrations of MMP-2 in both cervical cancer cell lines and HUVECs. Active caspase-3 and p62 were both increased by the treatment of TAO, indicating increased rates of apoptosis and decreased rates of autophagy, respectively. In vivo studies with PDXs revealed that TAO significantly decreased tumor weight for both primary squamous cell carcinoma and adenocarcinoma of the cervix. However, this anti-cancer effect was not seen in PDXs with recurrent cancers. Nevertheless, the combination of TAO with cisplatin significantly decreased tumor weight in PDX models for both primary and recurrent cancers. Conclusions: TAO exerted inhibitory effects on angiogenesis, cellular migration, and autophagy, and it showed stimulatory effects on apoptosis. Overall, it demonstrated anti-cancer effects in animal models for human cervical cancer.
Collapse
Affiliation(s)
- Joseph J. Noh
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.J.N.); (S.-Y.J.); (Y.-Y.L.)
| | - Myeong-Seon Kim
- Department of Obstetrics and Gynecology, St. Vincent’s Hospital, Catholic University of Korea, Seoul 16247, Korea;
| | - Young-Jae Cho
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.-J.C.); (J.-Y.R.); (J.-J.C.)
| | - Soo-Young Jeong
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.J.N.); (S.-Y.J.); (Y.-Y.L.)
| | - Yoo-Young Lee
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.J.N.); (S.-Y.J.); (Y.-Y.L.)
| | - Ji-Yoon Ryu
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.-J.C.); (J.-Y.R.); (J.-J.C.)
| | - Jung-Joo Choi
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.-J.C.); (J.-Y.R.); (J.-J.C.)
| | - Illju Bae
- Chemas Co., Ltd., Seoul 06163, Korea; (I.B.); (Z.W.)
| | - Zhaoyan Wu
- Chemas Co., Ltd., Seoul 06163, Korea; (I.B.); (Z.W.)
| | - Byoung-Gie Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.J.N.); (S.-Y.J.); (Y.-Y.L.)
- Correspondence: (B.-G.K.); (J.R.H.); (J.-W.L.); Tel.: +82-2-3410-1382 (J.-W.L.); Fax: +82-2-3410-0630 (J.-W.L.)
| | - Jae Ryoung Hwang
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (Y.-J.C.); (J.-Y.R.); (J.-J.C.)
- Correspondence: (B.-G.K.); (J.R.H.); (J.-W.L.); Tel.: +82-2-3410-1382 (J.-W.L.); Fax: +82-2-3410-0630 (J.-W.L.)
| | - Jeong-Won Lee
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.J.N.); (S.-Y.J.); (Y.-Y.L.)
- Correspondence: (B.-G.K.); (J.R.H.); (J.-W.L.); Tel.: +82-2-3410-1382 (J.-W.L.); Fax: +82-2-3410-0630 (J.-W.L.)
| |
Collapse
|
26
|
Lee EH, Chun SY, Kim B, Yoon BH, Lee JN, Kim BS, Yoo ES, Lee S, Song PH, Kwon TG, Ha YS. Knockdown of TRPM7 prevents tumor growth, migration, and invasion through the Src, Akt, and JNK pathway in bladder cancer. BMC Urol 2020; 20:145. [PMID: 32907556 PMCID: PMC7488071 DOI: 10.1186/s12894-020-00714-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Background Bladder cancer (BC) is one of the most common malignancies of the urinary tract. The role of transient receptor potential melastatin 7 (TRPM7) in BC remains unclear. The aim of this study was to investigate the function and signal transduction pathway of TRPM7 in BC. Methods T24 and UMUC3 cells were used to evaluate the molecular mechanism of TRPM7 by immunoblot analysis. Small interfering RNA was used to knockdown TRPM7, and the effect of silencing TRPM7 was studied by wound healing, migration, and invasion assays in T24 and UMUC3 cells. Xenograft model study was obtained to analyze the effect of TRPM7 inhibition in vivo. Results Silencing of TRPM7 decreased the migration and invasion ability of T24 and UMUC3 cells. The phosphorylation of Src, Akt, and JNK (c-Jun N-terminal kinase) was also suppressed by TRPM7 silencing. Src, Akt, and JNK inhibitors effectively inhibited the migration and invasion of T24 and UMUC3 cells. In addition, the TRPM7 inhibitor, carvacrol, limited the tumor size in a xenograft model. Conclusion Our data reveal that TRPM7 regulates the migration and invasion of T24 and UMUC3 cells via the Src, Akt, and JNK signaling pathway. Therefore, TRPM7 suppression could be a potential treatment for BC patients.
Collapse
Affiliation(s)
- Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - So Young Chun
- BioMedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Bomi Kim
- BioMedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Bo Hyun Yoon
- BioMedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jun Nyung Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.,Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea.,Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Eun Sang Yoo
- Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics Based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Phil Hyun Song
- Department of Urology, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Tae Gyun Kwon
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea. .,Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea. .,Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.
| | - Yun-Sok Ha
- Joint Institute for Regenerative Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea. .,Department of Urology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea. .,Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea.
| |
Collapse
|
27
|
Zhang S, Wang J, Yao T, Tao M. LncRNA ZFAS1/miR-589 regulates the PTEN/PI3K/AKT signal pathway in the proliferation, invasion and migration of breast cancer cells. Cytotechnology 2020; 72:415-425. [PMID: 32189153 PMCID: PMC7225242 DOI: 10.1007/s10616-020-00388-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) is a common clinical disease and the second leading cause of cancer death in women. Long noncoding RNA (lncRNA) and microRNA (miRNA) are reported to be involved in the development of BC. The present study aimed to investigate whether LncRNA ZFAS1 could regulate the proliferation, invasion and migration of breast cancer cells by targeting miR-589 through PTEN/PI3K/AKT signal pathway. The expression of ZFAS1 and miR-589 in BC cells and transfection effects were determined by RT-qPCR analysis. The abilities of proliferation, colony formation, invasion and migration of breast cancer cells were analyzed by CCK-8 assay, colony formation assay, transwell assay and wound healing assay respectively. The expression of MMP2, MMP9, Bcl2, Bax, cleaved caspase3, PTEN, p-PI3K, p-AKT, PI3K and AKT was detected by Western blot. The flow cytometry analysis was used to detect cell apoptosis. As a result, ZFAS1 expression was increased and miR-589 expression was decreased in BC cells. And, miR-589 was demonstrated to be a target of ZFAS1. ZFAS1 overexpression could inhibit the proliferation, colony formation, invasion and migration of BC cells while miR-589 overexpression could reverse the changes. In addition, ZFAS1 overexpression suppressed the expression of PI3K/AKT signal pathway by activating the PTEN expression while miR-589 overexpression could reverse the changes. Moreover, PTEN is one of the gene targets of miR-589. In conclusion, this study indicated that ZFAS1 inhibited the proliferation, invasion and migration of breast cancer cells by targeting miR-589 through regulating the PTEN/PI3K/AKT signal pathway.
Collapse
Affiliation(s)
- Song Zhang
- Department of Medical Oncology, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Jin Wang
- Department of Clinical Laboratory Science, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Tingjing Yao
- Department of Surgical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China
| | - Min Tao
- Department of Medical Oncology, First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
28
|
Sinomenine Inhibits Migration and Invasion of Human Lung Cancer Cell through Downregulating Expression of miR-21 and MMPs. Int J Mol Sci 2020; 21:ijms21093080. [PMID: 32349289 PMCID: PMC7247699 DOI: 10.3390/ijms21093080] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Sinomenine is an alkaloid derived from Sinomenium acutum. Recent studies have found that sinomenine can inhibit various cancers by inhibiting the proliferation, migration and invasion of tumors and inducing apoptosis. This study aims to investigate the effect and mechanism of sinomenine on inhibiting the migration and invasion of human lung adenocarcinoma cells in vitro. The results demonstrate that viabilities of A549 and H1299 cells were inhibited by sinomenine in a dose-dependent manner. When treated with sub-toxic doses of sinomenine, cell migration and invasion are markedly suppressed. Sinomenine decreases the mRNA level of matrix metalloproteinase-2 (MMP-2), MMP-9, and the extracellular inducer of matrix metalloproteinase (EMMPRIN/CD147), but elevates the expression of reversion-inducing cysteine-rich proteins with kazal motifs (RECK) and the tissue inhibitor of metalloproteinase-1 (TIMP-1) and TIMP-2. In addition, sinomenine significantly increases the expression of the epithelial marker E-cadherin but concomitantly decreases the expression of the mesenchymal marker vimentin, suggesting that it suppresses epithelial–mesenchymal transition (EMT). Moreover, sinomenine downregulates oncogenic microRNA-21 (miR-21), which has been known to target RECK. The downregulation of miR-21 decreases cell invasion, while the upregulation of miR-21 increases cell invasion. Furthermore, the downregulation of miR-21 stimulates the expression of RECK, TIMP-1/-2, and E-cadherin, but reduces the expression of MMP-2/-9, EMMPRIN/CD147, and vimentin. Taken together, the results reveal that the inhibition of A549 cell invasion by sinomenine may, at least in part, be through the downregulating expression of MMPs and miR-21. These findings demonstrate an attractive therapeutic potential for sinomenine in lung cancer anti-metastatic therapy.
Collapse
|
29
|
Chen YF, Shih PC, Kuo HM, Yang SN, Lin YY, Chen WF, Tzou SJ, Liu HT, Chen NF. TP3, an antimicrobial peptide, inhibits infiltration and motility of glioblastoma cells via modulating the tumor microenvironment. Cancer Med 2020; 9:3918-3931. [PMID: 32266797 PMCID: PMC7286473 DOI: 10.1002/cam4.3005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 02/19/2020] [Accepted: 02/27/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a cancer of the central nervous system with limited therapeutic outcomes. Infiltrating cancer cells are the contributing factor to high GBM malignancy. The intracranial brain cancer cell infiltration is a complex cascade involving adhesion, migration, and invasion. An arsenal of natural products has been under exploration to overcome GBM malignancy. This study applied the antimicrobial peptide tilapia piscidin 3 (TP3) to GBM8401, U87MG, and T98G cells. The cellular assays and microscopic observations showed that TP3 significantly attenuated cell adhesion, migration, and invasion. A live‐cell video clip showed the inhibition of filopodia protrusions and cell attachment. Probing at the molecular levels showed that the proteolytic activities (from secretion), the mRNA and protein expression levels of matrix metalloproteinases‐2 and ‐9 were attenuated. This result strongly evidenced that both invasion and metastasis were inhibited, although metastatic GBM is rare. Furthermore, the protein expression levels of cell‐mobilization regulators focal adhesion kinase and paxillin were decreased. Similar effects were observed in small GTPase (RAS), phosphorylated protein kinase B (AKT) and MAP kinases such as extracellular signal‐regulated kinases (ERK), JNK, and p38. Overall, TP3 showed promising activities to prevent cell infiltration and metastasis through modulating the tumor microenvironment balance, suggesting that TP3 merits further development for use in GBM treatments.
Collapse
Affiliation(s)
- Ying-Fa Chen
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center for Parkinson's Disease, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Po-Chang Shih
- UCL School of Pharmacy, University College London, London, UK.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.,Center for Neuroscience, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - San-Nan Yang
- Department of Internal Medicine, E-DA Hospital and College of Medicine, I-SHOU University, Kaohsiung, Taiwan
| | - Yen-You Lin
- Department of Orthopedic Surgery, Ping-Tung Christian Hospital, Pingtung, Taiwan
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan.,Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Neurosurgery, Xiamen Chang Gung Hospital, Xiamen, Fujian, China
| | - Shiow-Jyu Tzou
- Department of Nursing, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Nan-Fu Chen
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan.,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
30
|
Hwang SH, Bang S, Kim W, Chung J. Von Hippel-Lindau tumor suppressor (VHL) stimulates TOR signaling by interacting with phosphoinositide 3-kinase (PI3K). J Biol Chem 2020; 295:2336-2347. [PMID: 31959630 DOI: 10.1074/jbc.ra119.011596] [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: 10/24/2019] [Revised: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cell growth is positively controlled by the phosphoinositide 3-kinase (PI3K)-target of rapamycin (TOR) signaling pathway under conditions of abundant growth factors and nutrients. To discover additional mechanisms that regulate cell growth, here we performed RNAi-based mosaic analyses in the Drosophila fat body, the primary metabolic organ in the fly. Unexpectedly, the knockdown of the Drosophila von Hippel-Lindau (VHL) gene markedly decreased cell size and body size. These cell growth phenotypes induced by VHL loss of function were recovered by activation of TOR signaling in Drosophila Consistent with the genetic interactions between VHL and the signaling components of PI3K-TOR pathway in Drosophila, we observed that VHL loss of function in mammalian cells causes decreased phosphorylation of ribosomal protein S6 kinase and Akt, which represent the main activities of this pathway. We further demonstrate that VHL activates TOR signaling by directly interacting with the p110 catalytic subunit of PI3K. On the basis of the evolutionarily conserved regulation of PI3K-TOR signaling by VHL observed here, we propose that VHL plays an important role in the regulation and maintenance of proper cell growth in metazoans.
Collapse
Affiliation(s)
- Sun-Hong Hwang
- School of Biological Sciences, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea
| | - Sunhoe Bang
- School of Biological Sciences, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea
| | - Wonho Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea
| | - Jongkyeong Chung
- School of Biological Sciences, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea; Institute of Molecular Biology and Genetics, Seoul National University, Gwanak-Gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
31
|
Pang D, Yang C, Li C, Zou Y, Feng B, Li L, Liu W, Luo Q, Chen Z, Huang C. Polyphyllin II inhibits liver cancer cell proliferation, migration and invasion through downregulated cofilin activity and the AKT/NF-κB pathway. Biol Open 2020; 9:bio.046854. [PMID: 31988091 PMCID: PMC7044461 DOI: 10.1242/bio.046854] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The morbidity and mortality of primary liver cancer is one of the highest amongst all cancers. Deficiency of effective treatment and characteristics of cancer metastasis are believed to be responsible for this situation, thus a great demand is required for new agent development. Polyphyllin II (PP2), an important steroidal saponin extracted from Rhizoma Paris, has emerged as a potential anti-cancer agent, but the effects of PP2 in liver cancers and its underlying mechanisms remain unexplored. In our study, we found that PP2 could remarkably suppress the proliferation of two liver cancer cell lines, HepG2 and BEL7402, resulting in significant cell death. Besides, low doses of PP2 have displayed properties that inhibit cellular motility and invasion of liver cancer cells. In addition, we have found that PP2-mediated cofilin activity suppression was implicated in the inhibition of liver cancer cell motility. Decreased expression of two major hydrolytic enzymes (MMP2/MMP9), through the AKT/NF-κB signaling pathway may also be also responsible for this process. Rescue experiments done with either non-phosphorylatable mutant cofilin-1 (S3A) transfection or an activator of the AKT pathway significantly reversed the inhibition effects of PP2 on liver cancer cells. Taken together, we report a potential agent for liver cancer treatment and reveal its underlying mechanisms.
Collapse
Affiliation(s)
- Dejiang Pang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Chengcheng Yang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Chao Li
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuanfeng Zou
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Lixia Li
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Wentao Liu
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qihui Luo
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhengli Chen
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Authors for correspondence (; )
| | - Chao Huang
- Laboratory of Experimental Animal Disease Model, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China,Authors for correspondence (; )
| |
Collapse
|
32
|
Wen Y, Cai X, Chen S, Fu W, Chai D, Zhang H, Zhang Y. 7-Methoxy-1-Tetralone Induces Apoptosis, Suppresses Cell Proliferation and Migration in Hepatocellular Carcinoma via Regulating c-Met, p-AKT, NF-κB, MMP2, and MMP9 Expression. Front Oncol 2020; 10:58. [PMID: 32117722 PMCID: PMC7020565 DOI: 10.3389/fonc.2020.00058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022] Open
Abstract
This study aimed to determine the anti-proliferative and anti-migratory effects of 7-methoxy-1-tetralone (MT) in hepatocellular carcinoma (HCC) cells. MTT assay assessed HCC cell viability; cell apoptosis of HCC cells was determined by flow cytometry; wound healing assay evaluated HCC cell migratory ability; protein expression levels were assessed using western blot assay; the in vivo antitumor effects of MT were tested in BALB/c nude mice and the pathological changes within the tumor tissues were evaluated by immunohistochemistry. MT treatment significantly suppressed the cell proliferative and migratory potentials of HepG2 cells, and induced HepG2 cell apoptosis. The western blot assay showed that MT treatment caused a suppression on c-Met, phosphorylated AKT (p-AKT), NF-κB, matrix metallopeptidase 2 (MMP2)/MMP9 protein levels in HepG2 cells. Further in vivo animal studies deciphered that MT treatment suppressed tumor growth of HepG2 cells in the nude mice, but had no effect on the body weight and the organ index of liver and spleen. Further immunohistochemistry analysis of the dissected tumor tissues showed that MT treatment significantly suppressed the protein expression levels of NF-κB, MMP9, MMP2, and p-AKT. In summary, the present study demonstrated the anti-tumor effects of MT on the HCC, and MT suppressed HCC progression possibly via regulating proliferation- and migration-related mediators including c-Met, p-AKT, NF-κB, MMP2, and MMP9 in HepG2 cells.
Collapse
Affiliation(s)
- Ying Wen
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Cell Biology and Medical Genetics, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaoyan Cai
- Department of Cell Biology and Medical Genetics, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shaolian Chen
- Department of Clinical Laboratory, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wei Fu
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Cell Biology and Medical Genetics, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dong Chai
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Cell Biology and Medical Genetics, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huainian Zhang
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Cell Biology and Medical Genetics, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongli Zhang
- Guangzhou Key Laboratory of Construction and Application of New Drug Screening Model Systems, Guangdong Pharmaceutical University, Guangzhou, China
- Department of Cell Biology and Medical Genetics, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
33
|
Chen Z, Lai H, Hou L, Chen T. Rational design and action mechanisms of chemically innovative organoselenium in cancer therapy. Chem Commun (Camb) 2020; 56:179-196. [PMID: 31782422 DOI: 10.1039/c9cc07683b] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Organo-seleno compounds (org-Se) have been widely used in antitumor, antiviral, and antiinflammatory therapy; antioxidation and other biological fields. As such, they have made an important contribution to overcoming various kinds of diseases, and researchers are increasingly attracted to org-Se's synthesis and functional design. This review is mainly focused on the design and synthesis of various kinds of org-Se, followed by their anticancer mechanisms such as the mitochondria mediated pathway induced by ROS, death receptor mediated pathways involving p53 phosphorylation, and the activation of the AMPK pathway to promote apoptosis. Org-Se also serves as a sensitizer in chemotherapy and radiotherapy, and an antagonist against the cytotoxic effects induced by chemotherapeutic agents. Finally, we will summarize the development of cancer-targeted org-Se containing complexes, and nanotechnology-based org-Se for anticancer application. This review could provide information for the future design of chemically innovative org-Se with anticancer potential, and shed light on the discovery of nanomaterial-based pharmaceuticals to improve drug development and formation.
Collapse
Affiliation(s)
- Zhen Chen
- The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | | | | | | |
Collapse
|
34
|
Tomatidine Represses Invasion and Migration of Human Osteosarcoma U2OS and HOS Cells by Suppression of Presenilin 1 and c-Raf-MEK-ERK Pathway. Molecules 2020; 25:molecules25020326. [PMID: 31941156 PMCID: PMC7024336 DOI: 10.3390/molecules25020326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma, which is the most prevalent malignant bone tumor, is responsible for the great majority of bone cancer-associated deaths because of its highly metastatic potential. Although tomatidine is suggested to serve as a chemosensitizer in multidrug-resistant tumors, the anti-metastatic effect of tomatidine in osteosarcoma is still unknown. Here, we tested the hypothesis that tomatidine suppresses migration and invasion, features that are associated with metastatic process in human osteosarcoma cells and also investigate its underlying pathway. Tomatidine, up to 100 μM, without cytotoxicity, inhibited the invasion and migration capabilities of human osteosarcoma U2OS and HOS cells and repressed presenilin 1 (PS-1) expression of U2OS cells. After the knockdown of PS-1, U2OS and HOS cells’ biological behaviors of cellular invasion and migratory potential were significantly reduced. While tomatidine significantly decreased the phosphorylation of c-Raf, mitogen/extracellular signal-regulated kinase (MEK), and extracellular signal-regulated protein kinase (ERK)1/2 in U2OS cells, no obvious influences on p-Jun N-terminal kinase, p38, and Akt, including their phosphorylation, were observed. In ERK 1 silencing U2 OS cells, tomatidine further enhanced the decrease of their migratory potential and invasive activities. We conclude that both PS-1 derived from U2OS and HOS cells and the c-Raf–MEK–ERK pathway contribute to cellular invasion and migration and tomatidine could inhibit the phenomenons. These findings indicate that tomatidine might be a potential candidate for anti-metastasis treatment of human osteosarcoma.
Collapse
|
35
|
Hakimee H, Hutamekalin P, Tanasawet S, Chonpathompikunlert P, Tipmanee V, Sukketsiri W. Metformin Inhibit Cervical Cancer Migration by Suppressing the FAK/Akt Signaling Pathway. Asian Pac J Cancer Prev 2019; 20:3539-3545. [PMID: 31870092 PMCID: PMC7173373 DOI: 10.31557/apjcp.2019.20.12.3539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Indexed: 01/27/2023] Open
Abstract
Background: Metformin, an antidiabetic drug, has been previously reported to have anti-cancer activities. However, its role in the control of cancer cell migration remains elusive. Methods: To examine the possible effect of metformin on migration of cervical cancer cells. The related mechanisms were further determined by immunocytochemistry and Western’s blotting assay. Results: The results showed that metformin treatment substantially inhibited the migration ability of cervical cancer cells. Consistently, the filopodia and lamellipodia formation were depleted after exposure to metformin. The suppression of migration mediated through the regulatory proteins such as focal adhesion kinase (FAK), ATP-dependent tyrosine kinase (Akt), Rac1 and RhoA after metformin treatment. Conclusion: Metformin displays antimigration effects in cervical cancer cells by inhibiting filopodia and lamellipodia formation through the suppression of FAK, Akt and its downstream Rac1 and RhoA protein. We propose that metformin could be a novel potential candidate as an antimetastatic cancer drug in the cervical cancer management.
Collapse
Affiliation(s)
- Henna Hakimee
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Pilaiwanwadee Hutamekalin
- Department of Physiology, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla,Thailand
| | - Supita Tanasawet
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla,Thailand
| | - Pennapa Chonpathompikunlert
- Expert Centre of Innovative Health Food (InnoFood), Thailand Institute of Scientific and Technological Research (TISTR), Pathumthani, Thailand
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla,Thailand
| | - Wanida Sukketsiri
- Department of Pharmacology, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
36
|
Hinz N, Jücker M. Distinct functions of AKT isoforms in breast cancer: a comprehensive review. Cell Commun Signal 2019; 17:154. [PMID: 31752925 PMCID: PMC6873690 DOI: 10.1186/s12964-019-0450-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AKT, also known as protein kinase B, is a key element of the PI3K/AKT signaling pathway. Moreover, AKT regulates the hallmarks of cancer, e.g. tumor growth, survival and invasiveness of tumor cells. After AKT was discovered in the early 1990s, further studies revealed that there are three different AKT isoforms, namely AKT1, AKT2 and AKT3. Despite their high similarity of 80%, the distinct AKT isoforms exert non-redundant, partly even opposing effects under physiological and pathological conditions. Breast cancer as the most common cancer entity in women, frequently shows alterations of the PI3K/AKT signaling. MAIN CONTENT A plethora of studies addressed the impact of AKT isoforms on tumor growth, metastasis and angiogenesis of breast cancer as well as on therapy response and overall survival in patients. Therefore, this review aimed to give a comprehensive overview about the isoform-specific effects of AKT in breast cancer and to summarize known downstream and upstream mechanisms. Taking account of conflicting findings among the studies, the majority of the studies reported a tumor initiating role of AKT1, whereas AKT2 is mainly responsible for tumor progression and metastasis. In detail, AKT1 increases cell proliferation through cell cycle proteins like p21, p27 and cyclin D1 and impairs apoptosis e.g. via p53. On the downside AKT1 decreases migration of breast cancer cells, for instance by regulating TSC2, palladin and EMT-proteins. However, AKT2 promotes migration and invasion most notably through regulation of β-integrins, EMT-proteins and F-actin. Whilst AKT3 is associated with a negative ER-status, findings about the role of AKT3 in regulation of the key properties of breast cancer are sparse. Accordingly, AKT1 is mutated and AKT2 is amplified in some cases of breast cancer and AKT isoforms are associated with overall survival and therapy response in an isoform-specific manner. CONCLUSIONS Although there are several discussed hypotheses how isoform specificity is achieved, the mechanisms behind the isoform-specific effects remain mostly unrevealed. As a consequence, further effort is necessary to achieve deeper insights into an isoform-specific AKT signaling in breast cancer and the mechanism behind it.
Collapse
Affiliation(s)
- Nico Hinz
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
37
|
Leong MML, Cheung AKL, Kwok TCT, Lung ML. Functional characterization of a candidate tumor suppressor gene, Mirror Image Polydactyly 1, in nasopharyngeal carcinoma. Int J Cancer 2019; 146:2891-2900. [PMID: 31609475 DOI: 10.1002/ijc.32732] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/01/2019] [Accepted: 10/07/2019] [Indexed: 12/24/2022]
Abstract
Mirror Image Polydactyly 1 (MIPOL1) is generally associated with congenital anomalies. However, its role in cancer development is poorly understood. Previously, by utilizing the functional complementation approach, microcell-mediated chromosome transfer (MMCT), a tumor suppressor gene, MIPOL1, was identified. MIPOL1 was confirmed to be downregulated in nasopharyngeal carcinoma (NPC) cells and tumor tissues, and re-expression of MIPOL1 induced tumor suppression. The aim of the current study is to further elucidate the functional tumor suppressive role of MIPOL1. In our study, with an expanded sample size of different clinical stages of NPC tumor tissues, we further confirmed the downregulation of MIPOL1 in different cancer stages. MIPOL1 re-expression down-regulated angiogenic factors and reduced phosphorylation of metastasis-associated proteins including AKT, p65, and FAK. In addition, MIPOL1 was confirmed to interact with a tumor suppressor, RhoB, and re-expression of MIPOL1 enhanced RhoB activity. The functional role of MIPOL1 was further validated by utilizing a panel of wild-type (WT) and truncated MIPOL1 expression constructs. The MIPOL1 tumor-suppressive effect can only be observed in the WT MIPOL1-expressing cells. In vitro and nude mice in vivo functional studies further confirmed the critical role of WT MIPOL1 in inhibiting migration, invasion and metastasis in NPC. Overall, our study provides strong evidence about the tumor-suppressive role of MIPOL1 in inhibiting angiogenesis and metastasis in NPC.
Collapse
Affiliation(s)
- Merrin M L Leong
- Department of Clinical Oncology, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Arthur K L Cheung
- Department of Clinical Oncology, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Tommy C T Kwok
- Department of Clinical Oncology, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Maria L Lung
- Department of Clinical Oncology, University of Hong Kong, Pok Fu Lam, Hong Kong.,Centre for Nasopharyngeal Carcinoma Research, University of Hong Kong, Pok Fu Lam, Hong Kong
| |
Collapse
|
38
|
Hegazy RR, Mansour DF, Salama AA, Abdel-Rahman RF, Hassan AM. Regulation of PKB/Akt-pathway in the chemopreventive effect of lactoferrin against diethylnitrosamine-induced hepatocarcinogenesis in rats. Pharmacol Rep 2019; 71:879-891. [PMID: 31442665 DOI: 10.1016/j.pharep.2019.04.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Abnormal activation of protein kinase B (PKB) is associated with many cancers. This makes inhibition of PKB signaling pathway a promising strategy for cancer therapy. Lactoferrin (Lf) has been reported for its inhibition of tumor growth and metastasis, however, the mechanism is not completely understood. Its anti-hepatocarcinogenic activity has not taken the deserved recognition despite the additional advantages of Lf as an antiviral against hepatitis C virus, the main cause of hepatocellular carcinoma (HCC), and as a targeting ligand for delivering chemotherapeutics to hepatoma cells. METHODS This study evaluated the anti-hepatocarcinogenic effect of Lf, and the role of PKB in this effect using diethylnitrosamine (DENA)-induced HCC rat model, and a primary cell culture prepared from the induced hepatic lesions (DENA-HCC cell culture). RESULTS Up-regulation of activated PKB in the hepatocytes of rats with DENA-induced HCC was observed, as measured biochemically in the liver homogenate, and localized immunohistochemically. This was accompanied by increment of hepatocytes proliferation, and expression of vascular endothelial growth factor and endothelial nitric oxide synthase. Involvement of PKB in DENA-induced HCC was confirmed by the observed decrease in cell proliferation in DENA-HCC cell culture that was treated with PKB inhibitor. In Lf-treated rats, a dose-dependent chemopreventive effect was observed, with decreased expression and activation of PKB, amelioration of the other DENA-induced alterations, and stimulation of apoptosis. In vitro, Lf blocked PKB activator-induced cell proliferation. CONCLUSION These findings support the chemopreventive activity of Lf against HCC, and suggest regulation of PKB-pathway as a potential mechanism underlying this effect.
Collapse
Affiliation(s)
- Rehab R Hegazy
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt.
| | - Dina F Mansour
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Abeer A Salama
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Rehab F Abdel-Rahman
- Department of Pharmacology, Medical Division, National Research Centre, Giza, Egypt
| | - Azza M Hassan
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
39
|
Ciardiello C, Leone A, Lanuti P, Roca MS, Moccia T, Minciacchi VR, Minopoli M, Gigantino V, De Cecio R, Rippa M, Petti L, Capone F, Vitagliano C, Milone MR, Pucci B, Lombardi R, Iannelli F, Di Gennaro E, Bruzzese F, Marchisio M, Carriero MV, Di Vizio D, Budillon A. Large oncosomes overexpressing integrin alpha-V promote prostate cancer adhesion and invasion via AKT activation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:317. [PMID: 31319863 PMCID: PMC6639931 DOI: 10.1186/s13046-019-1317-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/09/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Molecular markers for prostate cancer (PCa) are required to improve the early definition of patient outcomes. Atypically large extracellular vesicles (EVs), referred as "Large Oncosomes" (LO), have been identified in highly migratory and invasive PCa cells. We recently developed and characterized the DU145R80 subline, selected from parental DU145 cells as resistant to inhibitors of mevalonate pathway. DU145R80 showed different proteomic profile compared to parental DU145 cells, along with altered cytoskeleton dynamics and a more aggressive phenotype. METHODS Immunofluorescence staining and western blotting were used to identify blebbing and EVs protein cargo. EVs, purified by gradient ultra-centrifugations, were analyzed by tunable resistive pulse sensing and multi-parametric flow cytometry approach coupled with high-resolution imaging technologies. LO functional effects were tested in vitro by adhesion and invasion assays and in vivo xenograft model in nude mice. Xenograft and patient tumor tissues were analyzed by immunohistochemistry. RESULTS We found spontaneous blebbing and increased shedding of LO from DU145R80 compared to DU145 cells. LO from DU145R80, compared to those from DU145, carried increased amounts of key-molecules involved in PCa progression including integrin alpha V (αV-integrin). By incubating DU145 cells with DU145R80-derived LO we demonstrated that αV-integrin on LO surface was functionally involved in the increased adhesion and invasion of recipient cells, via AKT. Indeed either the pre-incubation of LO with an αV-integrin blocking antibody, or a specific AKT inhibition in recipient cells are able to revert the LO-induced functional effects. Moreover, DU145R80-derived LO also increased DU145 tumor engraftment in a mice model. Finally, we identified αV-integrin positive LO-like structures in tumor xenografts as well as in PCa patient tissues. Increased αV-integrin tumor expression correlated with high Gleason score and lymph node status. CONCLUSIONS Overall, this study is the first to demonstrate the critical role of αV-integrin positive LO in PCa aggressive features, adding new insights in biological function of these large EVs and suggesting their potential use as PCa prognostic markers.
Collapse
Affiliation(s)
- Chiara Ciardiello
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy.
| | - Alessandra Leone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Paola Lanuti
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy.,Department of Medicine and Aging Sciences, University "G. d'Annunzio", Chieti-Pescara, Italy
| | - Maria S Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Tania Moccia
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Valentina R Minciacchi
- Georg-Speyer-Haus Institute for Tumor biology and Experimental Therapy, Frankfurt, Germany
| | - Michele Minopoli
- Neoplastic Progression Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Massimo Rippa
- Institute of Applied Sciences and Intelligent Systems 'E. Caianiello' of CNR, Pozzuoli, Italy
| | - Lucia Petti
- Institute of Applied Sciences and Intelligent Systems 'E. Caianiello' of CNR, Pozzuoli, Italy
| | - Francesca Capone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Carlo Vitagliano
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Maria R Milone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Biagio Pucci
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Rita Lombardi
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Federica Iannelli
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Elena Di Gennaro
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Francesca Bruzzese
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy
| | - Marco Marchisio
- Centre on Aging Sciences and Translational Medicine (Ce.S.I.-Me.T.), University "G.d'Annunzio", Chieti-Pescara, Italy.,Department of Medicine and Aging Sciences, University "G. d'Annunzio", Chieti-Pescara, Italy
| | - Maria V Carriero
- Neoplastic Progression Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Naples, Italy
| | - Dolores Di Vizio
- Departments of Surgery, Pathology & Lab Medicine, and Biochemical Science, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alfredo Budillon
- Experimental Pharmacology Unit, Istituto Nazionale Tumori - IRCCS- Fondazione G. Pascale, Via M. Semmola, 80131, Naples, Italy.
| |
Collapse
|
40
|
Summer R, Krishna R, Schriner D, Cuevas-Mora K, Sales D, Para R, Roman J, Nieweld C, Gochuico BR, Romero F. Matrix metalloproteinase activity in the lung is increased in Hermansky-Pudlak syndrome. Orphanet J Rare Dis 2019; 14:162. [PMID: 31272455 PMCID: PMC6610946 DOI: 10.1186/s13023-019-1143-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/25/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive disorder characterized by oculocutaneous albinism and platelet dysfunction and can sometimes lead to a highly aggressive form of pulmonary fibrosis that mimics the fatal lung condition called idiopathic pulmonary fibrosis (IPF). Although the activities of various matrix metalloproteinases (MMPs) are known to be dysregulated in IPF, it remains to be determined whether similar changes in these enzymes can be detected in HPS. RESULTS Here, we show that transcript and protein levels as well as enzymatic activities of MMP-2 and -9 are markedly increased in the lungs of mice carrying the HPS Ap3b1 gene mutation. Moreover, immunohistochemical staining localized this increase in MMP expression to the distal pulmonary epithelium, and shRNA knockdown of the Ap3b1 gene in cultured lung epithelial cells resulted in a similar upregulation in MMP-2 and -9 expression. Mechanistically, we found that upregulation in MMP expression associated with increased activity of the serine/threonine kinase Akt, and pharmacological inhibition of this enzyme resulted in a dramatic suppression of MMP expression in Ap3b1 deficient lung epithelial cells. Similarly, levels and activity of different MMPs were also found to be increased in the lungs of mice carrying the Bloc3 HPS gene mutation and in the bronchoalveolar lavage fluid of subjects with HPS. However, an association between MMP activity and disease severity was not detected in these individuals. CONCLUSIONS In summary, our findings indicate that MMP activity is dysregulated in the HPS lung, suggesting a role for these proteases as biological markers or pathogenic players in HPS lung disease.
Collapse
Affiliation(s)
- Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Rachana Krishna
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - DeLeila Schriner
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Karina Cuevas-Mora
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Dominic Sales
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Rachel Para
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Jesse Roman
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Carl Nieweld
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Bernadette R. Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD USA
| | - Freddy Romero
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
- Center for Translational Medicine and Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, 1020 Locust Street, JAH 354, Philadelphia, PA 19107 USA
| |
Collapse
|
41
|
Elzaiat M, Herman L, Legois B, Léger T, Todeschini AL, Veitia RA. High-throughput Exploration of the Network Dependent on AKT1 in Mouse Ovarian Granulosa Cells. Mol Cell Proteomics 2019; 18:1307-1319. [PMID: 30992313 PMCID: PMC6601207 DOI: 10.1074/mcp.ra119.0014613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Indexed: 12/22/2022] Open
Abstract
The PI3K/AKT signaling pathway is known to regulate a broad range of cellular processes, and it is often altered in several types of cancers. Recently, somatic AKT1 mutations leading to a strong activation of this kinase have been reported in juvenile granulosa cell tumors. However, the molecular role of AKT1 in the supporting cell lineage of the ovary is still poorly understood. To get insights into its function in such cells, we depleted Akt1 in murine primary granulosa cells and assessed the molecular consequences at both the transcript and protein levels. We were able to corroborate the involvement of AKT1 in the regulation of metabolism, apoptosis, cell cycle, or cytoskeleton dynamics in this ovarian cell type. Consistently, we showed in established granulosa cells that depletion of Akt1 provoked altered directional persistent migration and increased its velocity. This study also allowed us to put forward new direct and indirect targets of the kinase. Indeed, a series of proteins involved in intracellular transport and mitochondrial physiology were significantly affected by Akt1 depletion. Using in silico analyses, we also propose a set of kinases and transcription factors that can mediate the action of AKT1 on the deregulated transcripts and proteins. Taken altogether, our results provide a resource of direct and indirect AKT1 targets in granulosa cells and may help understand its roles in this ovarian cell type.
Collapse
Affiliation(s)
- Maëva Elzaiat
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France
| | - Laetitia Herman
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France
| | - Bérangère Legois
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France
| | - Thibaut Léger
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France
| | - Anne-Laure Todeschini
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France.
| | - Reiner A Veitia
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France.
| |
Collapse
|
42
|
Szweda M, Rychlik A, Babińska I, Pomianowski A. Significance of Cyclooxygenase-2 in Oncogenesis. J Vet Res 2019; 63:215-224. [PMID: 31276061 PMCID: PMC6598184 DOI: 10.2478/jvetres-2019-0030] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/23/2019] [Indexed: 12/25/2022] Open
Abstract
Abstract
The cyclooxygenase-2 (COX-2) enzyme catalyses the first stage of biosynthesis of prostanoids, proteins that are implicated in various physiological and pathological processes in humans and animals. The expression of COX-2 increases significantly during pathological processes accompanied by inflammation, pain and fever. Overexpression of COX-2 was determined in tumour tissues, which suggests that this enzyme participates in oncogenesis. In this paper the topics discussed are mechanisms regulating COX-2 expression, COX isoforms, their role in the body and the oncogenic mechanisms triggered by the overexpression of COX-2, including inhibition of apoptosis, intensification of neoangiogenesis, increased metastatic capacity, and weakening of the immune system. The significance of and the mechanisms by which COX-2 participates in oncogenesis have been studied intensively in recent years. The results are highly promising, and they expand our understanding of the complex processes and changes at the molecular, cellular and tissue level that promote oncogenesis and cancer progression. Notwithstanding the knowledge already gleaned, many processes and mechanisms have not yet been elucidated in human medicine and, in particular, in veterinary medicine. Further research is required to develop effective tumour diagnostic methods and treatment procedures for humans and animals.
Collapse
Affiliation(s)
- Marta Szweda
- Department of Internal Diseases with Clinic, 10-719Olsztyn, Poland
| | | | - Izabella Babińska
- Department of Pathophysiology, Forensic Medicine, and Administration Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, 10-719Olsztyn, Poland
| | | |
Collapse
|
43
|
Mierke CT. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2019; 82:064602. [PMID: 30947151 DOI: 10.1088/1361-6633/ab1628] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The minimal structural unit of a solid tumor is a single cell or a cellular compartment such as the nucleus. A closer look inside the cells reveals that there are functional compartments or even structural domains determining the overall properties of a cell such as the mechanical phenotype. The mechanical interaction of these living cells leads to the complex organization such as compartments, tissues and organs of organisms including mammals. In contrast to passive non-living materials, living cells actively respond to the mechanical perturbations occurring in their microenvironment during diseases such as fibrosis and cancer. The transformation of single cancer cells in highly aggressive and hence malignant cancer cells during malignant cancer progression encompasses the basement membrane crossing, the invasion of connective tissue, the stroma microenvironments and transbarrier migration, which all require the immediate interaction of the aggressive and invasive cancer cells with the surrounding extracellular matrix environment including normal embedded neighboring cells. All these steps of the metastatic pathway seem to involve mechanical interactions between cancer cells and their microenvironment. The pathology of cancer due to a broad heterogeneity of cancer types is still not fully understood. Hence it is necessary to reveal the signaling pathways such as mechanotransduction pathways that seem to be commonly involved in the development and establishment of the metastatic and mechanical phenotype in several carcinoma cells. We still do not know whether there exist distinct metastatic genes regulating the progression of tumors. These metastatic genes may then be activated either during the progression of cancer by themselves on their migration path or in earlier stages of oncogenesis through activated oncogenes or inactivated tumor suppressor genes, both of which promote the metastatic phenotype. In more detail, the adhesion of cancer cells to their surrounding stroma induces the generation of intracellular contraction forces that deform their microenvironments by alignment of fibers. The amplitude of these forces can adapt to the mechanical properties of the microenvironment. Moreover, the adhesion strength of cancer cells seems to determine whether a cancer cell is able to migrate through connective tissue or across barriers such as the basement membrane or endothelial cell linings of blood or lymph vessels in order to metastasize. In turn, exposure of adherent cancer cells to physical forces, such as shear flow in vessels or compression forces around tumors, reinforces cell adhesion, regulates cell contractility and restructures the ordering of the local stroma matrix that leads subsequently to secretion of crosslinking proteins or matrix degrading enzymes. Hence invasive cancer cells alter the mechanical properties of their microenvironment. From a mechanobiological point-of-view, the recognized physical signals are transduced into biochemical signaling events that guide cellular responses such as cancer progression after the malignant transition of cancer cells from an epithelial and non-motile phenotype to a mesenchymal and motile (invasive) phenotype providing cellular motility. This transition can also be described as the physical attempt to relate this cancer cell transitional behavior to a T1 phase transition such as the jamming to unjamming transition. During the invasion of cancer cells, cell adaptation occurs to mechanical alterations of the local stroma, such as enhanced stroma upon fibrosis, and therefore we need to uncover underlying mechano-coupling and mechano-regulating functional processes that reinforce the invasion of cancer cells. Moreover, these mechanisms may also be responsible for the awakening of dormant residual cancer cells within the microenvironment. Physicists were initially tempted to consider the steps of the cancer metastasis cascade as single events caused by a single mechanical alteration of the overall properties of the cancer cell. However, this general and simple view has been challenged by the finding that several mechanical properties of cancer cells and their microenvironment influence each other and continuously contribute to tumor growth and cancer progression. In addition, basement membrane crossing, cell invasion and transbarrier migration during cancer progression is explained in physical terms by applying physical principles on living cells regardless of their complexity and individual differences of cancer types. As a novel approach, the impact of the individual microenvironment surrounding cancer cells is also included. Moreover, new theories and models are still needed to understand why certain cancers are malignant and aggressive, while others stay still benign. However, due to the broad variety of cancer types, there may be various pathways solely suitable for specific cancer types and distinct steps in the process of cancer progression. In this review, physical concepts and hypotheses of cancer initiation and progression including cancer cell basement membrane crossing, invasion and transbarrier migration are presented and discussed from a biophysical point-of-view. In addition, the crosstalk between cancer cells and a chronically altered microenvironment, such as fibrosis, is discussed including the basic physical concepts of fibrosis and the cellular responses to mechanical stress caused by the mechanically altered microenvironment. Here, is highlighted how biophysical approaches, both experimentally and theoretically, have an impact on classical hallmarks of cancer and fibrosis and how they contribute to the understanding of the regulation of cancer and its progression by sensing and responding to the physical environmental properties through mechanotransduction processes. Finally, this review discusses various physical models of cell migration such as blebbing, nuclear piston, protrusive force and unjamming transition migration modes and how they contribute to cancer progression. Moreover, these cellular migration modes are influenced by microenvironmental perturbances such as fibrosis that can induce mechanical alterations in cancer cells, which in turn may impact the environment. Hence, the classical hallmarks of cancer need to be refined by including biomechanical properties of cells, cell clusters and tissues and their microenvironment to understand mechano-regulatory processes within cancer cells and the entire organism.
Collapse
|
44
|
Teng P, Liu Y, Dai Y, Zhang H, Liu WT, Hu J. Nicotine Attenuates Osteoarthritis Pain and Matrix Metalloproteinase-9 Expression via the α7 Nicotinic Acetylcholine Receptor. THE JOURNAL OF IMMUNOLOGY 2019; 203:485-492. [PMID: 31152077 DOI: 10.4049/jimmunol.1801513] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 05/08/2019] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that causes chronic disability among the elderly. Despite recent advances in symptomatic management of OA by pharmacological and surgical approaches, there remains a lack of optimal approaches to manage inflammation in the joints, which causes cartilage degradation and pain. In this study, we investigated the efficacy and underlying mechanisms of nicotine exposure in attenuating joint inflammation, cartilage degradation, and pain in a mouse model of OA. A mouse model of OA was induced by injection of monosodium iodoacetate into the knee joint. Cell culture models were also used to study the efficacy and underlying mechanisms of nicotine treatment in attenuating symptoms of OA. Nicotine treatment reduced mechanical allodynia, cartilage degradation, and the upregulation of matrix metalloproteinase-9 (MMP-9), a hallmark of joint inflammation in OA, in mice treated with monosodium iodoacetate. The effects of nicotine were abolished by the selective α7 nicotinic acetylcholine receptor (nAChR) blocker, methyllycaconitine . In RAW264.7 cells and murine primary bone marrow-derived macrophages, nicotine significantly inhibited MMP-9 production induced by LPS. In addition, nicotine significantly enhanced PI3K/Akt and inhibited NF-κB translocation from the cytosol to the nucleus in an α7-nAChR-dependent manner, suggesting that nicotine acts on α7-nAChRs to inhibit MMP-9 production by macrophages through modulation of the PI3K/Akt-NF-κB pathway. Our results provide novel evidence that nicotine can attenuate joint inflammation and pain in experimental OA via α7-nAChRs. α7-nAChR could thus serve as a highly promising target to manage joint inflammation and pain in OA.
Collapse
Affiliation(s)
- Peng Teng
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - Yuan Liu
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; and
| | - Yan Dai
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; and
| | - Haijun Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China; .,Department of Pharmacy, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Jun Hu
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| |
Collapse
|
45
|
Zhang S, Song Q, Wang X, Wei Z, Yu R, Wang X, Jiang T. Virtual Screening Guided Design, Synthesis and Bioactivity Study of Benzisoselenazolones (BISAs) on Inhibition of c-Met and Its Downstream Signalling Pathways. Int J Mol Sci 2019; 20:E2489. [PMID: 31137515 PMCID: PMC6566228 DOI: 10.3390/ijms20102489] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
Abstract
c-Met is a transmembrane receptor tyrosine kinase and an important therapeutic target for anticancer drugs. In this study, we designed a small library containing 300 BISAs molecules that consisted of carbohydrates, amino acids, isothiourea, tetramethylthiourea, guanidine and heterocyclic groups and screened c-Met targeting compounds using docking and MM/GBSA. Guided by virtual screening, we synthesised a series of novel compounds and their activity on inhibition of the autophosphorylation of c-Met and its downstream signalling pathway proteins were evaluated. We found a panel of benzisoselenazolones (BISAs) obtained by introducing isothiourea, tetramethylthiourea and heterocyclic groups into the C-ring of Ebselen, including 7a, 7b, 8a, 8b and 12c (with IC50 values of less than 20 μM in MET gene amplified lung cancer cell line EBC-1), exhibited more potent antitumour activity than Ebselen by cell growth assay combined with in vitro biochemical assays. In addition, we also tested the antitumour activity of three cancer cell lines without MET gene amplification/activation, including DLD1, MDA-MB-231 and A549. The neuroblastoma SK-N-SH cells with HGF overexpression which activates MET signalling are sensitive to MET inhibitors. The results reveal that our compounds may be nonspecific multitarget kinase inhibitors, just like type-II small molecule inhibitors. Western blot analysis showed that these inhibitors inhibited autophosphorylation of c-MET, and its downstream signalling pathways, such as PI3K/AKT and MARK/ERK. Results suggest that bensoisoselenones can be used as a scaffold for the design of c-Met inhibiting drug leads, and this study opens up new possibilities for future antitumour drug design.
Collapse
Affiliation(s)
- Siqi Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| | - Qiaoling Song
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Center for Innovative Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China.
| | - Xueting Wang
- Center for High Performance Computing & System simulation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China.
| | - Zhiqiang Wei
- Center for High Performance Computing & System simulation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China.
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Center for Innovative Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.
| | - Xin Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Center for Innovative Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
- Center for Innovative Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China.
| |
Collapse
|
46
|
Lin CC, Chen KB, Tsai CH, Tsai FJ, Huang CY, Tang CH, Yang JS, Hsu YM, Peng SF, Chung JG. Casticin inhibits human prostate cancer DU 145 cell migration and invasion via Ras/Akt/NF-κB signaling pathways. J Food Biochem 2019; 43:e12902. [PMID: 31353708 DOI: 10.1111/jfbc.12902] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/13/2019] [Accepted: 05/04/2019] [Indexed: 12/29/2022]
Abstract
Casticin, a polymethoxyflavone derived from natural plants, has biological activities including induction of cell apoptosis. In this study, we showed the beneficial effects of casticin on the inhibition of prostate cancer cell metastasis. Casticin reduced total viable cell number, thus, we selected low doses of casticin for following experiments. Casticin decreased cell mobility, suppressed cell migration and invasion, and reduced cell gelatinolytic activities of MMP-2/-9. Furthermore, casticin inhibited the protein levels of AKT, GSK3 αβ, Snail, and MMPs (MMP-2, -9, -13, and -7) at 24 and 48 hr treatment. Casticin diminished the expressions of NF-κB p65, GRB2, SOS-1, MEK, p-ERK1/2, and p-JNK1/2 at 48 hr treatment only. However, casticin reduced the level of E-cadherin at 24 hr treatment but elevated at 48 hr. The novel findings suggest that casticin may represent a new and promising therapeutic agent for the metastatic prostate cancer. PRACTICAL APPLICATIONS: Casticin derived from natural plants had been used for Chinese medicine in Chinese population for thousands of years. In the present study, casticin attenuated metastatic effects, including decreasing viable cell number, inhibiting the migration, invasion, and adhesion, and reducing matrix metalloproteinases activity on human prostate DU 145 cancer cells. In addition, the results also provided possible pathways involved in casticin anti-metastasis mechanism. We conclude that casticin may be an aptitude anticancer agent or adjuvant for the metastatic prostate cancer in the future.
Collapse
Affiliation(s)
- Chia-Chang Lin
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Kuen-Bao Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan.,School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chang-Hai Tsai
- China Medical University Children's Hospital, China Medical University, Taichung, Taiwan.,Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- China Medical University Children's Hospital, China Medical University, Taichung, Taiwan.,School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
47
|
Aras S, Maroun MC, Song Y, Bandyopadhyay S, Stark A, Yang ZQ, Long MP, Grossman LI, Fernández-Madrid F. Mitochondrial autoimmunity and MNRR1 in breast carcinogenesis. BMC Cancer 2019; 19:411. [PMID: 31046734 PMCID: PMC6498478 DOI: 10.1186/s12885-019-5575-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
Background Autoantibodies function as markers of tumorigenesis and have been proposed to enhance early detection of malignancies. We recently reported, using immunoscreening of a T7 complementary DNA (cDNA) library of breast cancer (BC) proteins with sera from patients with BC, the presence of autoantibodies targeting several mitochondrial DNA (mtDNA)-encoded subunits of the electron transport chain (ETC) in complexes I, IV, and V. Methods In this study, we have characterized the role of Mitochondrial-Nuclear Retrograde Regulator 1 (MNRR1, also known as CHCHD2), identified on immunoscreening, in breast carcinogenesis. We assessed the protein as well as transcript levels of MNRR1 in BC tissues and in derived cell lines representing tumors of graded aggressiveness. Mitochondrial function was also assayed and correlated with the levels of MNRR1. We studied the invasiveness of BC derived cells and the effect of MNRR1 levels on expression of genes associated with cell proliferation and migration such as Rictor and PGC-1α. Finally, we manipulated levels of MNRR1 to assess its effect on mitochondria and on some properties linked to a metastatic phenotype. Results We identified a nuclear DNA (nDNA)-encoded mitochondrial protein, MNRR1, that was significantly associated with the diagnosis of invasive ductal carcinoma (IDC) of the breast by autoantigen microarray analysis. In focusing on the mechanism of action of MNRR1 we found that its level was nearly twice as high in malignant versus benign breast tissue and up to 18 times as high in BC cell lines compared to MCF10A control cells, suggesting a relationship to aggressive potential. Furthermore, MNRR1 affected levels of multiple genes previously associated with cancer metastasis. Conclusions MNRR1 regulates multiple genes that function in cell migration and cancer metastasis and is higher in cell lines derived from aggressive tumors. Since MNRR1 was identified as an autoantigen in breast carcinogenesis, the present data support our proposal that both mitochondrial autoimmunity and MNRR1 activity in particular are involved in breast carcinogenesis. Virtually all other nuclear encoded genes identified on immunoscreening of invasive BC harbor an MNRR1 binding site in their promoters, thereby placing MNRR1 upstream and potentially making it a novel marker for BC metastasis.
Collapse
Affiliation(s)
- Siddhesh Aras
- Wayne State University School of Medicine, Center for Molecular Medicine and Genetics, 540 E. Canfield Ave, Detroit, MI, 48201, USA
| | - Marie-Claire Maroun
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA.,Division of Rheumatology, Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Yeohan Song
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA
| | | | - Azadeh Stark
- Department of Pathology, Henry Ford Health System, Detroit, MI, 48201, USA
| | - Zeng-Quan Yang
- Department of Oncology and Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Michael P Long
- Department of Pathology, Wayne State University, Detroit, MI, 48201, USA
| | - Lawrence I Grossman
- Wayne State University School of Medicine, Center for Molecular Medicine and Genetics, 540 E. Canfield Ave, Detroit, MI, 48201, USA.
| | - Félix Fernández-Madrid
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Division of Rheumatology, Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Wayne State University, University Health Center, 4H, 4201 St. Antoine, Detroit, MI, 48201, USA.
| |
Collapse
|
48
|
Chen JK, Peng SF, Lai KC, Liu HC, Huang YP, Lin CC, Huang AC, Chueh FS, Chung JG. Fisetin Suppresses Human Osteosarcoma U-2 OS Cell Migration and Invasion via Affecting FAK, uPA and NF-ĸB Signaling Pathway In Vitro. In Vivo 2019; 33:801-810. [PMID: 31028200 PMCID: PMC6559886 DOI: 10.21873/invivo.11542] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIM Evidence has indicated that fisetin induces cytotoxic effects in human cancer cell lines, including the inhibition of cell migration and invasion, however, the exact molecular mechanism of action of fisetin in human osteosarcoma cells remains unclear. MATERIALS AND METHODS The anti-metastatic mechanisms of fisetin in human osteosarcoma U-2 OS cells were investigated in vitro. RESULTS Fisetin reduced the viability of cells at different concentrations (2.5, 5 and 10 μM) as measured by flow cytometric assay. Fisetin suppressed cell mobility, migration and invasion of U-2 OS cells, as shown by wound healing assay and transwell filter chambers, respectively. The gelatin zymography assay showed that fisetin inhibited MMP-2 activity in U-2 OS cells. Results from western blotting indicated that fisetin reduced the levels of pEGFR, SOS-1, GRB2, Ras, PKC, p-ERK1/2, p-JNK, p-p-38, VEGF, FAK, RhoA, PI3K, p-AKT, NF-ĸB, uPA, MMP-7, MMP-9, and MMP-13, but increased GSK3β and E-cadherin in U-2 OS cells after 48 h of treatment. CONCLUSION Fisetin can be used in the future, as a target for the treatment of metastasis of human osteosarcoma cells.
Collapse
Affiliation(s)
- Jr-Kai Chen
- Attending Physician of Orthopaedadics, Department of Chang Bing Show-Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Kuang Chi Lai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan, R.O.C
- Department of Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan, R.O.C
| | - Hsin-Chung Liu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Yi-Ping Huang
- Department of Physiology, College of Medicine, China Medical University, Taichung, Taiwan, R.O.C
| | - Chin-Chung Lin
- Department of Chinese Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Executive Yuan, Taichung, Taiwan, R.O.C
- General Education Center, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C
| | - An-Cheng Huang
- Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yilan, Taiwan, R.O.C
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan, R.O.C.
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
- Department of Biotechnology, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
49
|
Elzaiat M, Herman L, Legois B, Léger T, Todeschini AL, Veitia RA. High-throughput Exploration of the Network Dependent on AKT1 in Mouse Ovarian Granulosa Cells. Mol Cell Proteomics 2019. [PMID: 30992313 DOI: 10.1074/mcp.ra119.001461] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The PI3K/AKT signaling pathway is known to regulate a broad range of cellular processes, and it is often altered in several types of cancers. Recently, somatic AKT1 mutations leading to a strong activation of this kinase have been reported in juvenile granulosa cell tumors. However, the molecular role of AKT1 in the supporting cell lineage of the ovary is still poorly understood. To get insights into its function in such cells, we depleted Akt1 in murine primary granulosa cells and assessed the molecular consequences at both the transcript and protein levels. We were able to corroborate the involvement of AKT1 in the regulation of metabolism, apoptosis, cell cycle, or cytoskeleton dynamics in this ovarian cell type. Consistently, we showed in established granulosa cells that depletion of Akt1 provoked altered directional persistent migration and increased its velocity. This study also allowed us to put forward new direct and indirect targets of the kinase. Indeed, a series of proteins involved in intracellular transport and mitochondrial physiology were significantly affected by Akt1 depletion. Using in silico analyses, we also propose a set of kinases and transcription factors that can mediate the action of AKT1 on the deregulated transcripts and proteins. Taken altogether, our results provide a resource of direct and indirect AKT1 targets in granulosa cells and may help understand its roles in this ovarian cell type.
Collapse
Affiliation(s)
- Maëva Elzaiat
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France
| | - Laetitia Herman
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France
| | - Bérangère Legois
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France
| | - Thibaut Léger
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France
| | - Anne-Laure Todeschini
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France.
| | - Reiner A Veitia
- From the ‡Institut Jacques Monod, Université Paris-Diderot, 75013 Paris, France;; §Université Paris-Diderot, 75013 Paris, France.
| |
Collapse
|
50
|
Fadhlullah SFB, Halim NBA, Yeo JYT, Ho RLY, Um P, Ang BT, Tang C, Ng WH, Virshup DM, Ho IAW. Pathogenic mutations in neurofibromin identifies a leucine-rich domain regulating glioma cell invasiveness. Oncogene 2019; 38:5367-5380. [PMID: 30967630 PMCID: PMC6755990 DOI: 10.1038/s41388-019-0809-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is the most aggressive tumor of the brain. NF1, a tumor suppressor gene and RAS-GTPase, is one of the highly mutated genes in GBM. Dysregulated NF1 expression promotes cell invasion, proliferation, and tumorigenesis. Loss of NF1 expression in glioblastoma is associated with increased aggressiveness of the tumor. Here, we show that NF1-loss in patient-derived glioma cells using shRNA increases self-renewal, heightens cell invasion, and promotes mesenchymal subtype and epithelial mesenchymal transition-specific gene expression that enhances tumorigenesis. The neurofibromin protein contains at least four major domains, with the GAP-related domain being the most well-studied. In this study, we report that the leucine-rich domain (LRD) of neurofibromin inhibits invasion of human glioblastoma cells without affecting their proliferation. Moreover, under conditions tested, the NF1-LRD fails to hydrolyze Ras-GTP to Ras-GDP, suggesting that its suppressive function is independent of Ras signaling. We further demonstrate that rare variants within the NF1-LRD domain found in a subset of the patients are pathogenic and reduce NF1-LRD’s invasion suppressive function. Taken together, our results show, for the first time, that NF1-LRD inhibits glioma invasion, and provides evidence of a previously unrecognized function of NF1-LRD in glioma biology.
Collapse
Affiliation(s)
- Siti Farah Bte Fadhlullah
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore.,Lucence Diagnostics Pte Ltd., Singapore, Singapore
| | | | - Jacqueline Y T Yeo
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Rachel L Y Ho
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Phoebe Um
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore.,University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Beng Ti Ang
- Department of Neurosurgery, National Neuroscience Institute, Singapore, 308433, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Carol Tang
- Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Division of Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore
| | - Wai H Ng
- Department of Neurosurgery, National Neuroscience Institute, Singapore, 308433, Singapore
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Department of Pediatrics, Duke University School of Medicine, Durham, NC, 27703, USA
| | - Ivy A W Ho
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore. .,Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|