1
|
Khiabani NA, Doustvandi MA, Story D, Nobari SA, Hajizadeh M, Petersen R, Dunbar G, Rossignol J. Glioblastoma therapy: State of the field and future prospects. Life Sci 2024; 359:123227. [PMID: 39537100 DOI: 10.1016/j.lfs.2024.123227] [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: 06/25/2024] [Revised: 09/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GB) is a cancerous brain tumor that originates from glial cells and leads to thousands of deaths each year and a five-year survival of only 6.8 %. Treatments for GB include surgery, chemotherapy, radiation, and immunotherapy. GB is an incurable fatal disease, necessitating the development of innovative strategies to find a developing effective therapy. Genetic therapies may be crucial in treating GB by identifying the mutations and amplifications of multiple genes, which drive its proliferation and spread. Use of small interfering RNAs (siRNAs) provides a novel technology used to suppress the genes associated with disease, which forms a basis for targeted therapy in GB and its stem cell population, which are recognized for their ability to develop resistance to chemotherapy and tumorigenic capabilities. This review examines the use of siRNAs in GB, emphasizing their effectiveness in suppressing key oncogenes and signaling pathways associated with tumor development, invasion, stemness, and resistance to standard treatments. siRNA-based gene silencing is a promising approach for developing targeted therapeutics against GB and associated stem cell populations, potentially enhancing patient outcomes and survival rates in this devastating disease.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | | | - Darren Story
- Department of Psychology, Saginaw Valley State University, University Center, MI 48710, USA
| | | | | | - Robert Petersen
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Gary Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, USA; Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, USA; College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
| |
Collapse
|
2
|
pan C, bai X, Li N, Zheng N, Si Y, Zhao Y. PBX3 as a biomarker for the early diagnosis and prediction of prognosis of glioma. PLoS One 2024; 19:e0293647. [PMID: 38324550 PMCID: PMC10849273 DOI: 10.1371/journal.pone.0293647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 10/17/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Increasing evidence have elucidated that PBX3 played a crucial role in cancer initiation and progression. PBX3 was differentially expressed in many cancer types. However, PBX3 potential involvement in gliomas remains to be explored. METHODS The expression level of PBX3 in glioma tissues and glioma cells, and its correlation with clinical features were analyzed by data from TCGA, GEPIA, CGGA and CCLE. Univariable survival and Multivariate Cox analysis was used to compare several clinical characteristics with survival. We also analyzed the correlation between PBX3 expression level and survival outcome and survival time of LGG and GBM patients by using linear regression equation. GSEA was used to generate an ordered list of all genes related to PBX3 expression and screening of genes co-expressed with PBX3 mRNA by "limma" package. RESULTS The results showed that PBX3 was highly expressed in gliomas and its expression increased with the increase of malignancy. Survival analysis found that PBX3 is more valuable in predicting the OS and PFI of LGG patients than that of GBM. For further study, TCGA and CGGA data were downloaded for univariate Cox analysis and multivariate Cox analysis which showed that the expression of PBX3 was independent influencing factors for poor prognosis of LGG patients. Meanwhile, Receiver operating characteristic (ROC) curve showed that PBX3 was a predictor of overall survival rate and progression-free survival rate of LGG. Linear regression model analysis indicated that the higher expression of PBX3 the higher the risk of death of LGG patients, and the higher expression of PBX3 the higher the risk of disease progression of LGG patients. Next, TCGA data were downloaded for GSEA and Co-expression analyses, which was performed to study the function of PBX3. CONCLUSION PBX3 may be involved in the occurrence and development of glioma, and has potential reference value for the early diagnosis and prediction of prognosis of glioma.
Collapse
Affiliation(s)
- Cuicui pan
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xueli bai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Na Li
- Department of Dermatology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ni Zheng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuanquan Si
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yueran Zhao
- Central Laboratory, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| |
Collapse
|
3
|
Pienkowski T, Kowalczyk T, Cysewski D, Kretowski A, Ciborowski M. Glioma and post-translational modifications: A complex relationship. Biochim Biophys Acta Rev Cancer 2023; 1878:189009. [PMID: 37913943 DOI: 10.1016/j.bbcan.2023.189009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 11/03/2023]
Abstract
Post-translational modifications (PTMs) are common covalent processes in biochemical pathways that alter protein function and activity. These modifications occur through proteolytic cleavage or attachment of modifying groups, such as phosphoryl, methyl, glycosyl, or acetyl groups, with one or more amino acid residues of a single protein. Some PTMs also present crosstalk abilities that affect both protein functionality and structure, creating new proteoforms. Any alteration in organism homeostasis may be a cancer hallmark. Cataloging PTMs and consequently, emerging proteoforms, present new therapeutic targets, approaches, and opportunities to discover additional discriminatory biomarkers in disease diagnostics. In this review, we focus on experimentally confirmed PTMs and their potential crosstalk in glioma research to introduce new opportunities for this tumor type, which emerge within the PTMomics area.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Tomasz Kowalczyk
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Dominik Cysewski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland.
| |
Collapse
|
4
|
Agrawal K, Chauhan S, Kumar D. Expression analysis and regulation of GLI and its correlation with stemness and metabolic alteration in human brain tumor. 3 Biotech 2023; 13:10. [PMID: 36532860 PMCID: PMC9755437 DOI: 10.1007/s13205-022-03419-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022] Open
Abstract
GLI gene-mediated hedgehog (Hh) signaling pathway plays a substantial role in brain cancer development and growth including glioblastoma multiforme (GBM), lower-grade glioma (LGG), and medulloblastoma (MB). GLI2 and GLI3 gene expression levels are extremely enhanced in these cancers with poor patient survival. Moreover, GLI genes are correlated with stemness-related factors SOX2, SOX9, POU5F1, and NANOG that work as the driving factors for brain cancer stem cells (CSCs) progression. It's critical to find new ways to combat this deadly malignancy and CSCs. Using in silico approaches, our study explored the role of GLI genes (GLI1, GLI2, and GLI3), the primary transcription factors of the sonic hedgehog (SHH) signaling pathway, in GBM, LGG, MB, and glioblastoma stem-like cells (GSCs). Additionally, we found strong association of angiogenic-related gene VEGFA, metabolic genes ENO1, ENO2, and pluripotency-related genes SOX2, SOX9, NANOG, POU5F1 with GLI genes, suggesting their role in brain tumor initiation and progression. We also studied their transcriptional network and functional category enrichment analysis about brain tumor development to find a better therapeutic strategy against brain cancer and their stem cells. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03419-5.
Collapse
Affiliation(s)
- Kirti Agrawal
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, Uttarakhand 248007 India
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sec 125, Noida, 201303 India
| | - Saumya Chauhan
- Amity Global School, Sector 46, Gurugram, Haryana 122018 India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, Uttarakhand 248007 India
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sec 125, Noida, 201303 India
| |
Collapse
|
5
|
Lv S, Chen Z, Mi H, Yu X. Cofilin Acts as a Booster for Progression of Malignant Tumors Represented by Glioma. Cancer Manag Res 2022; 14:3245-3269. [PMID: 36452435 PMCID: PMC9703913 DOI: 10.2147/cmar.s389825] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/10/2022] [Indexed: 07/20/2023] Open
Abstract
Cofilin, as a depolymerization factor of actin filaments, has been widely studied. Evidences show that cofilin has a role in actin structural reorganization and dynamic regulation. In recent years, several studies have demonstrated a regulatory role for cofilin in the migration and invasion mediated by cell dynamics and epithelial to mesenchymal transition (EMT)/EMT-like process, apoptosis, radiotherapy resistance, immune escape, and transcriptional dysregulation of malignant tumor cells, particularly glioma cells. On this basis, it is practical to evaluate cofilin as a biomarker for predicting tumor metastasis and prognosis. Targeting cofilin regulating kinases, Lin11, Isl-1 and Mec-3 kinases (LIM kinases/LIMKs) and their major upstream molecules inhibits tumor cell migration and invasion and targeting cofilin-mediated mitochondrial pathway induces apoptosis of tumor cells represent effective options for the development of novel anti-malignant tumor drug, especially anti-glioma drugs. This review explores the structure, general biological function, and regulation of cofilin, with an emphasis on the critical functions and prospects for clinical therapeutic applications of cofilin in malignant tumors represented by glioma.
Collapse
Affiliation(s)
- Shihong Lv
- Department of Gastroenterology, The Second Affiliated Hospital of Mudanjiang Medical College, Mudanjiang Medical College, Mudanjiang, 157011, People’s Republic of China
| | - Zhiye Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Hailong Mi
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xingjiang Yu
- Department of Histology and Embryology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
6
|
Guo X, Jiao H, Cao L, Meng F. Biological implications and clinical potential of invasion and migration related miRNAs in glioma. Front Integr Neurosci 2022; 16:989029. [PMID: 36479040 PMCID: PMC9720134 DOI: 10.3389/fnint.2022.989029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/07/2022] [Indexed: 12/01/2024] Open
Abstract
Gliomas are the most common primary malignant brain tumors and are highly aggressive. Invasion and migration are the main causes of poor prognosis and treatment resistance in gliomas. As migration and invasion occur, patient survival and prognosis decline dramatically. MicroRNAs (miRNAs) are small, non-coding 21-23 nucleotides involved in regulating the malignant phenotype of gliomas, including migration and invasion. Numerous studies have demonstrated the mechanism and function of some miRNAs in glioma migration and invasion. However, the biological and clinical significance (including diagnosis, prognosis, and targeted therapy) of glioma migration and invasion-related miRNAs have not been systematically discussed. This paper reviews the progress of miRNAs-mediated migration and invasion studies in glioma and discusses the clinical value of migration and invasion-related miRNAs as potential biomarkers or targeted therapies for glioma. In addition, these findings are expected to translate into future directions and challenges for clinical applications. Although many biomarkers and their biological roles in glioma invasion and migration have been identified, none have been specific so far, and further exploration of clinical treatment is still in progress; therefore, we aimed to further identify specific markers that may guide clinical treatment and improve the quality of patient survival.
Collapse
Affiliation(s)
| | | | | | - Facai Meng
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
7
|
Yan L, He Z, Li W, Liu N, Gao S. P76RBE silencing inhibits ovarian cancer cell proliferation, migration, and invasion via suppressing the integrin β1/NF-κB pathway. Cell Cycle 2021; 20:1875-1889. [PMID: 34382920 DOI: 10.1080/15384101.2021.1963910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Rhophilin Rho GTPase binding protein 2 (P76RBE) belongs to rhophilin family of Rho-GTPase-binding proteins and is found to contribute to the development of diverse cancers. Data in Oncomine and Kaplan-Meier Plotter databases showed that P76RBE was upregulated in ovarian cancer tissues compared with normal tissues, and patients with high P76RBE expression had worse overall survival, which indicated P76RBE may be associated with the pathogenesis of ovarian cancer. This study aimed to investigate the role of P76RBE in ovarian cancer and to reveal the possible underlying mechanisms. The results demonstrated that P76RBE was highly expressed in ovarian cancer tissues and ovarian cancer cell lines. Functionally, silencing of P76RBE suppressed the proliferation, induced cell cycle arrest, and inhibited migration and invasion in OVCAR-3 and OV-90 cells, while overexpression of P76RBE showed opposite effects on A2780 cells. Mechanically, P76RBE silencing resulted in downregulation of integrin β1, accompanying the reduced NF-κB p65 phosphorylation and nuclear translocation. Importantly, integrin β1 knockdown effectively rescued the effects of P76RBE overexpression on ovarian cancer cells with suppressed proliferation, migration, and invasion. Additionally, in the xenograft tumors derived from OVCAR-3 and OV-90 cell lines, P76RBE knockdown inhibited tumor growth. Meanwhile, the expression of integrin β1 and NF-κB p65 phosphorylation was decreased. In summary, our findings indicate that P76RBE contributes to the progression of ovarian cancer through regulating the integrin β1/NF-κB signaling, and it may be a promising target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Limei Yan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zeping He
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ning Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Song Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
8
|
Levenga J, Wong H, Milstead R, LaPlante L, Hoeffer CA. Immunohistological Examination of AKT Isoforms in the Brain: Cell-Type Specificity That May Underlie AKT's Role in Complex Brain Disorders and Neurological Disease. Cereb Cortex Commun 2021; 2:tgab036. [PMID: 34296180 PMCID: PMC8223503 DOI: 10.1093/texcom/tgab036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
Protein kinase B (PKB/AKT) is a central kinase involved in many neurobiological processes. AKT is expressed in the brain as three isoforms, AKT1, AKT2, and AKT3. Previous studies suggest isoform-specific roles in neural function, but very few studies have examined AKT isoform expression at the cellular level. In this study, we use a combination of histology, immunostaining, and genetics to characterize cell-type-specific expression of AKT isoforms in human and mouse brains. In mice, we find that AKT1 is the most broadly expressed isoform, with expression in excitatory neurons and the sole detectable AKT isoform in gamma-aminobutyric acid ergic interneurons and microglia. By contrast, we find that AKT2 is the sole isoform expressed in astroglia and is not detected in other neural cell types. We find that AKT3 is expressed in excitatory neurons with AKT1 but shows greater expression levels in dendritic compartments than AKT1. We extend our analysis to human brain tissues and find similar results. Using genetic deletion approaches, we also find that the cellular determinants restricting AKT isoform expression to specific cell types remain intact under Akt deficiency conditions. Because AKT signaling is linked to numerous neurological disorders, a greater understanding of cell-specific isoform expression could improve treatment strategies involving AKT.
Collapse
Affiliation(s)
- Josien Levenga
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Helen Wong
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Ryan Milstead
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA.,Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Lauren LaPlante
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Charles A Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO 80303, USA.,Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80303, USA.,Linda Crnic Institute, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Whitehouse JP, Howlett M, Hii H, Mayoh C, Wong M, Barahona P, Ajuyah P, White CL, Buntine MK, Dyke JM, Lee S, Valvi S, Stanley J, Andradas C, Carline B, Kuchibhotla M, Ekert PG, Cowley MJ, Gottardo NG, Endersby R. A Novel Orthotopic Patient-Derived Xenograft Model of Radiation-Induced Glioma Following Medulloblastoma. Cancers (Basel) 2020; 12:cancers12102937. [PMID: 33053751 PMCID: PMC7600047 DOI: 10.3390/cancers12102937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Radiation-induced glioma (RIG) is a highly aggressive brain cancer arising as a consequence of radiation therapy. We report a case of RIG that arose in the brain stem following treatment for paediatric medulloblastoma, and the development and characterisation of a matched orthotopic patient-derived xenograft (PDX) model (TK-RIG915). Patient and PDX tumours were analysed using DNA methylation profiling, whole genome sequencing (WGS) and RNA sequencing. While initially thought to be a diffuse intrinsic pontine glioma (DIPG) based on disease location, results from methylation profiling and WGS were not consistent with this diagnosis. Furthermore, clustering analyses based on RNA expression suggested the tumours were distinct from primary DIPG. Additional gene expression analysis demonstrated concordance with a published RIG expression profile. Multiple genetic alterations that enhance PI3K/AKT and Ras/Raf/MEK/ERK signalling were discovered in TK-RIG915 including an activating mutation in PIK3CA, upregulation of PDGFRA and AKT2, inactivating mutations in NF1, and a gain-of-function mutation in PTPN11. Additionally, deletion of CDKN2A/B, increased IDH1 expression, and decreased ARID1A expression were observed. Detection of phosphorylated S6, 4EBP1 and ERK via immunohistochemistry confirmed PI3K pathway and ERK activation. Here, we report one of the first PDX models for RIG, which recapitulates the patient disease and is molecularly distinct from primary brain stem glioma. Genetic interrogation of this model has enabled the identification of potential therapeutic vulnerabilities in this currently incurable disease.
Collapse
Affiliation(s)
- Jacqueline P. Whitehouse
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Meegan Howlett
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Hilary Hii
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Chelsea Mayoh
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Marie Wong
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Paulette Barahona
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
| | - Pamela Ajuyah
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
| | - Christine L. White
- Genetics and Molecular Pathology Laboratory, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; (C.L.W.); (M.K.B.)
- Department of Molecular and Translational Science, Monash University, Melbourne 3168, Victoria, Australia
| | - Molly K. Buntine
- Genetics and Molecular Pathology Laboratory, Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia; (C.L.W.); (M.K.B.)
- Department of Molecular and Translational Science, Monash University, Melbourne 3168, Victoria, Australia
| | - Jason M. Dyke
- Department of Neuropathology, PathWest Laboratory Medicine, Royal Perth Hospital, Perth 6000, Australia;
- Pathology and Laboratory Medicine, University of Western Australia, Nedlands 6009, Australia
| | - Sharon Lee
- Department of Neurosurgery, Perth Children’s Hospital, Nedlands 6009, Australia;
| | - Santosh Valvi
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children’s Hospital, Nedlands 6009, Australia
- Division of Paediatrics, University of Western Australia Medical School, Nedlands 6009, Australia
| | - Jason Stanley
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Clara Andradas
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
| | - Brooke Carline
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Mani Kuchibhotla
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
| | - Paul G. Ekert
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville 3052, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne 3000, Victoria, Australia
| | - Mark J. Cowley
- Children’s Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington 2033, Australia; (C.M.); (M.W.); (P.B.); (P.A.); (P.G.E.); (M.J.C.)
- School of Women’s and Children’s Health, UNSW Sydney, Kensington 2033, Australia
| | - Nicholas G. Gottardo
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
- Department of Paediatric and Adolescent Oncology/Haematology, Perth Children’s Hospital, Nedlands 6009, Australia
| | - Raelene Endersby
- Brain Tumour Research Program, Telethon Kids Institute, Nedlands 6009, Australia; (J.P.W.); (M.H.); (H.H.); (S.V.); (J.S.); (C.A.); (B.C.); (M.K.); (N.G.G.)
- Centre for Child Health Research, University of Western Australia, Nedlands 6009, Australia
- Correspondence:
| |
Collapse
|
10
|
Li Y, Lu S, Lan M, Peng X, Zhang Z, Lang J. A prognostic nomogram integrating novel biomarkers identified by machine learning for cervical squamous cell carcinoma. J Transl Med 2020; 18:223. [PMID: 32503630 PMCID: PMC7275455 DOI: 10.1186/s12967-020-02387-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023] Open
Abstract
Background Cervical cancer (CC) represents the fourth most frequently diagnosed malignancy affecting women all over the world. However, effective prognostic biomarkers are still limited for accurately identifying high-risk patients. Here, we provided a combination machine learning algorithm-based signature to predict the prognosis of cervical squamous cell carcinoma (CSCC). Methods and materials After utilizing RNA sequencing (RNA-seq) data from 36 formalin-fixed and paraffin-embedded (FFPE) samples, the most significant modules were highlighted by the weighted gene co-expression network analysis (WGCNA). A candidate genes-based prognostic classifier was constructed by the least absolute shrinkage and selection operator (LASSO) and then validated in an independent validation set. Finally, based on the multivariate analysis, a nomogram including the FIGO stage, therapy outcome, and risk score level was built to predict progression-free survival (PFS) probability. Results A mRNA-based signature was developed to classify patients into high- and low-risk groups with significantly different PFS and overall survival (OS) rate (training set: p < 0.001 for PFS, p = 0.016 for OS; validation set: p = 0.002 for PFS, p = 0.028 for OS). The prognostic classifier was an independent and powerful prognostic biomarker for PFS in both cohorts (training set: hazard ratio [HR] = 0.13, 95% CI 0.05–0.33, p < 0.001; validation set: HR = 0.02, 95% CI 0.01–0.04, p < 0.001). A nomogram that integrated the independent prognostic factors was constructed for clinical application. The calibration curve showed that the nomogram was able to predict 1-, 3-, and 5-year PFS accurately, and it performed well in the external validation cohorts (concordance index: 0.828 and 0.864, respectively). Conclusion The mRNA-based biomarker is a powerful and independent prognostic factor. Furthermore, the nomogram comprising our prognostic classifier is a promising predictor in identifying the progression risk of CSCC patients.
Collapse
Affiliation(s)
- Yimin Li
- School of Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Avenue, High-tech Zone (West District), Chengdu, 611731, Sichuan, People's Republic of China
| | - Shun Lu
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China.,Radiation Oncology Key Laboratory of Sichuan Province, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China
| | - Mei Lan
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xinhao Peng
- School of Medicine, University of Electronic Science and Technology of China, No. 2006, Xiyuan Avenue, High-tech Zone (West District), Chengdu, 611731, Sichuan, People's Republic of China
| | - Zijian Zhang
- Department of Oncology, Xiangya Hospital Central South University, Kaifu District, Changsha, 410008, Hunan, People's Republic of China
| | - Jinyi Lang
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China. .,Radiation Oncology Key Laboratory of Sichuan Province, No. 55, South Renmin Avenue Fourth Section, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
11
|
Xiao Y, Zhu Z, Li J, Yao J, Jiang H, Ran R, Li X, Li Z. Expression and prognostic value of long non-coding RNA H19 in glioma via integrated bioinformatics analyses. Aging (Albany NY) 2020; 12:3407-3430. [PMID: 32081833 PMCID: PMC7066912 DOI: 10.18632/aging.102819] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022]
Abstract
Numerous discoveries have elucidated that long noncoding RNAs (lncRNAs) play a critical role in cancer malignant progression. However, their potential involvement in gliomas remains to be explored. Herein, the expression level of lncRNA H19 in glioma tissues, and its relevance with clinical characteristics were analyzed through Oncomine. The results showed that H19 was highly expressed in glioma tissues and its expression increased with the increase of malignancy. Next, GTEx and TCGA data were downloaded for differently expressed genes (DEGs) identification, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and the correlation analyses between H19 expression and clinic features. Radiation therapy had a good effect on glioblastoma multiforme (GBM), but didn't have a good effect on low grade glioma (LGG). Meanwhile, the expression level of H19 could act as an indicator molecule indicating the effect of radiotherapy. Finally, gene set enrichment analysis (GSEA) and immune infiltration analysis were conducted. It was found that H19 could affect the immune infiltration level of glioma through copy number variations, thus affecting the prognosis of glioma patients. Collectively, H19 may be involved in the occurrence and development of glioma, and has potential reference value for the relief and immunotherapy of glioma.
Collapse
Affiliation(s)
- Yilei Xiao
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Zipeng Zhu
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Jianxiong Li
- Department of Chemotherapy, Chinese PLA General Hospital, Beijing 100036, P.R. China
| | - Jie Yao
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, P.R. China
| | - Haitao Jiang
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Ran Ran
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Xueyuan Li
- Department of Neurosurgery, Liaocheng People’s Hospital, Liaocheng 250000, Shandong Province, P.R. China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, P.R. China
| |
Collapse
|
12
|
Signaling Determinants of Glioma Cell Invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1202:129-149. [PMID: 32034712 DOI: 10.1007/978-3-030-30651-9_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
13
|
Liu Y, Li X, Jiang S, Ge Q. Inhibitory effect of Gypsophila oldhamiana gypsogenin on NCI-N87 gastric cancer cell line. EUR J INFLAMM 2018. [DOI: 10.1177/2058739218818958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Gastric cancer is one of the major cancers threatening people’s lives worldwide. Recent studies showed that Gypsophila oldhamiana gypsogenin (GOG) exhibits inhibition effects and cytotoxic activities against different cell lines. The aim of this study was to explore the inhibitory effect and dose response of GOG on gastric cancer cell line NCI-N87 and to provide the theoretical basis for clinical anti-tumor therapy. The experiments showed that GOG could inhibit the proliferation and promote the apoptosis of human gastric cancer cell line NCI-N87. GOG could dose dependently reduce the expression of vascular endothelial growth factor (VEGF) and matrix metalloprotein (MMP)-9 proteins, while increase the expression of caspase-3 and Bax proteins. Compared with model group, tumor volume (TV), relative tumor volume (RTV), and relative tumor increment rate (T/C) in the mid-dose and high-dose GOG groups were significantly reduced, and the inhibition rate (IR) in the two groups was significantly increased. The results indicated that the anti-tumor effect of GOG on gastric cancer cells may be related with the downregulation of caspase-3 and Bax and the upregulation of MMP-9 and VEGF.
Collapse
Affiliation(s)
- Yutao Liu
- Department of Pharmacy, Yantaishan Hospital, Yantai, China
| | - Xu Li
- Department of Pharmacy, Yantai Infectious Disease Hospital, Yantai, China
| | - Shanling Jiang
- Department of Pharmacy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Quanli Ge
- Department of Pharmacy, Yantaishan Hospital, Yantai, China
| |
Collapse
|
14
|
Wang H, He H, Meng H, Cui Y, Wang W. Effects of Grb2-associated binding protein 2-specific siRNA on the migration and invasion of MG-63 osteosarcoma cells. Oncol Lett 2018; 15:926-930. [PMID: 29422967 PMCID: PMC5772958 DOI: 10.3892/ol.2017.7375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 04/13/2017] [Indexed: 12/19/2022] Open
Abstract
To investigate the association between the expression of growth factor receptor binding protein 2-associated binding protein 2 (Gab2) in human osteosarcoma as well as the effects of Gab2 on invasion and metastasis, human MG-63 osteosarcoma cells were transfected with small interfering (si)RNA plasmid. Gab2 protein and mRNA expression levels were detected using western blotting and reverse transcription-polymerase chain reaction, respectively. The cell migration and invasion abilities were detected using in vitro chemotaxis and invasion assays, respectively, following siRNA vector expression. Gab2 was markedly expressed in MG-63 cells. The Gab2 protein and mRNA expression levels of the cells transfected with Gab2 siRNA (siGab2/MG-63) were reduced compared with those of the cells transfected with scrambled siRNA (Scr/MG-63). The chemotaxis assay demonstrated that the migration capacity of siGab2/MG-63 cells induced by 10 µg/l epidermal growth factor, was significantly reduced compared with that of the MG-63 and Scr/MG-63 cells (P<0.01). In comparison with Scr/MG-63 and MG-63 cells, a reduced number of siGab2/MG-63 cells invaded the Matrigel matrix, demonstrating that the in vitro invasion capacity was significantly decreased (P<0.01). Decreasing Gab2 expression levels using siRNA interference inhibited the migration and invasion ability of human MG-63 osteosarcoma cells.
Collapse
Affiliation(s)
- Huan Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hui He
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hongmei Meng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yang Cui
- Department of Orthopedic Surgery, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161000, P.R. China
| | - Wenbo Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
15
|
Silencing the Girdin gene enhances radio-sensitivity of hepatocellular carcinoma via suppression of glycolytic metabolism. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:110. [PMID: 28810896 PMCID: PMC5558715 DOI: 10.1186/s13046-017-0580-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/09/2017] [Indexed: 12/20/2022]
Abstract
Background Radiotherapy has been used increasingly to treat primary hepatocellular carcinoma. Clinically, the main cause of radiotherapy failure is cellular radioresistance, conferred via glycolytic metabolism. Our previous study demonstrated that Girdin is upregulated in primary hepatocellular carcinoma and promotes the invasion and metastasis of tumor cells. However, whether Girdin underlies the radio-sensitivity of hepatocellular carcinoma remains unclear. Methods A short hairpin RNA (shRNA) was used to silence CCDC88A (encoding Girdin), and real-time PCR was performed to determine CCDC88A mRNA expression. Then, cell proliferation, colony formation, flow cytometric, scratch, and transwell assays were to examine the influence of Girdin silencing on cellular radiosensitivity. Glycolysis assays were conducted to exam cell glycolysis process. Western blotting was performed to explore the signaling pathway downstream of Girdin. Finally, animal experiments were performed to demonstrate the effect of CCDC88A silencing on the radiosensitivity of hepatoma in vivo. Results shRNA-induced Girdin silencing suppressed glycolysis and enhanced the radio-sensitivity of hepatic cell lines, HepG2 and Huh-7. Furthermore, silencing of Girdin inhibited the PI3K/AKT/HIF-1α signaling pathway, which is a central regulator of glycolysis. Conclusion Girdin can regulate glycolysis in hepatocellular carcinoma cells through the PI3K/AKT/HIF-1α signaling pathway, which decreases the sensitivity of tumor cells to radiotherapy. Electronic supplementary material The online version of this article (doi:10.1186/s13046-017-0580-7) contains supplementary material, which is available to authorized users.
Collapse
|
16
|
Chen M, Li Y, Sun X, Zhang B, Li W, Wang S, Zhu X, Li F, Shi L. Grb2-associated binder 2 expression and its roles in uveal melanoma invasion. Mol Med Rep 2017; 16:4577-4582. [PMID: 28791340 PMCID: PMC5646995 DOI: 10.3892/mmr.2017.7151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/25/2017] [Indexed: 11/24/2022] Open
Abstract
Uveal melanoma (UM) is characterized by high metastasis and poor prognosis. A more improved understanding of the metastatic mechanism in UM cells is essential for the design of molecular therapy. Grb2-associated binder 2 (Gab2) has been reported to serve important roles in the progression of various types of human cancer. However, the role of Gab2 in the migration and invasion of UM remains unclear. The present study sought to further assess the expression of Gab2 in UM and the role of Gab2 in the invasion of UM cells. Clinical UM tissue samples and UM cell lines were analyzed using western blot analysis for the expression of Gab2. RNA interference was used to investigate the effect of Gab2 on the migratory and invasive characteristics of UM cells in vitro. The expression levels of matrix metalloproteinase (MMP)2, MMP9 and fascin in Gab2-knockdown, and control cells were also detected using western blot analysis. A total of 20 clinical UM samples and a subset of UM cell lines were investigated with uniformly high Gab2 expression. In the in vitro experiment, reduction of Gab2 using small interfering RNA inhibited the migration and invasion of UM cells by mediating MMPs, and fascin expression. These data suggest that Gab2 is a useful prognostic marker for UM and a novel therapeutic target for UM metastasis intervention.
Collapse
Affiliation(s)
- Meiling Chen
- Department of Pharmacology, Weifang Medical University, Weifang 261053, P.R. China
| | - Yuehua Li
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiuning Sun
- Department of Microbiology, Weifang Medical University, Weifang 261053, P.R. China
| | - Baogang Zhang
- Department of Pathology, Weifang Medical University, Weifang 261053, P.R. China
| | - Wei Li
- Department of Gynecology, Zhenjiang Maternity and Child Health Hospital, Zhenjiang 212001, P.R. China
| | - Shuxiao Wang
- Department of Pharmacology, Weifang Medical University, Weifang 261053, P.R. China
| | - Xuetao Zhu
- Department of Pharmacology, Weifang Medical University, Weifang 261053, P.R. China
| | - Feng Li
- Department of Pharmacology, Weifang Medical University, Weifang 261053, P.R. China
| | - Lihong Shi
- Department of Pharmacology, Weifang Medical University, Weifang 261053, P.R. China
| |
Collapse
|
17
|
Wang L, Huang D, Jiang Z, Luo Y, Norris C, Zhang M, Tian X, Tang Y. Akt3 is responsible for the survival and proliferation of embryonic stem cells. Biol Open 2017; 6:850-861. [PMID: 28483982 PMCID: PMC5483023 DOI: 10.1242/bio.024505] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/Akt) pathway plays an important role in regulating cell proliferation, metabolism, and survival. However, the distinct roles of Akt isoforms (Akt1, Akt2, and Akt3) in pluripotent stem cell maintenance are not fully defined. Using mouse embryonic stem cells (ESCs), we show that direct inhibition of Akt activity leads to ESC apoptosis. The Akt3, but not Akt1 or Akt2, activity specifically regulates this effect. Inhibiting Akt3 also leads to a cell cycle arrest at G1 phase. These regulatory roles of Akt3 are dependent on its kinase activity. Blocking the expression of Akt1 plus Akt2 in ESCs does not affect cell survival or proliferation, although blocking Akt1 aggravates the apoptotic effect induced by depletion of Akt3. We further show that blocking Akt3 in ESCs results in significant nuclear accumulation of p53, as well as the activation of its downstream targets, such as Mdm2, p21, and Fas. Inhibiting p53 and its downstream targets partially rescued the effects caused by Akt3-depletion. Our results revealed an Akt3 isoform-specific mechanism for ESC survival and proliferation involving the control of p53 activity. Summary: We identified that Akt isoform 3, but not Akt1 or Akt2, specifically regulates embryonic stem cell survival and proliferation. Mechanistically, this is achieved partially through controlling the p53 pathway activity.
Collapse
Affiliation(s)
- Ling Wang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Delun Huang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA.,Animal Reproduction Institute, Guangxi University, Nanning, 530004, People's Republic of China
| | - Zongliang Jiang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Yan Luo
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Carol Norris
- Center for Open Research Resources and Equipment, University of Connecticut, Storrs, CT 06269, USA
| | - Ming Zhang
- Animal Reproduction Institute, Guangxi University, Nanning, 530004, People's Republic of China
| | - Xiuchun Tian
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Young Tang
- Department of Animal Science, Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
18
|
Zhang B, Li H, Yin C, Sun X, Zheng S, Zhang C, Shi L, Liu Y, Lu S. Dock1 promotes the mesenchymal transition of glioma and is modulated by MiR‐31. Neuropathol Appl Neurobiol 2016; 43:419-432. [PMID: 26946516 DOI: 10.1111/nan.12321] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 02/25/2016] [Accepted: 03/04/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Baogang Zhang
- Department of Pathology Weifang Medical University Weifang China
| | - Hongli Li
- Medicine Research Center Weifang Medical University Weifang China
| | - Chonggao Yin
- College of Nursing Weifang Medical University Weifang China
| | - Xuemei Sun
- Department of Pathology Weifang Medical University Weifang China
| | - Shuxian Zheng
- Department of Pathology Weifang Medical University Weifang China
| | - Changjie Zhang
- Department of Pathology Weifang Medical University Weifang China
| | - Lihong Shi
- Department of Pharmacology Weifang Medical University Weifang China
| | - Yuqing Liu
- Department of Pathology Weifang Medical University Weifang China
| | - Shijun Lu
- Department of Pathology Weifang Medical University Weifang China
| |
Collapse
|
19
|
Sun B, Chen L, Fu H, Guo L, Guo H, Zhang N. Upregulation of RICTOR gene transcription by the proinflammatory cytokines through NF-κB pathway contributes to the metastasis of renal cell carcinoma. Tumour Biol 2015; 37:4457-66. [PMID: 26500094 DOI: 10.1007/s13277-015-4296-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022] Open
Abstract
Metastasis accounts for more than 50 % of deaths among renal cell carcinoma (RCC) patients, and therefore, it is important to study the biology of metastasis and identify metastasis-associated biomarkers for risk prognosis and stratification of patients for an individualized therapy of RCC. In cultured RCC cells, knockdown of Rictor by short hairpin RNA (shRNA) inhibited cell migration and invasion, probably due to impairments in activation of Akt. Pretreatment with tumor necrosis factor α (TNFα) or interleukin 6 (IL-6) enhanced the expression of Rictor and the migration of renal cancer cells. Mechanistic analysis showed that TNFα induced the activation of NF-κB in RCC cells. Luciferase reporter analysis revealed a NF-κB responding element (-301 to -51 bp) at the promoter region of Rictor. Chromatin immunoprecipitation (ChIP) analysis further confirmed that TNFα-induced binding of p65 with the promoter of Rictor. In a xenograft model, knockdown of Rictor-blocked RCC cells metastasis to the mouse lungs and livers. Taken together, our results suggest that the proinflammatory cytokine TNFα promotes the expression of Rictor through the NF-κB pathway.
Collapse
Affiliation(s)
- Bo Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People's Republic of China
| | - Liwei Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People's Republic of China
| | - Hui Fu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People's Republic of China
| | - Lin Guo
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People's Republic of China
| | - Hua Guo
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People's Republic of China.
| | - Ning Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, People's Republic of China. .,Research Center of Basic Medical Science, Tianjin Medical University, No. 22 Qixiangtai Road, Heping District, Tianjin, 300070, People's Republic of China.
| |
Collapse
|
20
|
Cai J, Zhao J, Zhang N, Xu X, Li R, Yi Y, Fang L, Zhang L, Li M, Wu J, Zhang H. MicroRNA-542-3p Suppresses Tumor Cell Invasion via Targeting AKT Pathway in Human Astrocytoma. J Biol Chem 2015; 290:24678-88. [PMID: 26286747 DOI: 10.1074/jbc.m115.649004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Indexed: 01/17/2023] Open
Abstract
The molecular mechanism underlying constitutive activation of AKT signaling, which plays essential roles in astrocytoma progression, is not fully characterized. Increasing numbers of studies have reported that microRNAs are involved in the malignant behavior of astrocytoma cells via directly targeting multiple oncogenes or tumor suppressors. Here, we found that microRNA (miR)-542-3p expression was decreased in glioblastoma cell lines and astrocytoma tissues, and reduced levels of miR-542-3p expression correlated with high histopathological grades and poor prognosis of astrocytoma patients. Exogenous miR-542-3p suppressed glioblastoma cell invasion through not only targeting AKT1 itself but also directly down-regulating its two important upstream regulators, namely, integrin-linked kinase and PIK3R1. Notably, overexpressing miR-542-3p decreased AKT1 phosphorylation and directly and indirectly repressed nuclear translocation and transactivation activity of β-catenin to exert its anti-invasive effect. Furthermore, the miR-542-3p expression level negatively correlated with AKT activity as well as levels of integrin-linked kinase and PIK3R1 in human astrocytoma specimens. These findings suggest that miR-542-3p acts as a negative regulator in astrocytoma progression and that miR-542-3p down-regulation contributes to aberrant activation of AKT signaling, leaving open the possibility that miR-542-3p may be a potential therapeutic target for high grade astrocytoma.
Collapse
Affiliation(s)
- Junchao Cai
- From the Departments of Microbiology and the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | | | | | - Xiaonan Xu
- From the Departments of Microbiology and the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | - Rong Li
- From the Departments of Microbiology and the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | - Yang Yi
- Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University
| | - Lishan Fang
- From the Departments of Microbiology and the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | - Le Zhang
- From the Departments of Microbiology and the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | - Mengfeng Li
- From the Departments of Microbiology and the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | - Jueheng Wu
- the Key Laboratory of Tropical Disease Control (Sun Yat-Sen University), Ministry of Education, and
| | - Heng Zhang
- Neurosurgery Intensive Care Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
21
|
Chen W, Zhang B, Guo W, Gao L, Shi L, Li H, Lu S, Liu Y, Li X. miR-429 inhibits glioma invasion through BMK1 suppression. J Neurooncol 2015; 125:43-54. [PMID: 26272601 DOI: 10.1007/s11060-015-1887-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 08/08/2015] [Indexed: 02/07/2023]
Abstract
The purpose of this research was to examine the relationship between big mitogen-activated protein kinase 1 (BMK1) and miRNA miR-429 and to determine the effect of miR-429 on glioma invasiveness. Immunohistochemistry was used to evaluate BMK1 expression in glioma tissues. Real-time PCR was used to measure the expression of miR-429 and other RNAs. Western blot was used to detect the expression of BMK1 and other related proteins. Wound healing, Matrigel invasion, and chemotaxis assays were performed to detect the invasion and migration of glioma cell lines. The actual binding site of miR-429 to the 3' untranslated region of BMK1 was confirmed by luciferase assay and RNA immunoprecipitation. BMK1 expression was associated with the World Health Organization grading of glioma and inversely correlated with patient survival. Suppression of BMK1 inhibited the migration and invasion of glioma cells by interfering with mesenchymal transition. Additionally, hepatocyte growth factor-induced GSK3β phosphorylation was suppressed through BMK1 knockdown. Interestingly, our findings validated a novel role for miR-429 in suppressing the migration and invasion of glioma by directly inhibiting BMK1 expression. We also found that miR-429 expression in glioma cells and tissues was lower than that in normal cells and adjacent non-neoplastic tissues, and miR-429 overexpression inhibited invasive activity of glioma cells both in vitro and in vivo. Furthermore, our data validated that miR-429 downregulation was due to the hypermethylation of its promoter region. Our results indicated that BMK1 modulation by miR-429 has an important function in glioma invasion both in vitro and in vivo.
Collapse
Affiliation(s)
- Weiyi Chen
- Department of Pathology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Baogang Zhang
- Department of Pathology, Weifang Medical University, Weifang, 261053, People's Republic of China.
| | - Wenjun Guo
- Department of Pathology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Linlin Gao
- Department of Internal Medicine, NO.3, Laixi Municipal Hospital, Laixi, 266600, People's Republic of China
| | - Lihong Shi
- Department of Pharmacology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Hongli Li
- Department of Medicine Research Center, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Shijun Lu
- Department of Pathology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Yuqing Liu
- Department of Pathology, Weifang Medical University, Weifang, 261053, People's Republic of China
| | - Xiaolong Li
- Department of Pathology, Weifang Medical University, Weifang, 261053, People's Republic of China
| |
Collapse
|
22
|
Ni W, Fang Y, Tong L, Tong Z, Yi F, Qiu J, Wang R, Tong X. Girdin regulates the migration and invasion of glioma cells via the PI3K-Akt signaling pathway. Mol Med Rep 2015; 12:5086-92. [PMID: 26151295 PMCID: PMC4581799 DOI: 10.3892/mmr.2015.4049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 06/18/2015] [Indexed: 01/10/2023] Open
Abstract
Girdin, an actin‑binding protein, is associated with cell migration and is expressed at high levels in glioma cells. However, the association between girdin and the development of glioma remains to be elucidated. In the present study, short‑hairpin RNA technology was used to silence the gene expression of girdin. The effects of girdin silencing on glioma cell proliferation, migration and invasion were then assessed using a cell viability assay, wound‑healing assay, transwell invasion assay, reverse transcription‑quantitative polymerase chain reaction, western blot analysis and gelatin zymography. The results suggested that girdin silencing inhibited the proliferation, migration and invasion of glioma cells. In addition, the expression levels and activity of matrix metalloproteinase (MMP)‑2 and MMP‑9 were also affected by girdin silencing. Further mechanistic investigation indicated that girdin may regulate glioma cell migration and invasion through the phosphatidylinositol‑3‑kinase/protein kinase B (PI3K‑Akt) signaling pathway. Therefore, the results of the present study provide a theoretical foundation for the development of anticancer drugs.
Collapse
Affiliation(s)
- Weimin Ni
- Department of Human Anatomy, Histology and Embryology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yan Fang
- Department of Human Anatomy, Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Lei Tong
- Department of Human Anatomy, Histology and Embryology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhaoxue Tong
- Department of Human Anatomy, Histology and Embryology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Fuxin Yi
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Jianwu Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Rui Wang
- Department of Neurosurgery, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xiaojie Tong
- Department of Human Anatomy, Histology and Embryology, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
23
|
Ghosh P. Heterotrimeric G proteins as emerging targets for network based therapy in cancer: End of a long futile campaign striking heads of a Hydra. Aging (Albany NY) 2015; 7:469-74. [PMID: 26224586 PMCID: PMC4543036 DOI: 10.18632/aging.100781] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 07/15/2015] [Indexed: 01/17/2023]
Abstract
Most common diseases, e.g., cancer are driven by not one, but multiple cell surface receptors that trigger and sustain a pathologic signaling network. The largest fraction of therapeutic agents that target individual receptors/pathways eventually fail due to the emergence of compensatory mechanisms that reestablish the pathologic network. Recently, a rapidly emerging paradigm has revealed GIV/Girdin as a central platform for receptor cross-talk which integrates signals downstream of a myriad of cell surface receptors, and modulates several key pathways within downstream signaling network, all via non-canonical activation of trimeric G proteins. Unlike canonical signal transduction via G proteins, which is spatially and temporally restricted, the temporal and spatial features of non-canonical activation of G protein via GIV is unusually unrestricted. Consequently, the GIV●G protein interface serves as a central hub allowing for control over several pathways within the pathologic signaling network, all at once. The relevance of this new paradigm in cancer and other disease states and the pros and cons of targeting the GIV●G protein interface are discussed.
Collapse
Affiliation(s)
- Pradipta Ghosh
- Department of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
24
|
Fortier AM, Asselin E, Cadrin M. Functional specificity of Akt isoforms in cancer progression. Biomol Concepts 2015; 2:1-11. [PMID: 25962016 DOI: 10.1515/bmc.2011.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Akt/PKB kinases are central mediators of cell homeostasis. There are three highly homologous Akt isoforms, Akt1/PKBα, Akt2/PKBβ and Akt3/PKBγ. Hyperactivation of Akt signaling is a key node in the progression of a variety of human cancer, by modulating tumor growth, chemoresistance and cancer cell migration, invasion and metastasis. It is now clear that, to understand the mechanisms on how Akt affects specific cancer cells, it is necessary to consider the relative importance of each of the three Akt isoforms in the altered cells. Akt1 is involved in tumor growth, cancer cell invasion and chemoresistance and is the predominant altered isoform found in various carcinomas. Akt2 is related to cancer cell invasion, metastasis and survival more than tumor induction. Most of the Akt2 alterations are observed in breast, ovarian, pancreatic and colorectal carcinomas. As Akt3 expression is limited to some tissues, its implication in tumor growth and resistance to drugs mostly occurs in melanomas, gliomas and some breast carcinomas. To explain how Akt isoforms can play different or even opposed roles, three mechanisms have been proposed: tissue-specificity expression/activation of Akt isoforms, distinct effect on same substrate as well as specific localization through the cyto-skeleton network. It is becoming clear that to develop an effective anticancer Akt inhibitor drug, it is necessary to target the specific Akt isoform which promotes the progression of the specific tumor.
Collapse
|
25
|
Gu F, Zhang H, Qin F, Liu X, Li W, Fu L, Ying G, Li B, Zhang M, Ma Y. Intersectin1-S, a multidomain adapter protein, is essential for malignant glioma proliferation. Glia 2015; 63:1595-605. [PMID: 25832561 DOI: 10.1002/glia.22830] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/17/2015] [Indexed: 01/07/2023]
Abstract
Glioblastomas, the most aggressive form of primary brain tumors with a tendency to invade surrounding healthy brain tissues, remains an incurable disease. Intersectin (ITSN) is a multidomain adapter protein implicated in endocytosis, exocytosis, and multiple signaling pathways. Prior research of ours has shown intersectin1-S (ITSN1-S) is critical for the migration and invasion of glioma cells by regulating several key proteins. In this study, we established ITSN1-S expression patterns in human tumor tissues. We discovered that ITSN1-S expression was positively correlated with histological grade of gliomas and with poor patient prognosis. We also found that the expression of ITSN1-S protein was essential to glioblastoma cell proliferation. Furthermore, through a series of expression constructs encoding different ITSN1-S domains, we identified the critical roles of ITSN1-S SH3 domains in the regulation of cell proliferation. This study also demonstrates evidence suggesting that the regulation of ITSN1-S on glioblastoma cells proliferation is through the Raf/MEK/ERK pathway. In conclusion, this study suggests critical roles of ITSN1-S in malignant glioma proliferation, indicating a potential usage of ITSN1-S in the therapeutic intervention as a novel molecular target.
Collapse
Affiliation(s)
- Feng Gu
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Huikun Zhang
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Fengxia Qin
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Xiaoli Liu
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Wenliang Li
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Li Fu
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Guoguang Ying
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Binghui Li
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, Institute of Bioinformatics, University of Georgia, Athens, Georgia
| | - Yongjie Ma
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, People's Republic of China
| |
Collapse
|
26
|
Shi L, Zhang B, Sun X, Lu S, Liu Z, Liu Y, Li H, Wang L, Wang X, Zhao C. MiR-204 inhibits human NSCLC metastasis through suppression of NUAK1. Br J Cancer 2014; 111:2316-2327. [PMID: 25412236 PMCID: PMC4264457 DOI: 10.1038/bjc.2014.580] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/07/2014] [Accepted: 09/16/2014] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer-related mortality worldwide and non-small-cell lung carcinoma (NSCLC) is responsible for almost 80% of lung cancer-related deaths. Identifying novel molecules that can repress the invasiveness and metastasis of lung cancer will facilitate the development of new antilung cancer strategies. The aim of this study is to determine the roles of NUAK1 (a downstream of Akt) and miR-204 in the invasiveness and metastasis of NSCLC and to reveal the correlation between NUAK1 and miR-204. METHODS The expression of NUAK1 in primary human NSCLC tissues was evaluated by immunohistochemistry. Real-time PCR was employed to measure the expression level of miR-204. The effect of NUAK1 and miR204 on the prognosis of NSCLC patients was evaluated by log-rank test. The siRNA transfection was used to manipulate the expression levels of NUAK1 and miR204 in cancer cells. Chemotaxis assay, Scratch assay, and Matrigel invasion assay were performed to evaluate the migration and invasion of cells. Cellular F-actin measurement was used to measure F-actin polymerisation in lung cancer cells. Western blot was used to detect the expression levels of corresponding proteins. The Luciferase assay and RNA immunoprecipitation were used to confirm the actual binding site of miR-204 to 3'UTR of NUAK1. RESULTS Increased expression of NUAK1 is correlated with the invasiveness and metastasis of human NSCLC. Knockdown of NUAK1 inhibited cell migration and invasion. In addition, this study showed that NUAK1 influenced mTOR phosphorylation and induced the phosphorylation of p70S6K1 and eukaryotic initiation factor 4E-binding protein1 (4E-BP1), two downstream targets of mTOR in NSCLC cells. At the same time, decreased expression of miR-204 promoted NSCLC progression and, contrarily, manipulated upregulation of miR-204-inhibited cell migration and invasion. There is clinical relevance between miR-204 downregulation and NUAK1 upregulation in human NSCLC. Furthermore, we found that miR-204 inhibited NSCLC tumour invasion by directly targeting and downregulating NUAK1 expression. Finally, our data suggested that the downregulation of miR-204 was due to hypermethylation of its promoter region. CONCLUSIONS Our results indicate that NUAK1 is excessively expressed in NSCLC and plays important roles in NSCLC invasion. The miR-204 acts as a tumour suppressor by inhibiting NUAK1 expression in NSCLC. Both NUAK1 and miR-204 may serve as potential targets of NSCLC therapy.
Collapse
Affiliation(s)
- L Shi
- Department of Pharmacology, Weifang Medical University, Weifang 261053, People's Republic China
| | - B Zhang
- Department of Pathology, Weifang Medical University, Weifang 261053, People's Republic China
| | - X Sun
- Department of Microbiology, Weifang Medical University, Weifang 261053, People's Republic China
| | - S Lu
- Department of Pathology, Weifang Medical University, Weifang 261053, People's Republic China
| | - Z Liu
- Department of Microbiology, Weifang Medical University, Weifang 261053, People's Republic China
| | - Y Liu
- Department of Pathology, Weifang Medical University, Weifang 261053, People's Republic China
| | - H Li
- Department of Medicine Research Center, Weifang Medical University, Weifang 261053, People's Republic China
| | - L Wang
- Department of Pharmacology, Weifang Medical University, Weifang 261053, People's Republic China
| | - X Wang
- Department of Pharmacology, Weifang Medical University, Weifang 261053, People's Republic China
| | - C Zhao
- Department of Pharmacology, Weifang Medical University, Weifang 261053, People's Republic China
| |
Collapse
|
27
|
Kang W, Tong JHM, Lung RWM, Dong Y, Yang W, Pan Y, Lau KM, Yu J, Cheng AS, To KF. let-7b/g silencing activates AKT signaling to promote gastric carcinogenesis. J Transl Med 2014; 12:281. [PMID: 25288334 PMCID: PMC4196013 DOI: 10.1186/s12967-014-0281-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/24/2014] [Indexed: 12/13/2022] Open
Abstract
Background Aberrant AKT activation contributes to gastric cancer cell survival and chemotherapy resistance, however its regulation is poorly understood. microRNAs have been established to be important regulators in gastric carcinogenesis. Here, we showed the functional role and putative target of let-7b and let-7g (let-7b/g) in gastric carcinogenesis. Methods The expression of let-7b/g in gastric cancer cell lines and primary tumors were evaluated by miRNA qRT-PCR. The putative target gene of let-7b/g was explored by TargetScan followed by further validation. Functional analyses including MTT proliferation, monolayer colony formation, cell invasion assays and in vivo study were performed in both ectopic expression and knockdown approaches. Results let-7b/g was found down-regulated in gastric cancer and its downregulation was associated with poor survival and correlated with lymph node metastasis. let-7b/g inhibited AKT2 expression by directly binding to its 3’UTR, reduced p-AKT (S473) activation and suppressed expression of the downstream effector pS6. AKT2 mRNA expression showed negative correlation with the expression of let-7b/g in primary tumors. Short interfering RNA (siRNA) mediated knockdown of AKT2 phenocopied the tumor-suppressive effects of let-7b/g. Moreover, AKT2 re-expression partly abrogated the growth-inhibitory effect of let-7b/g. Conclusion In conclusion, our findings reveal decreased let-7b/g contributes to aberrant AKT activation in gastric tumorigenesis and provide a potential therapeutic strategy for gastric cancer. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0281-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
28
|
Gu F, Wang L, He J, Liu X, Zhang H, Li W, Fu L, Ma Y. Girdin, an actin-binding protein, is critical for migration, adhesion, and invasion of human glioblastoma cells. J Neurochem 2014; 131:457-69. [PMID: 25060559 DOI: 10.1111/jnc.12831] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 01/05/2023]
Abstract
Girdin, an actin-binding protein, possesses versatile functions in a multitude of cellular processes. Although several studies have shown that Girdin is involved in the cell DNA synthesis, actin cytoskeleton rearrangement, and cell motility, the molecular mechanisms of Girdin in tumor development and progression remain elusive. In this study, through over-expression and siRNA experiments, we found that Girdin increased migration of LN229 human glioblastoma cells. On the other hand, reducing Girdin impaired F-actin polymerization, which is essential for cell morphogenesis and motility. Matrix metalloproteinase 2, critical in human glioma migration and invasion, was down-regulated upon Girdin reduction and led to decreased invasion in vitro and in vivo. In addition, silencing Girdin expression impaired the phosphorylation of two important adhesion molecules, integrin β1 and focal adhesion kinase, resulting in cell adhesion defects. Our immunohistochemical study on human gliomas tissue sections indicated that Girdin expression was positively related with glioma malignancy, supporting the in vitro and in vivo results from cell lines. Collectively, our findings suggest a critical role for Girdin in glioma infiltration. We show that reduction of Girdin, an actin-binding protein, leads to impaired F-actin polymerization and down-regulated expression of matrix metallopeptidase protein 2 (MMP-2), phosphorylated integrin β1, and phosphorylated focal adhesion kinase (FAK), which resulted in decreased migration, adhesion, and invasion of glioblastoma cells. Girdin was positively correlated with glioma malignancy and negatively associated with clinical prognosis, suggesting Girdin as a critical regulator in glioma infiltration.
Collapse
Affiliation(s)
- Feng Gu
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Tang Y, Jiang Z, Luo Y, Zhao X, Wang L, Norris C, Tian XC. Differential effects of Akt isoforms on somatic cell reprogramming. J Cell Sci 2014; 127:3998-4008. [PMID: 25037569 DOI: 10.1242/jcs.150029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Akt plays an important role in cell growth, proliferation and survival. The specific roles of the three Akt isoforms in somatic cell reprogramming have not been investigated. Here we report that, during iPSC generation, enhanced Akt1 activity promotes complete reprogramming mainly through increased activation of Stat3 in concert with leukemia inhibitory factor (LIF) and, to a lesser extent, through promotion of colony formation. Akt1 augments Stat3 activity through activation of mTOR and upregulation of LIF receptor expression. Similarly, enhanced Akt2 or Akt3 activation also promotes reprogramming and coordinates with LIF to activate Stat3. Blocking Akt1 or Akt3 but not Akt2 expression prohibits cell proliferation and reprogramming. Furthermore, the halt in cell proliferation and reprogramming caused by mTOR and Akt inhibitors can be reversed by inhibition of GSK3. Finally, we found that expressing the GSK3β target Esrrb overrides inhibition of Akt and restores reprogramming. Our data demonstrated that during reprogramming, Akt promotes establishment of pluripotency through co-stimulation of Stat3 activity with LIF. Akt1 and Akt3 are essential for the proliferation of reprogrammed cells, and Esrrb supports cell proliferation and complete reprogramming during Akt signaling.
Collapse
Affiliation(s)
- Yong Tang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Zongliang Jiang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Yan Luo
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Xueming Zhao
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Ling Wang
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| | - Carol Norris
- Department of Molecular and Cellular Biology/Biotechnology and Bioservices Center, University of Connecticut, Storrs, CT 06269, USA
| | - Xiuchun Cindy Tian
- Center for Regenerative Biology, Department of Animal Science, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
30
|
Paul P, Volny N, Lee S, Qiao J, Chung DH. Gli1 transcriptional activity is negatively regulated by AKT2 in neuroblastoma. Oncotarget 2014; 4:1149-57. [PMID: 23900341 PMCID: PMC3787147 DOI: 10.18632/oncotarget.1074] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Activation of the Hedgehog (Hh) signaling pathway has been implicated in a variety of malignancies including neuroblastoma. Expression of Gli1, a downstream effector of Hh, correlates with a favorable prognosis in patients with neuroblastoma. Moreover, Gli1 overexpression reduces mitotic index and induces transcription of genes involved in the differentiation of neuroblastoma cells; however, much remains unknown regarding the regulation of Gli1 transcriptional activity. Here, we report a novel negative regulation of Gli1 transcriptional activity by PI3K/AKT2 signal transduction pathway. Constitutively active PI3K subunit, p110α, inhibited Gli1 transcriptional activity in neuroblastoma cells, whereas, overexpression of an inactive form of PI3K subunit, p85, enhanced its activity. Specifically, the AKT2 isoform inhibited Gli1 luciferase activity. Silencing AKT2 using siRNA increased Gli1 transcriptional activity and conversely, overexpression of constitutively active AKT2 (myr-AKT2) decreased Gli1 transcriptional activity. Furthermore, Gli1 overexpression-mediated decrease in anchorage-independent growth was rescued by AKT2 overexpression. We also demonstrated that AKT2 overexpression regulates the nuclear-cytoplasmic distribution of exogenous Gli1 protein in neuroblastoma cells by relieving a GSK3β-mediated destabilization of SUFU, a negative regulator of Gli1 nuclear translocation. Inhibition of nuclear Gli1 accumulation may explain for the suppression of the tumor-suppressive function of Gli1. Collectively, our findings suggest an important role of Gli1 as a tumor suppressor in neuroblastoma, and offer a mechanism by which AKT2 regulates the subcellular localization, and in turn, inhibits the tumor-suppressive function of Gli1 in neuroblastoma.
Collapse
Affiliation(s)
- Pritha Paul
- Department of Pediatric Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | |
Collapse
|
31
|
Chin YR, Yuan X, Balk SP, Toker A. PTEN-deficient tumors depend on AKT2 for maintenance and survival. Cancer Discov 2014; 4:942-55. [PMID: 24838891 DOI: 10.1158/2159-8290.cd-13-0873] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
UNLABELLED Loss of PTEN is a common event in many cancers and leads to hyperactivation of the PI3K-AKT signaling pathway. The mechanisms by which AKT isoforms mediate signaling to phenotypes associated with PTEN inactivation in cancer have not been defined. Here, we show that AKT2 is exclusively required for PTEN-deficient prostate tumor spheroid maintenance, whereas AKT1 is dispensable. shRNA silencing of AKT2 but not AKT1 promotes regression of prostate cancer xenografts. Mechanistically, we show that AKT2 silencing upregulates p21 and the proapoptotic protein BAX and downregulates the insulin-like growth factor receptor-1. We also show that p21 is an effector of AKT2 in mediating prostate tumor maintenance. Moreover, AKT2 is also exclusively required for the maintenance and survival of other PTEN-deficient solid tumors, including breast cancer and glioblastoma. These findings identify a specific function for AKT2 in mediating survival of PTEN-deficient tumors and provide a rationale for developing therapeutics targeting AKT2. SIGNIFICANCE Depletion of AKT2, but not AKT1, induces potent tumor regression in PTEN-deficient prostate cancer xenografts, concomitant with upregulation of p21, which may serve as a potential biomarker for screening AKT2 activity in clinical samples. The specific role of AKT2 in tumor maintenance provides a rationale for the development of isoform-specific inhibitors for patients with PTEN-deficient cancers.
Collapse
Affiliation(s)
| | - Xin Yuan
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Steven P Balk
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
32
|
Wang C, Lin J, Li L, Wang Y. Expression and clinical significance of girdin in gastric cancer. Mol Clin Oncol 2014; 2:425-428. [PMID: 24772312 DOI: 10.3892/mco.2014.265] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/03/2014] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer is one of the most common malignant tumors and the second leading cause of cancer-related mortality. Elucidating the molecular mechanism underlying the development of gastric cancer is crucial in identifying gastric cancer-susceptible populations, screening for tumor markers and in the application of gene therapy. This study was conducted to investigate girdin expression in gastric cancer and para-cancer tissues and to elucidate the role of girdin in the development of gastric cancer. Tissue micro-array and streptavidin-peroxidase immunohistochemical staining were used to detect girdin expression in 105 gastric cancer and 72 para-cancer tissue samples. Analyses of the patients' clinical and pathological data were also performed. The expression ratio of girdin was 40.0% in gastric cancer and 11.1% in the para-cancer tissues and the difference was statistically significant (P<0.05). Girdin expression was found to be positively correlated (P<0.05) with tumor invasion depth and lymph node metastasis, while no significant associations were found between girdin expression and gender, age, tumor size, pathological grade and clinical stage (P>0.05). In conclusion, the upregulation of girdin expression in gastric cancer may contribute to tumor metastasis and cancer development, suggesting that girdin may be a novel indicator for evaluating lymph node metastasis and gastric cancer outcome.
Collapse
Affiliation(s)
- Caihua Wang
- Department of Gastroenterology, Guanghe Chinese and Western Medicine Hospital, Zhangjiagang, Jiangsu 215633
| | - Jie Lin
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Lurong Li
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Yun Wang
- Institute of Digestive Endoscopy and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
33
|
Chautard E, Ouédraogo ZG, Biau J, Verrelle P. Role of Akt in human malignant glioma: from oncogenesis to tumor aggressiveness. J Neurooncol 2014; 117:205-15. [PMID: 24477623 DOI: 10.1007/s11060-014-1382-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/19/2014] [Indexed: 12/21/2022]
Abstract
Gathering evidence has revealed that Akt signaling pathway plays an important role in glioma progression and aggressiveness. Among Akt kinases the most studied, Akt1, has been involved in many cellular processes that are in favor of cell malignancy. More recently, the actions of the two other isoforms, Akt2 and Akt3 have emerged in glioma. After a description of Akt pathway activation, we will explore the role of each isoform in malignant glioma that strengthens the current preclinical and clinical studies evaluating the impact of Akt pathway targeting in glioblastomas.
Collapse
Affiliation(s)
- Emmanuel Chautard
- Clermont Université, Université d'Auvergne, EA7283 CREaT, 28 Place Henri Dunant, 63000, Clermont-Ferrand, France,
| | | | | | | |
Collapse
|
34
|
Tamim S, Vo DT, Uren PJ, Qiao M, Bindewald E, Kasprzak WK, Shapiro BA, Nakaya HI, Burns SC, Araujo PR, Nakano I, Radek AJ, Kuersten S, Smith AD, Penalva LOF. Genomic analyses reveal broad impact of miR-137 on genes associated with malignant transformation and neuronal differentiation in glioblastoma cells. PLoS One 2014; 9:e85591. [PMID: 24465609 PMCID: PMC3899048 DOI: 10.1371/journal.pone.0085591] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 12/05/2013] [Indexed: 02/05/2023] Open
Abstract
miR-137 plays critical roles in the nervous system and tumor development; an increase in its expression is required for neuronal differentiation while its reduction is implicated in gliomagenesis. To evaluate the potential of miR-137 in glioblastoma therapy, we conducted genome-wide target mapping in glioblastoma cells by measuring the level of association between PABP and mRNAs in cells transfected with miR-137 mimics vs. controls via RIPSeq. Impact on mRNA levels was also measured by RNASeq. By combining the results of both experimental approaches, 1468 genes were found to be negatively impacted by miR-137--among them, 595 (40%) contain miR-137 predicted sites. The most relevant targets include oncogenic proteins and key players in neurogenesis like c-KIT, YBX1, AKT2, CDC42, CDK6 and TGFβ2. Interestingly, we observed that several identified miR-137 targets are also predicted to be regulated by miR-124, miR-128 and miR-7, which are equally implicated in neuronal differentiation and gliomagenesis. We suggest that the concomitant increase of these four miRNAs in neuronal stem cells or their repression in tumor cells could produce a robust regulatory effect with major consequences to neuronal differentiation and tumorigenesis.
Collapse
Affiliation(s)
- Saleh Tamim
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Dat T. Vo
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Philip J. Uren
- Molecular and Computational Biology Section, Division of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Mei Qiao
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Eckart Bindewald
- Basic Science Program, SAIC-Frederick, Inc., Center for Cancer Research Nanobiology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Wojciech K. Kasprzak
- Basic Science Program, SAIC-Frederick, Inc., Center for Cancer Research Nanobiology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Bruce A. Shapiro
- Center for Cancer Research Nanobiology Program, National Cancer Institute, Frederick, Maryland, California
| | - Helder I. Nakaya
- Department of Clinical Analyses and Toxicology, Institute of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Suzanne C. Burns
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Patricia R. Araujo
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Ichiro Nakano
- Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States of America
| | - Agnes J. Radek
- Epicentre (An Illumina Company), Madison, Wisconsin, United States of America
| | - Scott Kuersten
- Epicentre (An Illumina Company), Madison, Wisconsin, United States of America
| | - Andrew D. Smith
- Molecular and Computational Biology Section, Division of Biological Sciences, University of Southern California, Los Angeles, California, United States of America
| | - Luiz O. F. Penalva
- Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
35
|
Shi L, Sun X, Zhang J, Zhao C, Li H, Liu Z, Fang C, Wang X, Zhao C, Zhang X, Zhou F, Lu S, Luo R, Zhang B. Gab2 expression in glioma and its implications for tumor invasion. Acta Oncol 2013; 52:1739-50. [PMID: 23231021 DOI: 10.3109/0284186x.2012.750032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Gliomas are characterized by high invasiveness and poor prognosis. Better understanding of the mechanism of invasion in glioma cells is essential to the design of effective therapy. Recently Grb2-associated binder 2 (Gab2), a member of the DOS/Gab family of scaffolding adapters, has been reported to play important roles in the development and progression of human cancers. However, it is not known whether Gab2 has any role in the migration and invasion of gliomas. This study attempts to investigate the association between Gab2 expression and progression of gliomas and the molecular mechanism of Gab2 in the glioma cell invasion. Methods. The expression of Gab2 in pairs of matched glioma tissues and their normal brain tissues was detected by Western blot. Immunohistochemistry was applied to evaluate the expression of Gab2 in 163 cases of histologically diagnosed gliomas. The invasive character of Gab2 decreased glioma cells and control glioma cells were investigated in vitro and in vivo in SCID mice brain. Results. Gab2 is found to be high expressed in gliomas and a subset of cancer cell lines. Statistical analysis suggested that the up-regulation of Gab2 correlated with the WHO grade of gliomas (p < 0.01) and that patients with high Gab2 expression levels exhibited shorter survival time (p < 0.01). In an animal experiment, knockdown of Gab2 through siRNA inhibited invasive ability of glioma cells into the brain of SCID mice. In cell research, reduction of Gab2 by siRNA inhibits the migration and invasion of glioma cells by mediating cytoskeleton rearrangement and MMPs expression. Additionally, IGF-1-induced pAkt and pmTOR phosphorylation was suppressed by the knockdown of Gab2. Conclusion. Gab2 may be a useful prognostic marker for gliomas and a novel therapeutic target for glioma invasion intervention.
Collapse
Affiliation(s)
- Lihong Shi
- Department of Oncology, Nanfang Hospital of Southern Medical University , Guangzhou , PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fortin Ensign SP, Mathews IT, Symons MH, Berens ME, Tran NL. Implications of Rho GTPase Signaling in Glioma Cell Invasion and Tumor Progression. Front Oncol 2013; 3:241. [PMID: 24109588 PMCID: PMC3790103 DOI: 10.3389/fonc.2013.00241] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/02/2013] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GB) is the most malignant of primary adult brain tumors, characterized by a highly locally invasive cell population, as well as abundant proliferative cells, neoangiogenesis, and necrosis. Clinical intervention with chemotherapy or radiation may either promote or establish an environment for manifestation of invasive behavior. Understanding the molecular drivers of invasion in the context of glioma progression may be insightful in directing new treatments for patients with GB. Here, we review current knowledge on Rho family GTPases, their aberrant regulation in GB, and their effect on GB cell invasion and tumor progression. Rho GTPases are modulators of cell migration through effects on actin cytoskeleton rearrangement; in non-neoplastic tissue, expression and activation of Rho GTPases are normally under tight regulation. In GB, Rho GTPases are deregulated, often via hyperactivity or overexpression of their activators, Rho GEFs. Downstream effectors of Rho GTPases have been shown to promote invasiveness and, importantly, glioma cell survival. The study of aberrant Rho GTPase signaling in GB is thus an important investigation of cell invasion as well as treatment resistance and disease progression.
Collapse
Affiliation(s)
- Shannon Patricia Fortin Ensign
- Cancer and Cell Biology Division, Translational Genomics Research Institute , Phoenix, AZ , USA ; Cancer Biology Graduate Interdisciplinary Program, University of Arizona , Tucson, AZ , USA
| | | | | | | | | |
Collapse
|
37
|
Zhang B, Yin C, Li H, Shi L, Liu N, Sun Y, Lu S, Liu Y, Sun L, Li X, Chen W, Qi Y. Nir1 promotes invasion of breast cancer cells by binding to chemokine (C-C motif) ligand 18 through the PI3K/Akt/GSK3β/Snail signalling pathway. Eur J Cancer 2013; 49:3900-13. [PMID: 24001613 DOI: 10.1016/j.ejca.2013.07.146] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Revised: 06/30/2013] [Accepted: 07/29/2013] [Indexed: 12/22/2022]
Abstract
Chemokine (C-C motif) ligand 18 (CCL18), which is derived from tumour-associated macrophages (TAMs), plays a critical role in promoting breast cancer metastasis via its receptor, PYK2 N-terminal domain interacting receptor 1 (Nir1). However, the molecular mechanism by which Nir1 promotes breast cancer metastasis by binding to CCL18 remains elusive. In this study, Nir1 expression was associated with lymph node and distant metastasis in patients with invasive ductal carcinoma. For the first time, we report that Nir1 binding to CCL18 promotes the phosphorylation of Akt, LIN-11, Isl1 and MEC-3 protein domain kinase (LIMK), and cofilin, which is a critical step in cofilin recycling and actin polymerisation. Interestingly, Nir1 binding to CCL18 can enhance cell mesenchymal properties and induce epithelial-mesenchymal transition (EMT). Mechanistically, Nir1 binding to CCL18 stabilises Snail via the Akt/GSK3β signalling pathway. In support of these observations, Nir1 binding to CCL18 promoted lung metastasis and LY294002 could inhibit it in vivo. In summary, our in vitro and in vivo results indicate that Nir1 binding to CCL18 plays an important role in breast cancer invasion/metastasis. This study identified both Nir1 and CCL18 as potential anti-invasion targets for therapeutic intervention in breast cancer.
Collapse
Affiliation(s)
- Baogang Zhang
- Department of Pathology, Key Clinical Specialty for Pathology of Shandong Province, Affiliated Hospital of Weifang Medical University, Weifang 261053, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Knockdown a water channel protein, aquaporin-4, induced glioblastoma cell apoptosis. PLoS One 2013; 8:e66751. [PMID: 23950863 PMCID: PMC3741385 DOI: 10.1371/journal.pone.0066751] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 05/10/2013] [Indexed: 01/03/2023] Open
Abstract
Glioblastomas are the most aggressive forms of primary brain tumors due to their tendency to invade surrounding healthy brain tissues, rendering them largely incurable. The water channel protein, Aquaporin-4 (AQP4) is a key molecule for maintaining water and ion homeostasis in the central nervous system and has recently been reported with cell survival except for its well-known function in brain edema. An increased AQP4 expression has been demonstrated in glioblastoma multiforme (GBM), suggesting it is also involved in malignant brain tumors. In this study, we show that siRNA-mediated down regulation of AQP4 induced glioblastoma cell apoptosis in vitro and in vivo. We further show that several apoptotic key proteins, Cytochrome C, Bcl-2 and Bad are involved in AQP4 signaling pathways. Our results indicate that AQP4 may serve as an anti-apoptosis target for therapy of glioblastoma.
Collapse
|
39
|
Li H, Yin C, Zhang B, Sun Y, Shi L, Liu N, Liang S, Lu S, Liu Y, Zhang J, Li F, Li W, Liu F, Sun L, Qi Y. PTTG1 promotes migration and invasion of human non-small cell lung cancer cells and is modulated by miR-186. Carcinogenesis 2013; 34:2145-55. [PMID: 23671127 DOI: 10.1093/carcin/bgt158] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Deeper mechanistic understanding of non-small cell lung cancer (NSCLC), a leading cause of total cancer-related deaths, may facilitate the establishment of more effective therapeutic strategies. In this study, pituitary tumor transforming gene (PTTG1) expression was associated with lymph node and distant metastasis in patients with NSCLC and was correlated with patient survival. Reduction of PTTG1 by small interfering RNA (siRNA) inhibits the migration and invasion of NSCLC cells by mediating matrix metalloproteinases expression. To the best of our knowledge, this study is the first to report that PTTG1 promotes epidermal growth factor (EGF) induced the phosphorylation of LIN-11, Isl1 and MEC-3 protein domain kinase and cofilin, a critical step in cofilin recycling and actin polymerization. Additionally, EGF-induced Akt phosphorylation was suppressed through knockdown of PTTG1. Interestingly, miR-186 can modulate PTTG1 protein expression. As observed from the animal experiment in this study, knockdown of PTTG1 through siRNA and overexpression of miR-186 inhibited invasive activity of NSCLC cells toward the SCID mice lung. In summary, our in vitro and in vivo results indicate that PTTG1 modulated by miR-186 has an important function in NSCLC invasion/metastasis. This study identified both PTTG1 and miR-186 as potential anti-invasion targets for therapeutic intervention in NSCLC.
Collapse
Affiliation(s)
- Hongli Li
- Department of Pathology, Weifang Medical University, Weifang 261053, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lindemann O, Umlauf D, Frank S, Schimmelpfennig S, Bertrand J, Pap T, Hanley PJ, Fabian A, Dietrich A, Schwab A. TRPC6 regulates CXCR2-mediated chemotaxis of murine neutrophils. THE JOURNAL OF IMMUNOLOGY 2013; 190:5496-505. [PMID: 23636057 DOI: 10.4049/jimmunol.1201502] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Unraveling the mechanisms involved in chemotactic navigation of immune cells is of particular interest for the development of new immunoregulatory therapies. It is generally agreed upon that members of the classical transient receptor potential channel family (TRPC) are involved in chemotaxis. However, the regulatory role of TRPC channels in chemoattractant receptor-mediated signaling has not yet been clarified in detail. In this study, we demonstrate that the TRPC6 channels play a pronounced role in CXCR2-mediated intermediary chemotaxis, whereas N-formyl-methionine-leucine-phenylalanine receptor-mediated end-target chemotaxis is TRPC6 independent. The knockout of TRPC6 channels in murine neutrophils led to a strongly impaired intermediary chemotaxis after CXCR2 activation which is not further reinforced by CXCR2, PI3K, or p38 MAPK inhibition. Furthermore, CXCR2-mediated Ca(2+) influx but not Ca(2+) store release was attenuated in TRPC6(-/-) neutrophils. We demonstrate that the TRPC6 deficiency affected phosphorylation of AKT and MAPK downstream of CXCR2 receptor activation and led to altered remodeling of actin. The relevance of this TRPC6-depending defect in neutrophil chemotaxis is underscored by our in vivo findings. A nonseptic peritoneal inflammation revealed an attenuated recruitment of neutrophils in the peritoneal cavity of TRPC6(-/-) mice. In summary, this paper defines a specific role of TRPC6 channels in CXCR2-induced intermediary chemotaxis. In particular, TRPC6-mediated supply of calcium appears to be critical for activation of downstream signaling components.
Collapse
Affiliation(s)
- Otto Lindemann
- Institute of Physiology II, Westfälische Wilhelms-University, 48149 Münster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Deltex-1 activates mitotic signaling and proliferation and increases the clonogenic and invasive potential of U373 and LN18 glioblastoma cells and correlates with patient survival. PLoS One 2013; 8:e57793. [PMID: 23451269 PMCID: PMC3581491 DOI: 10.1371/journal.pone.0057793] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 01/29/2013] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma (GBM) is a highly malignant primary tumor of the central nervous system originating in glial cells. GBM results in more years of life lost than any other cancer type. Low levels of Notch receptor expression correlates with prolonged survival in various high grade gliomas independent of other markers. Different downstream pathways of Notch receptors have been identified. We tested if the Notch/Deltex pathway, which is distinct from the canonical, CSL-mediated pathway, has a role in GBM. We show that the alternative or non-canonical Notch pathway functioning through Deltex1 (DTX1) mediates key features of glioblastoma cell aggressiveness. For example, DTX1 activates the RTK/PI3K/PKB and the MAPK/ERK mitotic pathways and induces anti-apoptotic Mcl-1. The clonogenic and growth potential of established glioma cells correlated with DTX1 levels. Microarray gene expression analysis further identified a DTX1-specific, MAML1-independent transcriptional program - including microRNA-21- which is functionally linked to the changes in tumor cell aggressiveness. Over-expression of DTX1 increased cell migration and invasion correlating to ERK activation, miR-21 levels and endogenous Notch levels. In contrast to high and intermediate expressors, patients with low DTX1 levels have a more favorable prognosis. The alternative Notch pathway via DTX1 appears to be an oncogenic factor in glioblastoma and these findings offer new potential therapeutic targets.
Collapse
|
42
|
Lu S, Niu N, Guo H, Tang J, Guo W, Liu Z, Shi L, Sun T, Zhou F, Li H, Zhang J, Zhang B. ARK5 promotes glioma cell invasion, and its elevated expression is correlated with poor clinical outcome. Eur J Cancer 2013; 49:752-63. [PMID: 23063350 DOI: 10.1016/j.ejca.2012.09.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 09/17/2012] [Accepted: 09/17/2012] [Indexed: 12/30/2022]
|
43
|
Signaling determinants of glioma cell invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 986:121-41. [PMID: 22879067 DOI: 10.1007/978-94-007-4719-7_7] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor cell invasiveness is a critical challenge in the clinical management of glioma patients. In addition, there is accumulating evidence that current therapeutic modalities, including anti-angiogenic therapy and radiotherapy, can enhance glioma invasiveness. Glioma cell invasion is stimulated by both autocrine and paracrine factors that act on a large array of cell surface-bound receptors. Key signaling elements that mediate receptor-initiated signaling in the regulation of glioblastoma invasion are Rho family GTPases, including Rac, RhoA and Cdc42. These GTPases regulate cell morphology and actin dynamics and stimulate cell squeezing through the narrow extracellular spaces that are typical of the brain parenchyma. Transient attachment of cells to the extracellular matrix is also necessary for glioblastoma cell invasion. Interactions with extracellular matrix components are mediated by integrins that initiate diverse intracellular signalling pathways. Key signaling elements stimulated by integrins include PI3K, Akt, mTOR and MAP kinases. In order to detach from the tumor mass, glioma cells secrete proteolytic enzymes that cleave cell surface adhesion molecules, including CD44 and L1. Key proteases produced by glioma cells include uPA, ADAMs and MMPs. Increased understanding of the molecular mechanisms that control glioma cell invasion has led to the identification of molecular targets for therapeutic intervention in this devastating disease.
Collapse
|
44
|
Cui Y, Wang Q, Wang J, Dong Y, Luo C, Hu G, Lu Y. Knockdown of AKT2 expression by RNA interference inhibits proliferation, enhances apoptosis, and increases chemosensitivity to the anticancer drug VM-26 in U87 glioma cells. Brain Res 2012; 1469:1-9. [PMID: 22771706 DOI: 10.1016/j.brainres.2012.06.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 06/06/2012] [Accepted: 06/27/2012] [Indexed: 10/28/2022]
Abstract
The AKT2 kinase (protein kinas Bβ) is frequently overexpressed in malignant gliomas. In this study, the human glioblastoma cell line U87 was stably transfected with a lentivirus vector expressing a short hairpin RNA (shRNA) targeting AKT2. Knockdown of AKT2 by the shRNA inhibited U87 cell proliferation and increased the rate of apoptosis. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blot analysis revealed that cells stably underexpressing AKT2 showed lower expression of the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2) and enhanced expression of the apoptosis effector caspase-3 compared to U87 cells stably transfected with a control vector. Furthermore, expression levels of AKT2 were correlated with the IC50 of the antitumor drug VM-26 (teniposide); the VM-26 IC50 was reduced from 6.46±0.42μg/ml in control glioma cells to 1.15±0.22μg/ml in U87 cells underexpressing AKT2. Combined AKT2 knockdown and VM-26 treatment inhibited cell proliferation in vitro more effectively than either treatment alone. Knockdown of AKT2 expression was associated with decreased expression of the multidrug resistance-associated protein 1 (MRP1) without affecting MRP1 mRNA expression. However, the mRNA and protein levels of MDR1 (p-glycoprotein) were unaffected by AKT2 knockdown. These results indicate that inhibition of AKT2 expression may be an effective means for overcoming AKT2-associated chemoresistance in human malignant glioma cells and may represent a potential gene-targeting approach to treat glioma.
Collapse
Affiliation(s)
- Yong Cui
- Department of Neurosurgery, Changzheng Hospital, Second Military Medical University, No. 415 FengYang Road, Shanghai 200003, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
45
|
Zitzmann K, Vlotides G, Brand S, Lahm H, Spöttl G, Göke B, Auernhammer CJ. Perifosine-mediated Akt inhibition in neuroendocrine tumor cells: role of specific Akt isoforms. Endocr Relat Cancer 2012; 19:423-34. [PMID: 22499437 DOI: 10.1530/erc-12-0074] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system show aberrant Akt activity. Several inhibitors of the phosphoinositide 3-kinase (PI(3)K)-Akt-mTOR signaling pathway are currently being evaluated in clinical phase II and III studies for the treatment of NETs with promising results. However, the molecular mechanisms and particularly the role of different Akt isoforms in NET signaling are not fully understood. In this study, we examine the effect of Akt inhibition on NET cells of heterogeneous origin. We show that the Akt inhibitor perifosine effectively inhibits Akt phosphorylation and cell viability in human pancreatic (BON1), bronchus (NCI-H727), and midgut (GOT1) NET cells. Perifosine treatment suppressed the phosphorylation of Akt downstream targets such as GSK3α/β, MDM2, and p70S6K and induced apoptosis. To further investigate the role of individual Akt isoforms for NET cell function, we specifically blocked Akt1, Akt2, and Akt3 via siRNA transfection. In contrast to Akt2 knockdown, knockdown of Akt isoforms 1 and 3 decreased phosphorylation levels of GSK3α/β, MDM2, and p70S6K and suppressed NET cell viability and colony-forming capacity. The inhibitory effect of simultaneous downregulation of Akt1 and Akt3 on tumor cell viability was significantly stronger than that caused by downregulation of all Akt isoforms, suggesting a particular role for Akt1 and Akt3 in NET signaling. Akt3 siRNA-induced apoptosis while all three isoform-specific siRNAs impaired BON1 cell invasion. Together, our data demonstrate potent antitumor effects of the pan-Akt inhibitor perifosine on NET cells in vitro and suggest that selective targeting of Akt1 and/or Akt3 might improve the therapeutic potential of Akt inhibition in NET disease.
Collapse
Affiliation(s)
- Kathrin Zitzmann
- Department of Internal Medicine II, University-Hospital Munich-Grosshadern, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
46
|
PTEN regulates PDGF ligand switch for β-PDGFR signaling in prostate cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:1017-1027. [PMID: 22209699 DOI: 10.1016/j.ajpath.2011.11.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 10/10/2011] [Accepted: 11/28/2011] [Indexed: 12/29/2022]
Abstract
Platelet-derived growth factor (PDGF) family members are potent growth factors that regulate cell proliferation, migration, and transformation. Clinical studies have shown that both PDGF receptor β (β-PDGFR) and its ligand PDGF D are up-regulated in primary prostate cancers and bone metastases, whereas PDGF B, a classic ligand for β-PDGFR, is not frequently detected in clinical samples. In this study, we examined the role of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN) in the regulation of PDGF expression levels using both a prostate-specific, conditional PTEN-knockout mouse model and mouse prostate epithelial cell lines established from these mice. We found an increase in PDGF D and β-PDGFR expression levels in PTEN-null tumor cells, accompanied by a decrease in PDGF B expression. Among Akt isoforms, increased Akt3 expression was most prominent in mouse PTEN-null cells, and phosphatidylinositol 3-kinase/Akt activity was essential for the maintenance of increased PDGF D and β-PDGFR expression. In vitro deletion of PTEN resulted in a PDGF ligand switch from PDGF B to PDGF D in normal mouse prostate epithelial cells, further demonstrating that PTEN regulates this ligand switch. Similar associations between PTEN status and PDGF isoforms were noted in human prostate cancer cell lines. Taken together, these results suggest a mechanism by which loss of PTEN may promote prostate cancer progression via PDGF D/β-PDGFR signal transduction.
Collapse
|
47
|
Natsume A, Kato T, Kinjo S, Enomoto A, Toda H, Shimato S, Ohka F, Motomura K, Kondo Y, Miyata T, Takahashi M, Wakabayashi T. Girdin maintains the stemness of glioblastoma stem cells. Oncogene 2011; 31:2715-24. [PMID: 22020337 DOI: 10.1038/onc.2011.466] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glioblastomas (GBMs) are the most common and aggressive type of brain tumor. GBMs usually show hyperactivation of the PI3K-Akt pathway, a pro-tumorigenic signaling cascade that contributes to pathogenesis. Girdin, an actin-binding protein identified as a novel substrate of Akt, regulates the sprouting of axons and the migration of neural progenitor cells during early postnatal-stage neurogenesis in the hippocampus. Here, we show that Girdin is highly expressed in human glioblastoma (GBM). Stable Girdin knockdown in isolated GBM stem cells resulted in decreased expression of stem cell markers, including CD133, induced multilineage neural differentiation, and inhibited in vitro cell motility, ex vivo invasion, sphere-forming capacity and in vivo tumor formation. Furthermore, exogenous expression of the Akt-binding domain of Girdin, which competitively inhibits its Akt-mediated phosphorylation, diminished the expression of stem cell markers, SOX2 and nestin, and migration on the brain slice and induced the expression of neural differentiation markers glial fibrillary acidic protein/βIII Tubulin. Our results reveal that Girdin is required for GBM-initiating stem cells to sustain the stemness and invasive properties.
Collapse
Affiliation(s)
- A Natsume
- Department of Neurosurgery, Nagoya University School of Medicine, Showa-ku, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wani R, Bharathi NS, Field J, Tsang AW, Furdui CM. Oxidation of Akt2 kinase promotes cell migration and regulates G1-S transition in the cell cycle. Cell Cycle 2011; 10:3263-8. [PMID: 21957489 DOI: 10.4161/cc.10.19.17738] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Phosphorylation has long been recognized as the key mediator of protein signaling. New modes of signaling regulation are emerging with the development of specific chemical probes and application of high-throughput mass spectrometry technologies. Using biotin-tagged chemical probes for protein oxidation, mass spectrometry and functional assays, our group has recently reported isoform-specific oxidation of Akt2 in response to PDGF signaling. The studies included here investigate the functional consequence of oxidation on Akt2-mediated cell migration and cell cycle. Akt2-KO MEFs transduced with WT and Cys124Ser Akt2 were used as the model system for these studies. The implications of these findings on disease pathology are discussed.
Collapse
Affiliation(s)
- Revati Wani
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | | |
Collapse
|
49
|
Li X, Mikhalkova D, Gao E, Zhang J, Myers V, Zincarelli C, Lei Y, Song J, Koch WJ, Peppel K, Cheung JY, Feldman AM, Chan TO. Myocardial injury after ischemia-reperfusion in mice deficient in Akt2 is associated with increased cardiac macrophage density. Am J Physiol Heart Circ Physiol 2011; 301:H1932-40. [PMID: 21890689 DOI: 10.1152/ajpheart.00755.2010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Akt2 protein kinase has been shown to promote cell migration and actin polymerization in several cell types, including macrophages. Because migrating macrophages constitute an important inflammatory response after myocardial ischemia, we determined cardiac macrophage expression after ischemia-reperfusion (I/R) injury and cryo-injury in mice lacking Akt2 (Akt2-KO). At 7 days post-I/R, Akt2-KO cardiac tissues showed an increase in immunohistochemical staining for macrophage markers (Galectin 3 and F4/80) compared with wild-type (WT) mice, indicating macrophage density was increased in the injured Akt2-KO myocardium. This change was time dependent because macrophage density was similar between WT and Akt2-KO myocardium at 3 days post-I/R, but by 7 and 14 days post-I/R, macrophage density was significantly increased in Akt2-KO myocardium. Concomitantly, infarct size was larger and cardiac function was reduced in Akt2-KO mice subjected to I/R. However, when cryo-infarction produced similar infarct sizes in the anterior wall in both WT and Akt2-KO mice, macrophage density remained higher in Akt2-KO mouse myocardium, suggesting Akt2 regulates myocardial macrophage density independent of infarct size. Consistently, bone marrow from Akt2-KO mice enhanced myocardial macrophage density in both C57/B6 WT and Akt2-KO recipient mice. Finally, reciprocal ex-vivo coculturing of macrophages and cardiac myocytes showed that activated Akt2-KO peritoneal macrophages had reduced mobility and adhesion when compared with WT littermate controls. Thus, although Akt-2 KO mice did not affect the initial inflammation response after injury and Akt2 deficiency has been shown to impair cell migration or motility in macrophages, our data suggested a novel mechanism in which increasing retention of Akt2-KO macrophages resulted in increasing cardiac Akt2-KO macrophage density in the myocardial space.
Collapse
Affiliation(s)
- Xue Li
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Polisetty RV, Gupta MK, Nair SC, Ramamoorthy K, Tiwary S, Shiras A, Chandak GR, Sirdeshmukh R. Glioblastoma cell secretome: Analysis of three glioblastoma cell lines reveal 148 non-redundant proteins. J Proteomics 2011; 74:1918-25. [PMID: 21601021 DOI: 10.1016/j.jprot.2011.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 04/15/2011] [Accepted: 05/03/2011] [Indexed: 12/24/2022]
|