1
|
Xu J, Guo Y, Ning W, Wang J, Chen Y, Liu D, Yang J, Song Y, Zhang H. Comprehensive analysis of heat shock proteins in glioma revealed the association with glioma-associated myeloid cells. Genes Immun 2025:10.1038/s41435-025-00327-5. [PMID: 40234584 DOI: 10.1038/s41435-025-00327-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
In the central nervous system, glioma stands as the predominant primary brain tumor. Heat shock proteins exerted a critical influence on tumor progression and tumor immune microenvironment. However, research on heat shock proteins in glioma remained ambiguous. We analyzed data from the CPTAC, TCGA, and GTEx databases, identifying seven heat shock protein genes critical to glioma prognosis. Subsequently, through Lasso regression, a model based on heat shock protein genes (DNAJC7, DNAJC12, HSPB2, HSP90B1, HSPA5) was constructed. And the risk score showed a positive correlation to the immune score. Further investigation into immune cells revealed that HSPA5 and HSP90B1 were expressed at higher levels in glioma and significantly linked to M2 macrophage infiltration. Considering the limited research on HSP90B1 in glioma, we further revealed that HSP90B1 might have a connection with two crucial signaling pathways within tumors: PI3K/AKT and Wnt/β-catenin. Given that lactate could promote the M2 polarization of macrophages, we further found that HSP90B1 could enhance the transcription of glycolysis-related genes, including LDHA. Overall, our study demonstrated that heat shock protein genes were significantly linked to glioma patient prognosis. Additionally, we observed that HSP90B1 had a significant relationship with M2 macrophage infiltration and potentially regulated LDHA level in glioma.
Collapse
Affiliation(s)
- Jiacheng Xu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yuduo Guo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University/China International Neuroscience Institute (China-INI), Beijing, China
| | - Weihai Ning
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jun Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yujia Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Deshan Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jingjing Yang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Zhao J, Yang S, Xu Y, Qin S, Bie F, Chen L, Zhou F, Xie J, Liu X, Shu B, Qi S. Mechanical pressure-induced dedifferentiation of myofibroblasts inhibits scarring via SMYD3/ITGBL1 signaling. Dev Cell 2023:S1534-5807(23)00190-9. [PMID: 37192621 DOI: 10.1016/j.devcel.2023.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/21/2022] [Accepted: 04/24/2023] [Indexed: 05/18/2023]
Abstract
Pressure therapy (PT) is an effective intervention for reducing scarring, but its underlying mechanism remains largely unclear. Here, we demonstrate that human scar-derived myofibroblasts dedifferentiate into normal fibroblasts in response to PT, and we identify how SMYD3/ITGBL1 contributes to the nuclear relay of mechanical signals. In clinical specimens, reductions in SMYD3 and ITGBL1 expression levels are strongly associated with the anti-scarring effects of PT. The integrin β1/ILK pathway is inhibited in scar-derived myofibroblasts upon PT, leading to decreased TCF-4 and subsequently to reductions in SMYD3 expression, which reduces the levels of H3K4 trimethylation (H3K4me3) and further suppresses ITGBL1 expression, resulting the dedifferentiation of myofibroblasts into fibroblasts. In animal models, blocking SMYD3 expression results in reductions of scarring, mimicking the positive effects of PT. Our results show that SMYD3 and ITGBL1 act as sensors and mediators of mechanical pressure to inhibit the progression of fibrogenesis and provide therapeutic targets for fibrotic diseases.
Collapse
Affiliation(s)
- Jingling Zhao
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shuai Yang
- Department of Neurosurgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Yingbin Xu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Shitian Qin
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Fan Bie
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Lei Chen
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Fei Zhou
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Julin Xie
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Xusheng Liu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Bin Shu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| | - Shaohai Qi
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
3
|
Qiao P, Tian Z. Atractylenolide I inhibits EMT and enhances the antitumor effect of cabozantinib in prostate cancer via targeting Hsp27. Front Oncol 2023; 12:1084884. [PMID: 36686743 PMCID: PMC9853281 DOI: 10.3389/fonc.2022.1084884] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
Objective To investigate the effect of Hsp27 and the inhibitory effect of Atractylenolide I (ATL-1) on the proliferation of prostate cancer cell DU145 and PC-3. Methods MTT assay was used to detect the inhibitory effect of silencing Hsp27 and ATL-1 on DU145 and PC-3 proliferation of prostate cancer cells. TUNEL detected the apoptosis rate of prostate cancer cell DU145 and PC-3 after silencing Hsp27 and ATL-1 treated. qRT-PCR was used to detect the changes of apoptosis related genes caspase-3, PARP, Bax and Bcl-2 in prostate cancer cell DU145 and PC-3 after the effect of silencing Hsp27 and ATL-1 treated. At the same time, the antitumor effect of ATL-1 combined with cabozantinib was analyzed. Results Hsp27 was highly expressed in human prostate cancer. MTT assay showed that ATL-1 inhibited the proliferation of prostate cancer cells DU145 and PC-3 compared with the control group. TUNEL results showed that silencing Hsp27 and ATL-1 treated could significantly promote the apoptosis of prostate cancer cells DU145 and PC-3 compared with the control group. qRT-PCR results showed that compared with the control group, ATL-1 could promote the expression of caspase-3, PARP and Bax in DU145 and PC-3 prostate cancer cells. Inhibition of Hsp27 by ATL-1 reduced cell viability and induced apoptosis. ATL-1 inhibits the antitumor effect of Hsp27 - enhanced cabozantinib. Hsp27 regulates eIF4E and mediates cell protection. Conclusion Silencing Hsp27 inhibits EMT. ATL-1 can inhibit the malignant evolution of prostate cancer cells by inhibiting Hsp27/eIF4E. ATL-1 also enhanced chemosensitization of cabozantinib in prostate cancer.
Collapse
Affiliation(s)
- Pengfei Qiao
- The Department of Urology Surgery, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhentao Tian
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China,*Correspondence: Zhentao Tian,
| |
Collapse
|
4
|
Kim H, Gomez-Pastor R. HSF1 and Its Role in Huntington's Disease Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:35-95. [PMID: 36396925 PMCID: PMC12001818 DOI: 10.1007/5584_2022_742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW Heat shock factor 1 (HSF1) is the master transcriptional regulator of the heat shock response (HSR) in mammalian cells and is a critical element in maintaining protein homeostasis. HSF1 functions at the center of many physiological processes like embryogenesis, metabolism, immune response, aging, cancer, and neurodegeneration. However, the mechanisms that allow HSF1 to control these different biological and pathophysiological processes are not fully understood. This review focuses on Huntington's disease (HD), a neurodegenerative disease characterized by severe protein aggregation of the huntingtin (HTT) protein. The aggregation of HTT, in turn, leads to a halt in the function of HSF1. Understanding the pathways that regulate HSF1 in different contexts like HD may hold the key to understanding the pathomechanisms underlying other proteinopathies. We provide the most current information on HSF1 structure, function, and regulation, emphasizing HD, and discussing its potential as a biological target for therapy. DATA SOURCES We performed PubMed search to find established and recent reports in HSF1, heat shock proteins (Hsp), HD, Hsp inhibitors, HSF1 activators, and HSF1 in aging, inflammation, cancer, brain development, mitochondria, synaptic plasticity, polyglutamine (polyQ) diseases, and HD. STUDY SELECTIONS Research and review articles that described the mechanisms of action of HSF1 were selected based on terms used in PubMed search. RESULTS HSF1 plays a crucial role in the progression of HD and other protein-misfolding related neurodegenerative diseases. Different animal models of HD, as well as postmortem brains of patients with HD, reveal a connection between the levels of HSF1 and HSF1 dysfunction to mutant HTT (mHTT)-induced toxicity and protein aggregation, dysregulation of the ubiquitin-proteasome system (UPS), oxidative stress, mitochondrial dysfunction, and disruption of the structural and functional integrity of synaptic connections, which eventually leads to neuronal loss. These features are shared with other neurodegenerative diseases (NDs). Currently, several inhibitors against negative regulators of HSF1, as well as HSF1 activators, are developed and hold promise to prevent neurodegeneration in HD and other NDs. CONCLUSION Understanding the role of HSF1 during protein aggregation and neurodegeneration in HD may help to develop therapeutic strategies that could be effective across different NDs.
Collapse
Affiliation(s)
- Hyuck Kim
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Parma B, Wurdak H, Ceppi P. Harnessing mitochondrial metabolism and drug resistance in non-small cell lung cancer and beyond by blocking heat-shock proteins. Drug Resist Updat 2022; 65:100888. [DOI: 10.1016/j.drup.2022.100888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/10/2022] [Accepted: 10/25/2022] [Indexed: 11/30/2022]
|
6
|
The kinase activity of integrin-linked kinase regulates cellular senescence in gastric cancer. Cell Death Dis 2022; 13:577. [PMID: 35778385 PMCID: PMC9249761 DOI: 10.1038/s41419-022-05020-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 01/21/2023]
Abstract
The activity of integrin-linked kinase (ILK) in cancerous cells is often oncogenic and associated with malignant properties, such as uncontrolled cell cycle progression and evasion from senescence. However, the role of ILK in cellular senescence in gastric cancer (GC) has not been previously examined. We generated single-cell clones of ILK knock-out using CRISPR-Cas9 in human GC lines with mesenchymal or epithelial histology. Cells with no residual ILK expression exhibited strong cellular senescence with diminished clathrin-mediated endocytosis, Surprisingly, ILK loss-induced cellular senescence appeared to be independent of its function in integrin signaling. The low dose of CPD22, a small molecule inhibitor of ILK activity-induced senescence in three GC cell lines with different histologies. Furthermore, senescent cells with ILK depletion transfected with N-terminal truncated ILK mutant remaining catalytic domains displayed the reduction of senescent phenotypes. RNA sequencing and cytokine array results revealed the enrichment of multiple pro-inflammatory signaling pathways in GC lines in the absence of ILK. Our study identified the important role and the potential mechanism of ILK in the cellular senescence of cancerous epithelial cells. The inhibition of ILK activity using small molecule compounds could have a pro-senescent effect as a therapeutic option for GC.
Collapse
|
7
|
Katopodis P, Kerslake R, Zikopoulos A, Beri N, Anikin V. p38β - MAPK11 and its role in female cancers. J Ovarian Res 2021; 14:84. [PMID: 34174910 PMCID: PMC8236201 DOI: 10.1186/s13048-021-00834-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
Background The p38MAPK family of Mitogen Activated Protein Kinases are a group of signalling molecules involved in cell growth, survival, proliferation and differentiation. The widely studied p38α isoform is ubiquitously expressed and is implicated in a number of cancer pathologies, as are p38γ and p38δ. However, the mechanistic role of the isoform, p38β, remains fairly elusive. Recent studies suggest a possible role of p38β in both breast and endometrial cancer with research suggesting involvement in bone metastasis and cancer cell survival. Female tissue specific cancers such as breast, endometrial, uterine and ovary account for over 3,000,000 cancer related incidents annually; advancements in therapeutics and treatment however require a deeper understanding of the molecular aetiology associated with these diseases. This study provides an overview of the MAPK signalling molecule p38β (MAPK11) in female cancers using an in-silico approach. Methods A detailed gene expression and methylation analysis was performed using datasets from cBioportal, CanSar and MEXPRESS. Breast, Uterine Endometrial, Cervical, Ovarian and Uterine Carcinosarcoma TCGA cancer datasets were used and analysed. Results Data using cBioportal and CanSAR suggest that expression of p38β is lower in cancers: BRCA, UCEC, UCS, CESC and OV compared to normal tissue. Methylation data from SMART and MEXPRESS indicate significant probe level variation of CpG island methylation status of the gene MAPK11. Analysis of the genes’ two CpG islands shows that the gene was hypermethylated in the CpG1 with increased methylation seen in BRCA, CESC and UCEC cancer data sets with a slight increase of expression recorded in cancer samples. CpG2 exhibited hypomethylation with no significant difference between samples and high levels of expression. Further analysis from MEXPRESS revealed no significance between probe methylation and altered levels of expression. In addition, no difference in the expression of BRCA oestrogen/progesterone/HER2 status was seen. Conclusion This data provides an overview of the expression of p38β in female tissue specific cancers, showing a decrease in expression of the gene in BRCA, UCEC, CESC, UCS and OV, increasing the understanding of p38β MAPK expression and offering insight for future in-vitro investigation and therapeutic application. Supplementary Information The online version contains supplementary material available at 10.1186/s13048-021-00834-9.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK. .,Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London, UB9 6JH, UK.
| | - Rachel Kerslake
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
| | - Athanasios Zikopoulos
- Obstetrics and Gynaecology Department, Royal Cornwall Hospitals NHS Foundation Trust, Royal Cornwall Hospital, Truro, TR1 3LJ, UK
| | - Nefeli Beri
- Department of Medicine, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London, UB9 6JH, UK.,Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, Moscow, Russian Federation, 119146
| |
Collapse
|
8
|
Lu W, Wang Y, Gan M, Duan Q. Prognosis and predictive value of heat-shock proteins expression in oral cancer: A PRISMA-compliant meta-analysis. Medicine (Baltimore) 2021; 100:e24274. [PMID: 33546049 PMCID: PMC7837937 DOI: 10.1097/md.0000000000024274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/11/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Heat-shock proteins (HSP) is a key chaperone protein which maintains intracellular proteostasis and is expressed on the surface of solid and hematological malignancies. Several studies have reported paradoxical evidence of the association between HSP expression and prognosis of oral cancer. To address the discrepancy, we carried out the meta-analysis to assess the role of HSP such as: HSP70, HSP90, HSP27, HSP60, and HSP105 in susceptibility, progression, and prognosis of oral cancer. MATERIALS AND METHODS We retrieved the PubMed, Embase, Web of science, China National Knowledge Infrastructure (CNKI), and Wanfang databases to acquire the eligible studies which were associated with HSP70, HSP90, HSP27, HSP60, and HSP105 protein expression and oral cancer. We applied hazard ratio (HR) and its 95% confidence interval (95% CI) to assess the value of HSP protein expression in overall survival of oral cancer; odds ratio (OR) and its 95% CI were used to evaluate the association of risk and clinical features of oral cancer. Funnel plot, Begg test, and Egger line regression test were utilized to observe publication bias among studies. All statistical analysis was performed with Stata 14.0 software (Stata Corporation, College Station, TX). RESULTS A total of 26 studies were included in the present meta-analysis. On based of the results, HSP70 and HSP27 had no significant association with progression of oral cancer. However, the pooled HR and 95% CI revealed a significant well effects of HSP70 and HSP27 expression on survival of oral cancer. Moreover, the susceptibility of oral cancer was significantly associated with HSP70 and HSP60 overexpression. CONCLUSION HSP70 and HSP27 protein overexpression might be valuable biomarkers for the prognosis of oral cancer. And HSP70 and HSP60 might have potential predictive effects on the risk of oral cancer.
Collapse
|
9
|
Roche O, Fernández-Aroca DM, Arconada-Luque E, García-Flores N, Mellor LF, Ruiz-Hidalgo MJ, Sánchez-Prieto R. p38β and Cancer: The Beginning of the Road. Int J Mol Sci 2020; 21:ijms21207524. [PMID: 33053909 PMCID: PMC7589630 DOI: 10.3390/ijms21207524] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/08/2020] [Accepted: 10/10/2020] [Indexed: 12/26/2022] Open
Abstract
The p38 mitogen-activated protein kinase (MAPK) signaling pathway is implicated in cancer biology and has been widely studied over the past two decades as a potential therapeutic target. Most of the biological and pathological implications of p38MAPK signaling are often associated with p38α (MAPK14). Recently, several members of the p38 family, including p38γ and p38δ, have been shown to play a crucial role in several pathologies including cancer. However, the specific role of p38β (MAPK11) in cancer is still elusive, and further investigation is needed. Here, we summarize what is currently known about the role of p38β in different types of tumors and its putative implication in cancer therapy. All evidence suggests that p38β might be a key player in cancer development, and could be an important therapeutic target in several pathologies, including cancer.
Collapse
Affiliation(s)
- Olga Roche
- Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (O.R.); (D.M.F.-A.); (E.A.-L.); (N.G.-F.); (L.F.M.); (M.J.R.-H.)
- Departamento de Ciencias Médicas, Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Diego M. Fernández-Aroca
- Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (O.R.); (D.M.F.-A.); (E.A.-L.); (N.G.-F.); (L.F.M.); (M.J.R.-H.)
| | - Elena Arconada-Luque
- Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (O.R.); (D.M.F.-A.); (E.A.-L.); (N.G.-F.); (L.F.M.); (M.J.R.-H.)
| | - Natalia García-Flores
- Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (O.R.); (D.M.F.-A.); (E.A.-L.); (N.G.-F.); (L.F.M.); (M.J.R.-H.)
| | - Liliana F. Mellor
- Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (O.R.); (D.M.F.-A.); (E.A.-L.); (N.G.-F.); (L.F.M.); (M.J.R.-H.)
| | - María José Ruiz-Hidalgo
- Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (O.R.); (D.M.F.-A.); (E.A.-L.); (N.G.-F.); (L.F.M.); (M.J.R.-H.)
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Ricardo Sánchez-Prieto
- Departamento de Ciencias Médicas, Facultad de Medicina de Albacete, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Consejo Superior de Investigaciones Cientificas, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-915-854-420
| |
Collapse
|
10
|
Zhao Y, Wu H, Xing X, Ma Y, Ji S, Xu X, Zhao X, Wang S, Jiang W, Fang C, Zhang L, Yan F, Wang X. CD13 Induces Autophagy to Promote Hepatocellular Carcinoma Cell Chemoresistance Through the P38/Hsp27/CREB/ATG7 Pathway. J Pharmacol Exp Ther 2020; 374:512-520. [PMID: 32571958 DOI: 10.1124/jpet.120.265637] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The chemoresistance of hepatocellular carcinoma (HCC) is a serious problem that directly hinders the effect of chemotherapeutic agents. We previously reported that Aminopeptidase N (CD13) inhibition can enhance the cytotoxic efficacy of chemotherapy agents. In the present study, we use liver cancer cells to explore the molecular mechanism accounting for the relationship between CD13 and chemoresistance. We demonstrate that CD13 overexpression activates the P38/heat shock protein 27/cAMP response element-binding protein (CREB) signaling pathway to limit the efficacy of cytotoxic agents. Moreover, blockade of P38 or CREB sensitizes HCC cells to 5-fluorouracil. Then we reveal that CREB binds to the autophagy related 7 (ATG7) promoter to induce autophagy and promote HCC cell chemoresistance. CD13 inhibition also downregulates the expression of ATG7, autophagy, and tumor cell growth in vivo. Overall, the combination a CD13 inhibitor and chemotherapeutic agents may be a potential strategy for overcoming drug resistance in HCC. SIGNIFICANCE STATEMENT: Our study demonstrates that Aminopeptidase N (CD13) promotes hepatocellular carcinoma (HCC) cell chemoresistance via the P38/heat shock protein 27/cAMP response element-binding protein (CREB) pathway. CREB regulates autophagy related 7 transcription and expression to induce autophagy. Our results collectively suggest that CD13 may serve as a potential target for overcoming HCC resistance.
Collapse
Affiliation(s)
- Yan Zhao
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Huina Wu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Xiaoyan Xing
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Yuqian Ma
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Shengping Ji
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Xinyue Xu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Xin Zhao
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Sensen Wang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Wenyan Jiang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Chunyan Fang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Lei Zhang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Fang Yan
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| | - Xuejian Wang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China (Y.Z., H.W., X.Xi., Y.M., S.J., X.Xu., X.Z., S.W., W.J., C.F., L.Z., F.Y., X.W.) and Department of pharmacy, Southwestern Lu Hospital, Liaocheng, Shandong, China (H.W.)
| |
Collapse
|
11
|
Ning Z, Zhu X, Jiang Y, Gao A, Zou S, Gu C, He C, Chen Y, Ding WQ, Zhou J. Integrin-Linked Kinase Is Involved In the Proliferation and Invasion of Esophageal Squamous Cell Carcinoma. J Cancer 2020; 11:324-333. [PMID: 31897228 PMCID: PMC6930430 DOI: 10.7150/jca.33737] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/18/2019] [Indexed: 11/09/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive type of cancer with high mortality rate in China, largely due to its high invasive and metastatic potential. The purposes of this study are to investigate the potential molecular mechanisms behind the aggressive nature of ESCC and search for new prognostic biomarkers. By employing the quantitative proteomic based strategy, we compared the proteomic profile between three ESCC samples and paired adjacent tissues. After bioinformatics analysis, four candidate proteins were validated in thirteen paired patient samples. Further validation of the key candidate, integrin-linked kinase (ILK), was carried out in one hundred patient samples. The specific inhibitor compound 22 (cpd22) was used to assess the influence of ILK to ESCC cell motility and invasiveness by applying wound-healing and transwell assay. Western blot analysis was performed to elucidate the signaling pathways involved in ILK-mediated ESCC invasion. Total 236 proteins were identified by proteomic analysis. Bioinformatics analysis suggested a key role of the collagen/integrin/ILK signaling pathway during ESCC progression. Further validation indicated that ILK is overexpressed in ESCC tissues and is correlated with poor patient prognosis. Inhibition of ILK kinase activity suppresses proliferation and blocks invasion and migration of ESCC cells. Signaling pathway analysis revealed that ILK regulates AKT phosphorylation on Ser473 but not GSK-3β on Ser9 to promote proliferation and motility of ESCC cells. In conclusion, our results indicated that ILK may play a crucial role in ESCC invasion and metastasis and may serve as a prognostic biomarker and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Zhonghua Ning
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, P.R. China
| | - Xiaozhong Zhu
- Department of Thoracic Surgery, the Affiliated Hospital of the Jiangsu University, Zhenjiang, Jiangsu, P.R. China
| | - Youqin Jiang
- Department of Radiation Oncology, The Third People's Hospital of Yancheng, Yancheng, Jiangsu, P.R. China
| | - Aidi Gao
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Shitao Zou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Chao Gu
- Department of Gastrointestinal surgery, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Chao He
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| | - Yihong Chen
- Department of Radio-Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, P.R. China
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
12
|
Sun M, Zhao W, Chen Z, Li M, Li S, Wu B, Bu R. Circular RNA CEP128 promotes bladder cancer progression by regulating Mir-145-5p/Myd88 via MAPK signaling pathway. Int J Cancer 2019; 145:2170-2181. [PMID: 30939216 DOI: 10.1002/ijc.32311] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 02/09/2019] [Accepted: 03/21/2019] [Indexed: 12/23/2022]
Abstract
The present experiment was designed for exploring the regulatory mechanism of circ-CEP128/miR-145-5p/MYD88 axis in bladder cancer. MiRNAs and circRNAs expression data were derived from Gene Expression Omnibus database with bladder tumor tissues and paracarcinoma tissue samples. Differentially expressed genes in tumor were analyzed via R software. Interaction network of differently expressed miRNAs and differently expressed mRNA was established by means of Cytoscape software. CircCEP128 and miR-145-5p expression levels were determined using qRT-PCR. The expression of MAPK signaling-related proteins MYD88, p38, ERK and JNK was examined by western blot. The relationship between circCEP128 and miR-145-5p was validated using RNA immunoprecipitation. The level of cell propagation and migration was determined by CCK8 and wound healing assay, 5-bromo-2'-deoxyuridine assay and migration assay. Cell apoptosis rate and cell cycle were detected via flow cytometry. Tumor xenograft assay was implemented to investigate the function of circCEP128 in vivo. CircCEP128 and MYD88 were overexpressed in bladder cancer based on microarray analysis and miR-145-5p was a potential targeting factor in bladder cancer. CircCEP128 targeted miR-145-5p and miR-145-5p targeted MYD88. Expression of miR-145-5p was decreased in cancer samples. Knockdown of circCEP128 induced the inhibition of cell viability and mobility and cell cycle arrest. Overexpression of miR-145-5p or knockdown of circCEP128 promoted MAKP signaling pathway and related proteins expression. In addition, knockdown of circCEP128 suppressed the growth of bladder cancer tumor tissues in vivo. Overexpression of circCEP128 promoted bladder cancer progression through modulating miR-145-5p and MYD88 via MAKP signaling pathway.
Collapse
Affiliation(s)
- Ming Sun
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wenyan Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhaofu Chen
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuqiang Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bin Wu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renge Bu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
13
|
Chen J, Bell J, Lau BT, Whittaker T, Stapleton D, Ji HP. A functional CRISPR/Cas9 screen identifies kinases that modulate FGFR inhibitor response in gastric cancer. Oncogenesis 2019; 8:33. [PMID: 31076567 PMCID: PMC6510732 DOI: 10.1038/s41389-019-0145-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/28/2019] [Accepted: 04/17/2019] [Indexed: 01/23/2023] Open
Abstract
Some gastric cancers have FGFR2 amplifications, making them sensitive to FGFR inhibitors. However, cancer cells inevitably develop resistance despite initial response. The underlying resistance mechanism to FGFR inhibition is unclear. In this study, we applied a kinome-wide CRISPR/Cas9 screen to systematically identify kinases that are determinants of sensitivity to a potent FGFR inhibitor AZD4547 in KatoIII cells, a gastric cancer cell line with FGFR2 amplification. In total, we identified 20 kinases, involved in ILK, SRC, and EGFR signaling pathways, as determinants that alter cell sensitivity to FGFR inhibition. We functionally validated the top negatively selected and positively selected kinases, ILK and CSK, from the CRISPR/Cas9 screen using RNA interference. We observed synergistic effects on KatoIII cells as well as three additional gastric cancer cell lines with FGFR2 amplification when AZD4547 was combined with small molecular inhibitors Cpd22 and lapatinib targeting ILK and EGFR/HER2, respectively. Furthermore, we demonstrated that GSK3b is one of the downstream effectors of ILK upon FGFR inhibition. In summary, our study systematically evaluated the kinases and associated signaling pathways modulating cell response to FGFR inhibition, and for the first time, demonstrated that targeting ILK would enhance the effectiveness of AZD4547 treatment of gastric tumors with amplifications of FGFR2.
Collapse
Affiliation(s)
- Jiamin Chen
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - John Bell
- Stanford Genome Technology Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Billy T Lau
- Stanford Genome Technology Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tyler Whittaker
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Darren Stapleton
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Stanford Genome Technology Center, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Min P, Zhao S, Liu L, Zhang Y, Ma Y, Zhao X, Wang Y, Song Y, Zhu C, Jiang H, Gu L, Du J. MICAL-L2 potentiates Cdc42-dependent EGFR stability and promotes gastric cancer cell migration. J Cell Mol Med 2019; 23:4475-4488. [PMID: 31034158 PMCID: PMC6533512 DOI: 10.1111/jcmm.14353] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/29/2019] [Accepted: 04/12/2019] [Indexed: 01/21/2023] Open
Abstract
Enhanced migration potential is a common characteristic of cancer cells induced by mechanisms that are incompletely defined. The present study was designed to investigate relationship of a new discovered cytoskeleton regulator MICAL‐L2 and the endogenous epidermal growth factor receptor (EGFR) signalling pathways in gastric cancer cell migration. Increased expression of MICAL‐L2 in gastric cancer cells up‐regulated EGFR protein level, accompanied by the increase of cell migration, whereas silencing MICAL‐L2 down‐regulated EGFR and inhibited cell migration. Expression of MICAL‐L2 was also shown positively correlated with the activation of HSP27/cytoskeleton and HSP27/β‐catenin signalling pathways that provide key mechanisms controlling cell migration. The up‐regulating effect of MICAL‐L2 on EGFR is mediated through a transcription‐independent mechanism that involves inhibiting EGFR protein degradation in lysosome. Further analysis indicated that Cdc42 activation contributed in maintaining the effect of MICAL‐L2 on EGFR stability. Furthermore analysis of clinic specimens revealed increased expression of MICAL‐L2 in carcinoma tissues and a positive correlation between MICAL‐L2 and EGFR expression levels. The above results indicate that MICAL‐L2 potentiates gastric cell migration via inhibiting EGFR degradation in lysosome via a Cdc42‐dependent manner that leads to the activation of EGFR/HSP27 signalling pathways.
Collapse
Affiliation(s)
- Pengxiang Min
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuo Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yadong Ma
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuyang Zhao
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yueyuan Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yixuan Song
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenchen Zhu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haonan Jiang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Luo Gu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
Rsu1-dependent control of PTEN expression is regulated via ATF2 and cJun. J Cell Commun Signal 2019; 13:331-341. [PMID: 30680530 DOI: 10.1007/s12079-018-00504-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
The Rsu1 protein contributes to cell adhesion and migration via its association with the adaptor complex of Integrin linked kinase (ILK), PINCH, and Parvin (IPP), which binds to the cytoplasmic domain of β1 integrins joining integrins to the actin cytoskeleton. Rsu1 binding to PINCH in the IPP complex is required for EGF-induced adhesion, spreading and migration in MCF10A mammary epithelial cells. In addition, Rsu1 expression inhibits Jun kinase but is necessary for the activation of MKK4 and p38 Map kinase signaling essential for migration in MCF10A cells. The data reported here examines the links between MKK4-p38-ATF2 signaling and AKT regulation in MCF10A cells. Ectopic Rsu1 inhibited AKT1 phosphorylation while Rsu1 depletion induced AKT activation and AKT1 phosphorylation of MKK4 on serine 80, blocking MKK4 activity. Rsu1 depletion also reduced the RNA for lipid phosphatase PTEN thus implicating PTEN in modulating levels of activated AKT in these conditions. ChIP analysis of the PTEN promoter revealed that Rsu1 depletion prevented binding of ATF2 to a positive regulatory site in the PTEN promoter and the enhanced binding of cJun to a negatively regulatory PTEN promoter site. These results demonstrate a mechanism by which Rsu1 adhesion signaling alters the balance between MKK4-p38-ATF2 and cJun activation thus altering PTEN expression in MCF10A cells.
Collapse
|
16
|
Wang Y, Deng W, Zhang Y, Sun S, Zhao S, Chen Y, Zhao X, Liu L, Du J. MICAL2 promotes breast cancer cell migration by maintaining epidermal growth factor receptor (EGFR) stability and EGFR/P38 signalling activation. Acta Physiol (Oxf) 2018; 222. [PMID: 28719045 DOI: 10.1111/apha.12920] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/08/2017] [Accepted: 07/10/2017] [Indexed: 01/08/2023]
Abstract
AIM MICAL2, a cytoskeleton dynamics regulator, is identified associated with survival and metastasis of several types of cancers recently. This study was designed to investigate the role of MICAL2 in breast cancer cell migration as well as its underlying mechanisms. METHODS The relationship between MICAL2 and EGF/EGFR signalling was analysed by gene overexpression and knock-down techniques. Cell migration was measured by wound-healing assays. Activation of EGF/EGFR signalling pathways were evaluated by immunofluorescence, qPCR, Western blotting and zymography techniques. Rac1 activity was assessed by pull-down assay. Correlation of MICAL2 and EGFR in breast cancer specimens was examined by immunohistochemical analysis. RESULTS Ectopic expression of MICAL2 in MCF-7 cells augmented EGFR protein level, accompanied by the promotion of cell migration. Silencing MICAL2 in MDA-MB-231 cells destabilized EGFR and inhibited cell migration. In mechanism, the maintaining effect of MICAL2 on EGFR protein content was due to a delay in EGFR degradation. Expression of MICAL2 was also shown positively correlated with the activation of P38/HSP27 and P38/MMP9 signallings, which are the main downstream signalling cascades of EGF/EGFR involved in cell migration. Further analysis indicated that Rac1 activation contributed to the maintaining effect of MICAL2 on EGFR stability. In addition, analysis of breast cancer specimens revealed a positive correlation between MICAL2 and EGFR levels and an association between MICAL2 expression and worse prognosis. CONCLUSION MICAL2 is a major regulator of breast cancer cell migration, maintaining EGFR stability and subsequent EGFR/P38 signalling activation through inhibiting EGFR degradation in a Rac1-dependent manner.
Collapse
Affiliation(s)
- Y Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - W Deng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Y Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - S Sun
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - S Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Y Chen
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - X Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - L Liu
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - J Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
17
|
Yuan X, Wang W, Li J, Zheng P, Dong P, Chen L, Zhou Y, Xie G, Xu D, Liu Y, Shen L. Gelsolin suppresses gastric cancer metastasis through inhibition of PKR-p38 signaling. Oncotarget 2018; 7:53459-53470. [PMID: 27419625 PMCID: PMC5288199 DOI: 10.18632/oncotarget.10557] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/05/2016] [Indexed: 01/22/2023] Open
Abstract
The biological function of gelsolin in gastric cancer and its mechanism remained undefined. Here, we demonstrated that gelsolin was down-regulated in human gastric cancer tissues, and lower tumorous gelsolin significantly correlated with gastric cancer metastasis. Functionally, gelsolin suppressed the migration of gastric cancer cells in vitro and inhibited lung metastasis in vivo. In mechanism, gelsolin decreased epithelial–mesenchymal transition (EMT) inducing cytoskeleton remolding through inhibition of p38 signaling to suppress the migration of gastric cancer cell. Moreover, gelsolin bound to and decreased the phosphorylation of PKR, and then inhibited p38 signaling pathway. Finally, similar to the gastric cancer cell lines, PKR-p38 signaling pathway proteins tend to be activated and correlated with low expression of gelsolin in clinical gastric cancer tissues. Altogether, these results highlight the importance of gelsolin in suppression of gastric cancer metastasis through inhibition of PKR-p38 signaling pathway.
Collapse
Affiliation(s)
- Xiangliang Yuan
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Weiwei Wang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Junhua Li
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Peiming Zheng
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ping Dong
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lei Chen
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guohua Xie
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dakang Xu
- MIMR-PHI Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| | - Yingbin Liu
- Department of Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
18
|
Targeting Heat Shock Proteins in Cancer: A Promising Therapeutic Approach. Int J Mol Sci 2017; 18:ijms18091978. [PMID: 28914774 PMCID: PMC5618627 DOI: 10.3390/ijms18091978] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022] Open
Abstract
Heat shock proteins (HSPs) are a large family of chaperones that are involved in protein folding and maturation of a variety of "client" proteins protecting them from degradation, oxidative stress, hypoxia, and thermal stress. Hence, they are significant regulators of cellular proliferation, differentiation and strongly implicated in the molecular orchestration of cancer development and progression as many of their clients are well established oncoproteins in multiple tumor types. Interestingly, tumor cells are more HSP chaperonage-dependent than normal cells for proliferation and survival because the oncoproteins in cancer cells are often misfolded and require augmented chaperonage activity for correction. This led to the development of several inhibitors of HSP90 and other HSPs that have shown promise both preclinically and clinically in the treatment of cancer. In this article, we comprehensively review the roles of some of the important HSPs in cancer, and how targeting them could be efficacious, especially when traditional cancer therapies fail.
Collapse
|
19
|
Wu J, Liu T, Rios Z, Mei Q, Lin X, Cao S. Heat Shock Proteins and Cancer. Trends Pharmacol Sci 2016; 38:226-256. [PMID: 28012700 DOI: 10.1016/j.tips.2016.11.009] [Citation(s) in RCA: 477] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/23/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022]
Abstract
Heat shock proteins (HSPs) constitute a large family of proteins involved in protein folding and maturation whose expression is induced by heat shock or other stressors. The major groups are classified based on their molecular weights and include HSP27, HSP40, HSP60, HSP70, HSP90, and large HSPs. HSPs play a significant role in cellular proliferation, differentiation, and carcinogenesis. In this article we comprehensively review the roles of major HSPs in cancer biology and pharmacology. HSPs are thought to play significant roles in the molecular mechanisms leading to cancer development and metastasis. HSPs may also have potential clinical uses as biomarkers for cancer diagnosis, for assessing disease progression, or as therapeutic targets for cancer therapy.
Collapse
Affiliation(s)
- Jianming Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tuoen Liu
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV 24901, USA.
| | - Zechary Rios
- University of Illinois College of Medicine at Chicago, Chicago, IL 60612, USA
| | - Qibing Mei
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
20
|
Stempien-Otero A, Kim DH, Davis J. Molecular networks underlying myofibroblast fate and fibrosis. J Mol Cell Cardiol 2016; 97:153-61. [PMID: 27167848 PMCID: PMC5482716 DOI: 10.1016/j.yjmcc.2016.05.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/02/2016] [Accepted: 05/05/2016] [Indexed: 01/06/2023]
Abstract
Fibrotic remodeling is a hallmark of most forms of cardiovascular disease and a strong prognostic indicator of the advancement towards heart failure. Myofibroblasts, which are a heterogeneous cell-type specialized for extracellular matrix (ECM) secretion and tissue contraction, are the primary effectors of the heart's fibrotic response. This review is focused on defining myofibroblast physiology, its progenitor cell populations, and the core signaling network that orchestrates myofibroblast differentiation as a way of understanding the basic determinants of fibrotic disease in the heart and other tissues.
Collapse
Affiliation(s)
- April Stempien-Otero
- Division of Cardiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jennifer Davis
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
21
|
Zheng S, Yang C, Liu T, Liu Q, Dai F, Sheyhidin I, Lu X. Clinicopathological significance of p38β, p38γ, and p38δ and its biological roles in esophageal squamous cell carcinoma. Tumour Biol 2016; 37:7255-7266. [PMID: 26666822 DOI: 10.1007/s13277-015-4610-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/07/2015] [Indexed: 01/09/2023] Open
Abstract
P38β, p38γ, and p38δ have been sporadically and scarcely reported to be involved in the carcinogenesis of cancers, compared with p38α isoform. However, little has been known regarding their clinicopathological significance and biological roles in esophageal squamous cell carcinoma (ESCC). Expression status of p38β, p38γ, and p38δ was assayed using immunohistochemistry with ESCC tissue microarray; ensuing clinicopathological significance was statistically analyzed. To define its biological roles on proliferation, migration and invasion of ESCC cell line Eca109 in vitro, MTT, wound healing, and Transwell assays were employed, respectively. As confirmation, athymic nude mice were taken to verify the effect over proliferation in vivo. It was found that both p38β and p38δ expression, other than p38γ, were significantly higher in ESCC tissues compared with paired normal controls. In terms of prognosis, only p38β expression was observed to be significantly associated with overall prognosis. Clinicopathologically, there was significant association between p38γ expression and clinical stage, lymph nodes metastases, and tumor volume. No significant association was found for p38β and p38δ between its expression and other clinicopathological parameters other than significant difference of expression between ESCC versus normal control. In Eca109, it was observed that p38β, p38γ, and p38δ can promote the cell growth and motility. As verification, over-expression of p38δ can promote, whereas knockdown of p38γ can prevent, the tumorigenesis in nude mice model xenografted with Eca109 cells whose basal level of p38δ was stably over-expressed and p38γ was stably knocked down. Together, our results demonstrate that p38β, p38γ, and p38δ played oncogenic roles in ESCC.
Collapse
Affiliation(s)
- Shutao Zheng
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Chenchen Yang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Tao Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Qing Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Fang Dai
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Ilyar Sheyhidin
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China
| | - Xiaomei Lu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China.
- State Key Lab Incubation Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, People's Republic of China.
- Clinical Medical Research Institute, State Key Lab Breeding Base of Xinjiang Major Diseases Research, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, Xinjiang Uygur Autonomous Region, People's Republic of China.
| |
Collapse
|
22
|
Sooraj D, Xu D, Cain JE, Gold DP, Williams BRG. Activating Transcription Factor 3 Expression as a Marker of Response to the Histone Deacetylase Inhibitor Pracinostat. Mol Cancer Ther 2016; 15:1726-39. [PMID: 27196751 DOI: 10.1158/1535-7163.mct-15-0890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 03/30/2016] [Indexed: 11/16/2022]
Abstract
Improved treatment strategies are required for bladder cancer due to frequent recurrence of low-grade tumors and poor survival rate from high-grade tumors with current therapies. Histone deacetylase inhibitors (HDACi), approved as single agents for specific lymphomas, have shown promising preclinical results in solid tumors but could benefit from identification of biomarkers for response. Loss of activating transcription factor 3 (ATF3) expression is a feature of bladder tumor progression and correlates with poor survival. We investigated the utility of measuring ATF3 expression as a marker of response to the HDACi pracinostat in bladder cancer models. Pracinostat treatment of bladder cancer cell lines reactivated the expression of ATF3, correlating with significant alteration in proliferative, migratory, and anchorage-dependent growth capacities. Pracinostat also induced growth arrest at the G0-G1 cell-cycle phase, coincident with the activation of tumor suppressor genes. In mouse xenograft bladder cancer models, pracinostat treatment significantly reduced tumor volumes compared with controls, accompanied by reexpression of ATF3 in nonproliferating cells from early to late stage of therapy and in parallel induced antiangiogenesis and apoptosis. Importantly, cells in which ATF3 expression was depleted were less sensitive to pracinostat treatment in vitro, exhibiting significantly higher proliferative and migratory properties. In vivo, control xenograft tumors were significantly more responsive to treatment than ATF3 knockdown xenografts. Thus, reactivation of ATF3 is an important factor in determining sensitivity to pracinostat treatment, both in vitro and in vivo, and could serve as a potential biomarker of response and provide a rationale for therapeutic utility in HDACi-mediated treatments for bladder cancer. Mol Cancer Ther; 15(7); 1726-39. ©2016 AACR.
Collapse
Affiliation(s)
- Dhanya Sooraj
- Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Dakang Xu
- Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Jason E Cain
- Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | | | - Bryan R G Williams
- Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
23
|
Transcriptional Activation of Inflammatory Genes: Mechanistic Insight into Selectivity and Diversity. Biomolecules 2015; 5:3087-111. [PMID: 26569329 PMCID: PMC4693271 DOI: 10.3390/biom5043087] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/11/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022] Open
Abstract
Acute inflammation, an integral part of host defence and immunity, is a highly conserved cellular response to pathogens and other harmful stimuli. An inflammatory stimulation triggers transcriptional activation of selective pro-inflammatory genes that carry out specific functions such as anti-microbial activity or tissue healing. Based on the nature of inflammatory stimuli, an extensive exploitation of selective transcriptional activations of pro-inflammatory genes is performed by the host to ensure a defined inflammatory response. Inflammatory signal transductions are initiated by the recognition of inflammatory stimuli by transmembrane receptors, followed by the transmission of the signals to the nucleus for differential gene activations. The differential transcriptional activation of pro-inflammatory genes is precisely controlled by the selective binding of transcription factors to the promoters of these genes. Among a number of transcription factors identified to date, NF-κB still remains the most prominent and studied factor for its diverse range of selective transcriptional activities. Differential transcriptional activities of NF-κB are dictated by post-translational modifications, specificities in dimer formation, and variability in activation kinetics. Apart from the differential functions of transcription factors, the transcriptional activation of selective pro-inflammatory genes is also governed by chromatin structures, epigenetic markers, and other regulators as the field is continuously expanding.
Collapse
|
24
|
Yokota T, Wang Y. p38 MAP kinases in the heart. Gene 2015; 575:369-376. [PMID: 26390817 DOI: 10.1016/j.gene.2015.09.030] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 07/31/2015] [Accepted: 09/15/2015] [Indexed: 12/28/2022]
Abstract
p38 kinases are members of the mitogen-activated protein kinases (MAPK) with established contribution to a wide range of signaling pathways and different biological processes. The prototypic p38 MAPK, p38α was originally identified as an essential signaling kinase for inflammatory cytokine production Extensive studies have now revealed that p38s have critical roles in many different tissues far beyond immune regulation and inflammatory responses. In this review, we will focus on the structure and molecular biology of p38s, and their specific roles in heart, especially regarding myocyte proliferation, apoptosis, and hypertrophic responses.
Collapse
Affiliation(s)
- Tomohiro Yokota
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Physiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Physiology and Medicine, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Mazalouskas M, Jessen T, Varney S, Sutcliffe JS, Veenstra-VanderWeele J, Cook EH, Carneiro AMD. Integrin β3 Haploinsufficiency Modulates Serotonin Transport and Antidepressant-Sensitive Behavior in Mice. Neuropsychopharmacology 2015; 40:2015-24. [PMID: 25684064 PMCID: PMC4839525 DOI: 10.1038/npp.2015.51] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/06/2015] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
Converging lines of evidence have identified genetic interactions between the serotonin transporter (SERT) gene and ITGB3, which encodes the β3 subunit that forms the αIIbβ3 and αvβ3 integrin receptor complexes. Here we examine the consequences of haploinsufficiency in the mouse integrin β3 subunit gene (Itgb3) on SERT function and selective 5-hydroxytryptamine (5-HT) reuptake inhibitor (SSRI) effectiveness in vivo. Biochemical fractionation studies and immunofluorescent staining of murine brain slices reveal that αvβ3 receptors and SERTs are enriched in presynaptic membranes from several brain regions and that αvβ3 colocalizes with a subpopulation of SERT-containing synapses in raphe nuclei. Notably, we establish that loss of a single allele of Itgb3 in murine neurons is sufficient to decrease 5-HT uptake by SERT in midbrain synaptosomes. Pharmacological assays to elucidate the αvβ3-mediated mechanism of reduced SERT function indicate that decreased integrin β3 subunit expression scales down the population size of active SERT molecules and, as a consequence, lowers the effective dose of SSRIs. These data are consistent with the existence of a subpopulation of SERTs that are tightly modulated by integrin αvβ3 and significantly contribute to global SERT function at 5-HT synapses in the midbrain. Importantly, our screen of a normal human population for single nucleotide polymorphisms in human ITGB3 identified a variant associated with reductions in integrin β3 expression levels that parallel our mouse findings. Thus, polymorphisms in human ITGB3 may contribute to the differential responsiveness of select patients to SSRIs.
Collapse
Affiliation(s)
- Matthew Mazalouskas
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tammy Jessen
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Seth Varney
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - James S Sutcliffe
- Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN, USA,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Edwin H Cook
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University School of Medicine, 461 Preston Research Building, 23rd Avenue South at Pierce, Nashville, TN 37232, USA, Tel: +1 615 875 5635, Fax: 615-343-1084, E-mail:
| |
Collapse
|
26
|
Bhat A, Heinzel A, Mayer B, Perco P, Mühlberger I, Husi H, Merseburger AS, Zoidakis J, Vlahou A, Schanstra JP, Mischak H, Jankowski V. Protein interactome of muscle invasive bladder cancer. PLoS One 2015; 10:e0116404. [PMID: 25569276 PMCID: PMC4287622 DOI: 10.1371/journal.pone.0116404] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 12/09/2014] [Indexed: 12/31/2022] Open
Abstract
Muscle invasive bladder carcinoma is a complex, multifactorial disease caused by disruptions and alterations of several molecular pathways that result in heterogeneous phenotypes and variable disease outcome. Combining this disparate knowledge may offer insights for deciphering relevant molecular processes regarding targeted therapeutic approaches guided by molecular signatures allowing improved phenotype profiling. The aim of the study is to characterize muscle invasive bladder carcinoma on a molecular level by incorporating scientific literature screening and signatures from omics profiling. Public domain omics signatures together with molecular features associated with muscle invasive bladder cancer were derived from literature mining to provide 286 unique protein-coding genes. These were integrated in a protein-interaction network to obtain a molecular functional map of the phenotype. This feature map educated on three novel disease-associated pathways with plausible involvement in bladder cancer, namely Regulation of actin cytoskeleton, Neurotrophin signalling pathway and Endocytosis. Systematic integration approaches allow to study the molecular context of individual features reported as associated with a clinical phenotype and could potentially help to improve the molecular mechanistic description of the disorder.
Collapse
Affiliation(s)
- Akshay Bhat
- Charité-Universitätsmedizin Berlin, Med. Klinik IV, Berlin, Germany
- Mosaiques diagnostics GmbH, Hannover, Germany
| | | | - Bernd Mayer
- emergentec biodevelopment GmbH, Vienna, Austria
| | - Paul Perco
- emergentec biodevelopment GmbH, Vienna, Austria
| | | | - Holger Husi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Axel S. Merseburger
- Department of Urology and Urological Oncology, Hannover Medical School, Hannover, Germany
| | - Jerome Zoidakis
- Biomedical Research Foundation Academy of Athens, Biotechnology Division, Athens, Greece
| | - Antonia Vlahou
- Biomedical Research Foundation Academy of Athens, Biotechnology Division, Athens, Greece
| | - Joost P. Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Diseases, Toulouse, France
- Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Harald Mischak
- Mosaiques diagnostics GmbH, Hannover, Germany
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), Aachen, Germany
- * E-mail:
| |
Collapse
|
27
|
Chow SC, Gowing SD, Cools-Lartigue JJ, Chen CB, Berube J, Yoon HW, Chan CHF, Rousseau MC, Bourdeau F, Giannias B, Roussel L, Qureshi ST, Rousseau S, Ferri LE. Gram negative bacteria increase non-small cell lung cancer metastasis via Toll-like receptor 4 activation and mitogen-activated protein kinase phosphorylation. Int J Cancer 2014; 136:1341-50. [PMID: 25082668 DOI: 10.1002/ijc.29111] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 06/26/2014] [Indexed: 12/13/2022]
Abstract
Surgery is required for the curative treatment of lung cancer but is associated with high rates of postoperative pneumonias predominantly caused by gram negative bacteria. Recent evidence suggests that these severe infectious complications may decrease long term survival after hospital discharge via cancer recurrence, but the mechanism is unclear. Lung cancer cells have recently been demonstrated to express Toll-like receptors (TLR) that mediate pathogen recognition. We hypothesized that incubation of non-small cell lung cancer (NSCLC) cells with heat-inactivated Escherichia coli can augment cancer cell adhesion, migration and metastasis via TLR4 signaling. Incubation of murine and human NSCLC cells with E. coli increased in vitro cell adhesion to collagen I, collagen IV and fibronectin, and enhanced in vitro migration. Using hepatic intravital microscopy, we demonstrated that NSCLC cells have increased in vivo adhesion to hepatic sinusoids after coincubation with gram negative bacteria. These enhanced cell adhesion and migration phenotypes following incubation with E. coli were attenuated at three levels: inhibition of TLR4 (Eritoran), p38 MAPK (BIRB0796) and ERK1/2 phosphorylation (PD184352). Incubation of murine NSCLC cells in vitro with E. coli prior to intrasplenic injection significantly augmented formation of in vivo hepatic metastases 2 weeks later. This increase was abrogated by NSCLC TLR4 blockade using Eritoran. TLR4 represents a potential therapeutic target to help prevent severe postoperative infection driven cancer metastasis.
Collapse
Affiliation(s)
- Simon C Chow
- Department of Surgery, L.D. MacLean Surgical Research Laboratories, McGill University Health Centre, McGill University, Montreal, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gupta S, Hau AM, Beach JR, Harwalker J, Mantuano E, Gonias SL, Egelhoff TT, Hansel DE. Mammalian target of rapamycin complex 2 (mTORC2) is a critical determinant of bladder cancer invasion. PLoS One 2013; 8:e81081. [PMID: 24312263 PMCID: PMC3842329 DOI: 10.1371/journal.pone.0081081] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/08/2013] [Indexed: 01/12/2023] Open
Abstract
Bladder cancer is the fourth most common cause of cancer in males in the United States. Invasive behavior is a major determinant of prognosis. In this study, we identified mammalian target of rapamycin complex 2 (mTORC2) as a central regulator of bladder cancer cell migration and invasion. mTORC2 activity was assessed by the extent of phosphorylation of Ser473 in AKT and determined to be approximately 5-fold higher in specimens of invasive human bladder cancer as opposed to non-invasive human bladder cancer. The immortalized malignant bladder cell lines, UMUC-3, J82 and T24 demonstrated higher baseline mTORC2 activity relative to the benign bladder papilloma-derived cell line RT4 and the normal urothelial cell line HU1. The malignant bladder cancer cells also demonstrated increased migration in transwell and denudation assays, increased invasion of matrigel, and increased capacity to invade human bladder specimens. Gene silencing of rictor, a critical component of mTORC2, substantially inhibited bladder cancer cell migration and invasion. This was accompanied by a significant decrease in Rac1 activation and paxillin phosphorylation. These studies identify mTORC2 as a major target for neutralizing bladder cancer invasion.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Andrew M. Hau
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Jordan R. Beach
- Department of Cell Biology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Jyoti Harwalker
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Elisabetta Mantuano
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Steven L. Gonias
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Thomas T. Egelhoff
- Department of Cell Biology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Donna E. Hansel
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
- *
| |
Collapse
|