1
|
Zhang Z, Xue B, Chen Y, Shao Y, Wang D. A systematic review and meta-analysis combined with bioinformatic analysis on the predictive value of E-cadherin in patients with renal cell carcinoma. Expert Rev Mol Diagn 2024; 24:859-871. [PMID: 39187988 DOI: 10.1080/14737159.2024.2392641] [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: 05/13/2023] [Accepted: 03/08/2024] [Indexed: 08/28/2024]
Abstract
OBJECTIVES Renal cell carcinoma (RCC) is the most common cancer of the kidney. This study aims to evaluate the potential predictive value of E-cadherin, a marker of the epithelial mesenchymal transit (EMT) process that has been associated with tumor metastasis. METHODS We searched PubMed, Embase, and Cochrane Library to identify prospective studies. Hazard ratios (HRs), odds ratios (ORs), and 95% confidence intervals (CIs) were summarized to validate the relationship between E-cadherin and survival and clinical characteristics. The quality of the included studies was assessed using the NOS table. Then, we analyzed genetic data and clinical characteristics from The Cancer Genome Atlas Program (TCGA) database using R language with the dplyr package for validation. RESULTS Including 21 articles. The analysis revealed a strong link between high E-cadherin expression and favorable prognosis (for OS, HR = 0.35, 95% CI: 0.19-0.62; for PFS, HR = 0.19, 95% CI: 0.03-0.53; for DSS, HR = 0.25, 95% CI: 0.08-0.76; for RFS, HR = 0.71, 95% CI: 0.44-1.16; for DFS, HR = 0.28, 95% CI: 0.13-0.61; for T stage, OR = 0.21, 95% CI: 0.11-0.41; for N stage, OR = 0.07, 95%CI: 0.02-0.25; for M stage, OR = 0.12, 95% CI: 0.02-0.60; for clinical stage, OR = 0.29, 95% CI: 0.18-0.47; for nuclear grade, OR = 0.23, 95% CI: 0.13-0.41; for tumor size, OR = 0.49, 95% CI: 0.26-0.92). The findings were supported by bioinformatic analysis which used TCGA RCC patient's cohort (P < 0.01). CONCLUSION Based on the current data, E-cadherin may predict a better prognosis in RCC patients.
Collapse
Affiliation(s)
- Zikuan Zhang
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bo Xue
- Shanxi Medical University, Taiyuan, Shanxi, China
| | | | - Yuan Shao
- Tianjin Medical University, Tianjin, China
| | - Dongwen Wang
- Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Yan S, Wang Z, Lan D, Niu J, Jian X, He F, Tang W, Hu C, Liu W. Circ_PABPC1 promotes the malignancy of gastric cancer through interacting with ILK to activate NF-κB pathway. Exp Cell Res 2024; 438:114058. [PMID: 38688434 DOI: 10.1016/j.yexcr.2024.114058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a common cancer type with both high incidence and mortality. Recent studies have revealed an important role of circRNA in the development of GC. However, more experiments are needed to reveal the precise molecular mechanisms of circRNA in GC development. METHODS Bioinformatics analysis was conducted to predict the potential role of circ_PABPC1 in GC and the target proteins of circ_PABPC1. Quantitative RT-PCR, Western blot and immunohistochemistry assays were conducted to detect the levels of circ_PABPC1, NF-κB p65, NF-κB p65 (Ser536) and ILK. MTT, Edu staining, cell scratch-wound and trans-well assays were carried out to detect cell proliferation, migration and invasion. The interaction between ILK and circ_PABPC1 was confirmed by RNA immunoprecipitation (RIP), RNA pull-down and fluorescence in situ hybridization assays. Genetically modified GC cells were injected into mice to evaluate the tumor growth performance. RESULTS This study found that the high expression of circ_PABPC1 was associated with a poor prognosis of GC. The up-regulation of circ_PABPC1 promoted the proliferation, migration and invasion of GC cells. Circ_PABPC1 bound to ILK protein, thereby preventing the degradation of ILK. ILK mediated the effect of circ_PABPC1 on GC cells through activating NF-κB. CONCLUSION circ_PABPC1 promotes the malignancy of GC cells through binding to ILK to activate NF-κB signaling pathway.
Collapse
Affiliation(s)
- Siqi Yan
- Departments of Oncology, The Second Xiangya Hospital of Central-South University, Changsha, Hunan, 410011, China; Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China
| | - Zhu Wang
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China
| | - Dongqiang Lan
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China
| | - Junjie Niu
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China
| | - Xiaolan Jian
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China
| | - Fengjiao He
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China; Departments of Oncology, Xiangya Hospital of Central-South University, Changsha, Hunan, 410008, China
| | - Weizhi Tang
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China
| | - Chunhong Hu
- Departments of Oncology, The Second Xiangya Hospital of Central-South University, Changsha, Hunan, 410011, China.
| | - Wei Liu
- Departments of Radiotherapy, Hunan Provincial Hospital of Integrated Chinese and Western Medicine, The Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, 410006, China.
| |
Collapse
|
3
|
Pontrelli P, Gigante M, Spadaccino F, Netti GS, Saldarelli M, Balducci L, Gigante M, Battaglia M, Storkus WJ, Castellano G, Stallone G, Gesualdo L, Ranieri E. CD40 Cross-Linking Induces Migration of Renal Tumor Cell through Nuclear Factor of Activated T Cells (NFAT) Activation. Int J Mol Sci 2021; 22:ijms22168871. [PMID: 34445576 PMCID: PMC8396205 DOI: 10.3390/ijms22168871] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
CD40 crosslinking plays an important role in regulating cell migration, adhesion and proliferation in renal cell carcinoma (RCC). CD40/CD40L interaction on RCC cells activates different intracellular pathways but the molecular mechanisms leading to cell scattering are not yet clearly defined. Aim of our study was to investigate the main intracellular pathways activated by CD40 ligation and their specific involvement in RCC cell migration. CD40 ligation increased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun NH (2)-terminal kinase (JNK) and p38 MAPK. Furthermore, CD40 crosslinking activated different transcriptional factors on RCC cell lines: AP-1, NFkB and some members of the Nuclear Factor of Activated T cells (NFAT) family. Interestingly, the specific inhibition of NFAT factors by cyclosporine A, completely blocked RCC cell motility induced by CD40 ligation. In tumor tissue, we observed a higher expression of NFAT factors and in particular an increased activation and nuclear migration of NFATc4 on RCC tumor tissues belonging to patients that developed metastases when compared to those who did not. Moreover, CD40-CD40L interaction induced a cytoskeleton reorganization and increased the expression of integrin β1 on RCC cell lines, and this effect was reversed by cyclosporine A and NFAT inhibition. These data suggest that CD40 ligation induces the activation of different intracellular signaling pathways, in particular the NFATs factors, that could represent a potential therapeutic target in the setting of patients with metastatic RCC.
Collapse
Affiliation(s)
- Paola Pontrelli
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Margherita Gigante
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Federica Spadaccino
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Giuseppe Stefano Netti
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Marilisa Saldarelli
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Luigi Balducci
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Maddalena Gigante
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Michele Battaglia
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Walter J. Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | - Giuseppe Castellano
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Giovanni Stallone
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Elena Ranieri
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
- Correspondence: ; Tel.: +39-0881-732611; Fax: +39-0881-732627
| |
Collapse
|
4
|
Masi I, Caprara V, Spadaro F, Chellini L, Sestito R, Zancla A, Rainer A, Bagnato A, Rosanò L. Endothelin-1 drives invadopodia and interaction with mesothelial cells through ILK. Cell Rep 2021; 34:108800. [PMID: 33657382 DOI: 10.1016/j.celrep.2021.108800] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 01/02/2021] [Accepted: 02/05/2021] [Indexed: 01/09/2023] Open
Abstract
Cancer cells use actin-based membrane protrusions, invadopodia, to degrade stroma and invade. In serous ovarian cancer (SOC), the endothelin A receptor (ETAR) drives invadopodia by a not fully explored coordinated function of β-arrestin1 (β-arr1). Here, we report that β-arr1 links the integrin-linked kinase (ILK)/βPIX complex to activate Rac3 GTPase, acting as a central node in the adhesion-based extracellular matrix (ECM) sensing and degradation. Downstream, Rac3 phosphorylates PAK1 and cofilin and promotes invadopodium-dependent ECM proteolysis and invasion. Furthermore, ETAR/ILK/Rac3 signaling supports the communication between cancer and mesothelial cells, favoring SOC cell adhesion and transmigration. In vivo, ambrisentan, an ETAR antagonist, inhibits the adhesion and spreading of tumor cells to intraperitoneal organs, and invadopodium marker expression. As prognostic factors, high EDNRA/ILK expression correlates with poor SOC clinical outcome. These findings provide a framework for the ET-1R/β-arr1 pathway as an integrator of ILK/Rac3-dependent adhesive and proteolytic signaling to invadopodia, favoring cancer/stroma interactions and metastatic behavior.
Collapse
Affiliation(s)
- Ilenia Masi
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome 00128, Italy
| | - Valentina Caprara
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome 00128, Italy
| | - Francesca Spadaro
- Confocal Microscopy Unit, Core Facilities, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Lidia Chellini
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome 00128, Italy
| | - Rosanna Sestito
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome 00128, Italy
| | - Andrea Zancla
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, Rome 00128, Italy; Department of Engineering, Università degli Studi Roma Tre, via Vito Volterra 62, Rome 00146, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, Rome 00128, Italy; Institute of Nanotechnology (NANOTEC), National Research Council (CNR), c/o Campus Ecotekne, via Monteroni, Lecce 73100, Italy
| | - Anna Bagnato
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome 00128, Italy
| | - Laura Rosanò
- Unit of Preclinical Models and New Therapeutic Agents, IRCCS - Regina Elena National Cancer Institute, Rome 00128, Italy; Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome 00185, Italy.
| |
Collapse
|
5
|
Huang M, Zhu S, Huang H, He J, Tsuji K, Jin WW, Xie D, Ham O, Capen DE, Lu W, Păunescu TG, Yang B, Lu HAJ. Integrin-Linked Kinase Deficiency in Collecting Duct Principal Cell Promotes Necroptosis of Principal Cell and Contributes to Kidney Inflammation and Fibrosis. J Am Soc Nephrol 2019; 30:2073-2090. [PMID: 31653783 PMCID: PMC6830785 DOI: 10.1681/asn.2018111162] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 07/15/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Necroptosis is a newly discovered cell death pathway that plays a critical role in AKI. The involvement of integrin-linked kinase (ILK) in necroptosis has not been studied. METHODS We performed experiments in mice with an Ilk deletion in collecting duct (CD) principal cells (PCs), and cultured tubular epithelial cells treated with an ILK inhibitor or ILK siRNA knockdown. RESULTS Ilk deletion in CD PCs resulted in acute tubular injury and early mortality in mice. Progressive interstitial fibrosis and inflammation associated with the activation of the canonical TGF-β signaling cascade were detected in the kidneys of the mice lacking ILK in the CD PCs. In contrast to the minimal apoptosis detected in the animals' injured CDs, widespread necroptosis was present in ILK-deficient PCs, characterized by cell swelling, deformed mitochondria, and rupture of plasma membrane. In addition, ILK deficiency resulted in increased expression and activation of necroptotic proteins MLKL and RIPK3, and membrane translocation of MLKL in CD PCs. ILK inhibition and siRNA knockdown reduced cell survival in cultured tubular cells, concomitant with increased membrane accumulation of MLKL and/or phospho-MLKL. Administration of a necroptosis inhibitor, necrostatin-1, blocked cell death in vitro and significantly attenuated inflammation, interstitial fibrosis, and renal failure in ILK-deficient mice. CONCLUSIONS The study demonstrates the critical involvement of ILK in necroptosis through modulation of the RIPK3 and MLKL pathway and highlights the contribution of CD PC injury to the development of inflammation and interstitial fibrosis of the kidney.
Collapse
Affiliation(s)
- Ming Huang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuai Zhu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Huihui Huang
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jinzhao He
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kenji Tsuji
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - William W Jin
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dongping Xie
- Department of Physiology, Tongji University School of Medicine, Shanghai, China; and
| | - Onju Ham
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Diane E Capen
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Weining Lu
- Renal Section, Departments of Medicine, and Pathology & Laboratory Medicine, Boston University Medical Center, Boston, Massachusetts
| | - Teodor G Păunescu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China;
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
6
|
Clinical significance of integrin-linked kinase in laryngeal squamous cell carcinoma. Auris Nasus Larynx 2017; 44:458-463. [DOI: 10.1016/j.anl.2016.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 07/05/2016] [Accepted: 09/08/2016] [Indexed: 11/21/2022]
|
7
|
Khan MI, Dębski KJ, Dabrowski M, Czarnecka AM, Szczylik C. Gene set enrichment analysis and ingenuity pathway analysis of metastatic clear cell renal cell carcinoma cell line. Am J Physiol Renal Physiol 2016; 311:F424-36. [PMID: 27279483 DOI: 10.1152/ajprenal.00138.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/06/2016] [Indexed: 11/22/2022] Open
Abstract
In recent years, genome-wide RNA expression analysis has become a routine tool that offers a great opportunity to study and understand the key role of genes that contribute to carcinogenesis. Various microarray platforms and statistical approaches can be used to identify genes that might serve as prognostic biomarkers and be developed as antitumor therapies in the future. Metastatic renal cell carcinoma (mRCC) is a serious, life-threatening disease, and there are few treatment options for patients. In this study, we performed one-color microarray gene expression (4×44K) analysis of the mRCC cell line Caki-1 and the healthy kidney cell line ASE-5063. A total of 1,921 genes were differentially expressed in the Caki-1 cell line (1,023 upregulated and 898 downregulated). Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) approaches were used to analyze the differential-expression data. The objective of this research was to identify complex biological changes that occur during metastatic development using Caki-1 as a model mRCC cell line. Our data suggest that there are multiple deregulated pathways associated with metastatic clear cell renal cell carcinoma (mccRCC), including integrin-linked kinase (ILK) signaling, leukocyte extravasation signaling, IGF-I signaling, CXCR4 signaling, and phosphoinositol 3-kinase/AKT/mammalian target of rapamycin signaling. The IPA upstream analysis predicted top transcriptional regulators that are either activated or inhibited, such as estrogen receptors, TP53, KDM5B, SPDEF, and CDKN1A. The GSEA approach was used to further confirm enriched pathway data following IPA.
Collapse
Affiliation(s)
- Mohammed I Khan
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland; and
| | - Konrad J Dębski
- Bioinformatics Laboratory, Center of Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Michał Dabrowski
- Bioinformatics Laboratory, Center of Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Anna M Czarnecka
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland; and
| | - Cezary Szczylik
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland; and
| |
Collapse
|
8
|
Activated Integrin-Linked Kinase Negatively Regulates Muscle Cell Enhancement Factor 2C in C2C12 Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2015:748470. [PMID: 26788505 PMCID: PMC4695646 DOI: 10.1155/2015/748470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 11/25/2015] [Accepted: 11/29/2015] [Indexed: 01/28/2023]
Abstract
Our previous study reported that muscle cell enhancement factor 2C (MEF2C) was fully activated after inhibition of the phosphorylation activity of integrin-linked kinase (ILK) in the skeletal muscle cells of goats. It enhanced the binding of promoter or enhancer of transcription factor related to proliferation of muscle cells and then regulated the expression of these genes. In the present investigation, we explored whether ILK activation depended on PI3K to regulate the phosphorylation and transcriptional activity of MEF2C during C2C12 cell proliferation. We inhibited PI3K activity in C2C12 with LY294002 and then found that ILK phosphorylation levels and MEF2C phosphorylation were decreased and that MCK mRNA expression was suppressed significantly. After inhibiting ILK phosphorylation activity with Cpd22 and ILK-shRNA, we found MEF2C phosphorylation activity and MCK mRNA expression were increased extremely significantly. In the presence of Cpd22, PI3K activity inhibition increased MEF2C phosphorylation and MCK mRNA expression indistinctively. We conclude that ILK negatively and independently of PI3K regulated MEF2C phosphorylation activity and MCK mRNA expression in C2C12 cells. The results provide new ideas for the study of classical signaling pathway of PI3K-ILK-related proteins and transcription factors.
Collapse
|
9
|
Wu H, Ren Y, Pan W, Dong Z, Cang M, Liu D. The mammalian target of rapamycin signaling pathway regulates myocyte enhancer factor-2C phosphorylation levels through integrin-linked kinase in goat skeletal muscle satellite cells. Cell Biol Int 2015; 39:1264-73. [PMID: 26041412 DOI: 10.1002/cbin.10499] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/30/2015] [Indexed: 12/21/2022]
Abstract
Mammalian target of rapamycin (mTOR) signaling pathway plays a key role in muscle development and is involved in multiple intracellular signaling pathways. Myocyte enhancer factor-2 (MEF2) regulates muscle cell proliferation and differentiation. However, how the mTOR signaling pathway regulates MEF2 activity remains unclear. We isolated goat skeletal muscle satellite cells (gSSCs) as model cells to explore mTOR signaling pathway regulation of MEF2C. We inhibited mTOR activity in gSSCs with PP242 and found that MEF2C phosphorylation was decreased and that muscle creatine kinase (MCK) expression was suppressed. Subsequently, we detected integrin-linked kinase (ILK) using MEF2C coimmunoprecipitation; ILK and MEF2C were colocalized in the gSSCs. We found that inhibiting mTOR activity increased ILK phosphorylation levels and that inhibiting ILK activity with Cpd 22 and knocking down ILK with small interfering RNA increased MEF2C phosphorylation and MCK expression. In the presence of Cpd 22, mTOR activity inhibition did not affect MEF2C phosphorylation. Moreover, ILK dephosphorylated MEF2C in vitro. These results suggest that the mTOR signaling pathway regulates MEF2C positively and regulates ILK negatively and that ILK regulates MEF2C negatively. It appears that the mTOR signaling pathway regulates MEF2C through ILK, further regulating the expression of muscle-related genes in gSSCs.
Collapse
Affiliation(s)
- Haiqing Wu
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology Ministry of Education, Inner Mongolia University, China
| | - Yu Ren
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology Ministry of Education, Inner Mongolia University, China
| | - Wei Pan
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology Ministry of Education, Inner Mongolia University, China
| | - Zhenguo Dong
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology Ministry of Education, Inner Mongolia University, China
| | - Ming Cang
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology Ministry of Education, Inner Mongolia University, China
| | - Dongjun Liu
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology Ministry of Education, Inner Mongolia University, China
| |
Collapse
|
10
|
Brunner SM, Weber F, Werner JM, Agha A, Farkas SA, Schlitt HJ, Hornung M. Neuroendocrine tumors of the pancreas: a retrospective single-center analysis using the ENETS TNM-classification and immunohistochemical markers for risk stratification. BMC Surg 2015; 15:49. [PMID: 25928025 PMCID: PMC4451725 DOI: 10.1186/s12893-015-0033-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 04/20/2015] [Indexed: 02/06/2023] Open
Abstract
Background This study was performed to assess the 2006 introduced ENETS TNM-classification with respect to patient survival and surgical approach for patients who underwent surgery for a neuroendocrine tumor of the pancreas (PNET). Methods Between 2001 and 2010 38 patients after resection of a PNET were investigated regarding tumor localization and size. Further, patient survival with regards to the new TNM-classification, the operation methods and immunohistochemical markers was analyzed. Results The estimated mean survival time of the 38 patients was 91 ± 10 months (female 116 ± 9, male 56 ± 14 months; p = 0.008). The 5-year survival rate was 63.9%. Patient survival differed significantly depending on tumor size (pT1 107 ± 13, pT2 94 ± 16, pT3 44 ± 7 and pT4 18 ± 14 months; P = 0.006). Patients without lymph node metastasis survived significantly longer compared to patients with positive lymph node status (108 ± 9 vs. 19 ± 5 months; P < 0.001). However, survival in patients with and without distant metastasis did not differ significantly (92 ± 11 vs. 80 ± 23 months; P = 0.876). Further, the tumor grading significantly influenced patient survival (G1 111 ± 12, G2 68 ± 12 and G3 21 ± 14 months; P = 0.037). Conclusions As part of the TNM-classification especially lymph node status and also tumor size and grading were identified as important factors determining patient survival. Further, gender was demonstrated to significantly influence survival time. If an R0 resection was achieved in patients with distant metastases patient survival was comparable to patients without metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s12893-015-0033-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stefan M Brunner
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| | - Florian Weber
- Institute of Pathology, University Medical Center Regensburg, Regensburg, Germany
| | - Jens M Werner
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Ayman Agha
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Stefan A Farkas
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Matthias Hornung
- Department of Surgery, University Medical Center Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
11
|
Han KS, Li N, Raven PA, Fazli L, Ettinger S, Hong SJ, Gleave ME, So AI. Targeting Integrin-Linked Kinase Suppresses Invasion and Metastasis through Downregulation of Epithelial-to-Mesenchymal Transition in Renal Cell Carcinoma. Mol Cancer Ther 2015; 14:1024-34. [PMID: 25657336 DOI: 10.1158/1535-7163.mct-14-0771] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/28/2015] [Indexed: 11/16/2022]
Abstract
Renal cell carcinoma (RCC) is the most common malignancy in the kidney. Antiangiogenic targeted therapies inhibit the progression of RCC, but have limited impacts on invasion or metastasis of tumor cells. Integrin-linked kinase (ILK) is a serine/threonine kinase implicated in the regulation of cell growth/survival, cell-cycle progression, epithelial-mesenchymal transition (EMT), invasion/migration, and angiogenesis. However, the role of ILK in RCC has not been evaluated. We investigated the role of ILK on cancer progression and metastasis and the therapeutic potential of ILK inhibition in RCC. Our investigation reveals that ILK is expressed at a low level in normal cells and low-stage RCC cells and is highly expressed in advanced and metastatic cells. Caki-1, a metastatic RCC cell line, showed higher expression of molecular EMT markers, including Snail and Zeb1, but decreased activity of GSK3β. Knockdown of ILK using small interference (si)-ILK minimally inhibited tumor proliferation and cell-cycle progression was not significantly affected. However, ILK knockdown suppressed the formation of stress fibers and focal adhesions and impeded phenotypic EMT markers, including cell migration and invasion, in Caki-1 and UMRC-3 cells. Finally, in vivo knockdown of ILK suppressed the progression, invasion, and metastasis of primary RCC in nude mice by downregulation of EMT markers (Snail, Zeb1, vimentin, and E-cadherin). Our results show that ILK may be essential for invasion and metastasis in RCC and regulates vimentin and E-cadherin expression by regulating the EMT-related transcription factors Snail and Zeb1. These results suggest that ILK may be a potential target in RCC.
Collapse
Affiliation(s)
- Kyung Seok Han
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada. Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Na Li
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter A Raven
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Susan Ettinger
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sung Joon Hong
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Martin E Gleave
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alan I So
- Vancouver Prostate Centre and Department of Urologic Science, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
12
|
Araújo WF, Naves MA, Ravanini JN, Schor N, Teixeira VPC. Renin-angiotensin system (RAS) blockade attenuates growth and metastatic potential of renal cell carcinoma in mice. Urol Oncol 2015; 33:389.e1-7. [PMID: 25595575 DOI: 10.1016/j.urolonc.2014.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/27/2014] [Accepted: 11/27/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Renal cell carcinoma (RCC) is the most frequent type of cancer among renal neoplasms in adults and responds poorly to radiotherapy and chemotherapy. There is evidence that blockade of the renin-angiotensin system (RAS) might have antineoplastic effects. The aim of this study was to investigate the effects of RAS blockade on RCC in a murine model. METHODS AND MATERIALS Murine renal cancer cells (Renca) were injected (1 × 10(5)) into the subcapsular space of the left kidney of BALB/c mice (8 wk of age). The animals were divided into 4 groups: a control group (no treatment), angiotensin-receptor blockers group (losartan 100mg/kg/d), angiotensin-converting enzyme inhibitor group (captopril 10mg/kg/d), and angiotensin-receptor blockers +angiotensin-converting enzyme inhibitor group (losartan 100mg/kg/d +captopril 10mg/kg/d). The animals received the drugs by gavage for 21 days after inoculation, beginning 2 days before tumor induction, and were then euthanized. After killing the animals, the kidneys and lungs were removed, weighed, and processed for histopathological and immunohistochemical analyses. Angiogenesis and vascular microvessels were assessed with the antibodies anti-vascular endothelial growth factor and anti-CD34. RESULTS Angiotensin II-inoculated animals developed renal tumors. Treated animals presented smaller tumors, regardless of the therapeutic regimen, and far fewer lung metastases in both quantity and dimension compared with the controls. The expression of vascular endothelial growth factor and CD34 were significantly decreased in renal tumors of treated animals compared with the controls. CONCLUSIONS Our findings suggest that blockade of RAS decreases tumor proliferation and metastatic capacity of RCC in this experimental model.
Collapse
Affiliation(s)
- Wedson F Araújo
- Department of Pathology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil.
| | - Marcelo A Naves
- Department of Medicine, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | - Juliana N Ravanini
- Department of Pathology, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Nestor Schor
- Department of Medicine, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | - Vicente P C Teixeira
- Department of Pathology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil; Department of Medicine, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| |
Collapse
|
13
|
Kirik U, Hansson K, Krogh M, Jönsson M, Nilbert M, James P, Carneiro A. Discovery-based protein expression profiling identifies distinct subgroups and pathways in leiomyosarcomas. Mol Cancer Res 2014; 12:1729-39. [PMID: 25069693 DOI: 10.1158/1541-7786.mcr-14-0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Soft tissue sarcomas (STS) are malignant tumors of mesenchymal origin. A substantial portion of these tumors exhibits complex karyotypes and lack characterized chromosomal aberrations. Owing to such properties, both histopathologic and molecular classification of these tumors has been a significant challenge. This study examines the protein expression of a large number of human STS, including subtype heterogeneity, using two-dimensional gel proteomics. In addition, detailed proteome profiles of a subset of pleomorphic STS specimens using an in-depth mass-spectrometry approach identified subgroups within the leiomyosarcomas with distinct protein expression patterns. Pathways analysis indicates that key biologic nodes like apoptosis, cytoskeleton remodeling, and telomere regulation are differentially regulated among these subgroups. Finally, investigating the similarities between protein expression of leiomyosarcomas and undifferentiated pleomorphic sarcomas (UPS) revealed similar protein expression profiles for these tumors, in comparison with pleomorphic leiomyosarcomas. IMPLICATIONS These results suggest that UPS tumors share a similar lineage as leiomyosarcomas and are likely to originate from different stages of differentiation from mesenchymal stem cells to smooth muscle cells.
Collapse
Affiliation(s)
- Ufuk Kirik
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Karin Hansson
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | - Mef Nilbert
- Institute of Clinical Sciences, Department of Oncology, Lund University, Sweden. Clinical Research Centre, Hvidovre Hospital, Copenhagen University, Denmark
| | - Peter James
- Department of Immunotechnology, Lund University, Lund, Sweden.
| | - Ana Carneiro
- Institute of Clinical Sciences, Department of Oncology, Lund University, Sweden
| |
Collapse
|
14
|
Rhee SH, Han I, Lee MR, Cho HS, Oh JH, Kim HS. Role of integrin-linked kinase in osteosarcoma progression. J Orthop Res 2013; 31:1668-75. [PMID: 23784942 DOI: 10.1002/jor.22409] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 05/13/2013] [Indexed: 02/04/2023]
Abstract
Although integrin-linked kinase (ILK) has been suggested to play a role in the tumorigenesis of a number of human epithelial carcinomas, little is known of its role in musculoskeletal sarcoma. The authors studied ILK expression by immunohistochemistry using osteosarcoma prechemotherapy specimens from 56 patients, and investigated the prognostic implications of the findings obtained. It was found that ILK overexpression was significantly correlated with the presence of distant metastasis (p = 0.008) and that it was an independent prognostic factor for both poor overall survival and poor event-free survival (p = 0.015 and 0.010, respectively). During a transfection experiment conducted by transfecting osteosarcoma cells with ILK siRNA, VEGF concentrations were measured using an ELISA kit, and then compared with those of untransfected controls to evaluate its angiogenic effects. In addition, apoptotic percentages were measured by Annexin-V flow cytometry, and invasive properties were evaluated by measuring the numbers of non-migrating cells in a Boyden chamber. It was found that ILK downregulation significantly decreased angiogenesis, increased apoptosis, and decreased invasiveness of osteosarcoma cells. These results show that ILK is a promising prognostic factor in osteosarcoma and a novel potential therapeutic target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Seung Hwan Rhee
- Department of Orthopaedic Surgery, Seoul National University Boramae Medical Center, Seoul, Korea
| | | | | | | | | | | |
Collapse
|