1
|
Min X, Cheng H, Cao X, Chen Z, Zhang X, Li Y, Mao Q, Xue B, Fang L, Liu L, Ding Z. Heat shock protein A12A activates migration of hepatocellular carcinoma cells in a monocarboxylate transporter 4-dependent manner. Cell Stress Chaperones 2022; 27:83-95. [PMID: 35050463 PMCID: PMC8821763 DOI: 10.1007/s12192-021-01251-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022] Open
Abstract
Metastasis is responsible for most of the hepatocellular carcinoma (HCC)-associated death. However, its underlying mechanism has yet to be fully elucidated. Glycolysis-derived lactate has been shown to be a powerful regulator of cancer metastasis. Heat shock protein A12A (HSPA12A) encodes a novel member of HSP70 family. We have recently demonstrated that heat shock protein A12A (HSPA12A) inhibited renal cancer cell migration by suppressing lactate output and glycolytic activity, which were mediated by unstabilizing CD147 and promoting its degradation. By striking contrast, here we demonstrated that HSPA12A promoted migration of human HCC cells. Extracellular acidification, lactate export, and glycolytic activity in HCC cells were also promoted following HSPA12A overexpression. Further analysis revealed that HSPA12A interacted with MCT4 and increased its membrane localization, thereby promoting export of lactate generated from glycolysis; this led, ultimately, to HCC cell migration. Our results revealed the opposite effect of HSPA12A on migration of renal cancer cells and that of HCC cells. Of note, in contrast to the inhibitory effect on CD147 expression in renal cancer cells, we found that HSPA12A increased CD147 expression in HCC cells, indicating that the expression of CD147 might exist heterogeneity in different cancer cell types. Taken together, we identified HSPA12A as an activator of HCC migration, a role opposite to that of renal cancer cells. Inhibiting HSPA12A might be a potential therapeutic intervention for HCC metastasis.
Collapse
Affiliation(s)
- Xinxu Min
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China
| | - Hao Cheng
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China
- Department of Anesthesiology, The First Affiliated Hospital With Wannan Medical College, Wuhu, 241001, China
| | - Xiaofei Cao
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China
| | - Ziyang Chen
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
| | - Yunfan Li
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China
| | - Qian Mao
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China
| | - Bin Xue
- Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211166, China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, 210093, China
| | - Li Liu
- Department of Geriatrics, First Affiliated Hospital With Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 210029, China
| | - Zhengnian Ding
- Department of Anesthesiology, The First Affiliated Hospital With Nanjing Medical University, Guangzhou Rd 300, Nanjing, 210029, China.
| |
Collapse
|
2
|
Tegel BR, Huber S, Savic LJ, Lin M, Gebauer B, Pollak J, Chapiro J. Quantification of contrast-uptake as imaging biomarker for disease progression of renal cell carcinoma after tumor ablation. Acta Radiol 2020; 61:1708-1716. [PMID: 32216452 DOI: 10.1177/0284185120909964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The prognosis of patients with renal cell carcinoma (RCC) depends greatly on the presence of extra-renal metastases. PURPOSE To investigate the value of total tumor volume (TTV) and enhancing tumor volume (ETV) as three-dimensional (3D) quantitative imaging biomarkers for disease aggressiveness in patients with RCC. MATERIAL AND METHODS Retrospective, HIPAA-compliant, IRB-approved study including 37 patients with RCC treated with image-guided thermal ablation during 2007-2015. TNM stage, RENAL Nephrometry Score, largest tumor diameter, TTV, and ETV were assessed on cross-sectional imaging at baseline and correlated with outcome measurements. The primary outcome was time-to-occurrence of extra-renal metastases and the secondary outcome was progression-free survival (PFS). Correlation was assessed using a Cox regression model and differences in outcomes were shown by Kaplan-Meier plots with significance and odds ratios (OR) calculated by Log-rank test/generalized Wilcoxon and continuity-corrected Woolf logit method. RESULTS Patients with a TTV or ETV > 5 cm3 were more likely to develop distant metastases compared to patients with TTV (OR 6.69, 95% confidence interval [CI] 0.33-134.4, P=0.022) or ETV (OR 8.48, 95% CI 0.42-170.1, P=0.016) < 5 cm3. Additionally, PFS was significantly worse in patients with larger ETV (P = 0.039; median PFS 51.87 months vs. 69.97 months). In contrast, stratification by median value of the established, caliper-based measurements showed no significant correlation with outcome parameters. CONCLUSION ETV, as surrogate of lesion vascularity, is a sensitive imaging biomarker for occurrence of extra-renal metastatic disease and PFS in patients with RCC.
Collapse
Affiliation(s)
- Bruno R Tegel
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Steffen Huber
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Lynn J Savic
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - MingDe Lin
- U/S Imaging and Interventions, Philips Research North America, Cambridge, MA, USA
| | - Bernhard Gebauer
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Jeffrey Pollak
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Julius Chapiro
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| |
Collapse
|
3
|
Min X, Zhang X, Li Y, Cao X, Cheng H, Li Y, Li C, Kong Q, Mao Q, Peng P, Ni Y, Li J, Duan Y, Liu L, Ding Z. HSPA12A unstabilizes CD147 to inhibit lactate export and migration in human renal cell carcinoma. Am J Cancer Res 2020; 10:8573-8590. [PMID: 32754264 PMCID: PMC7392002 DOI: 10.7150/thno.44321] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Metastasis accounts for 90% of cancer-associated mortality in patients with renal cell carcinoma (RCC). However, the clinical management of RCC metastasis is challenging. Lactate export is known to play an important role in cancer cell migration. This study investigated the role of heat shock protein A12A (HSPA12A) in RCC migration. Methods: HSPA12A expression was examined in 82 pairs of matched RCC tumors and corresponding normal kidney tissues from patients by immunoblotting and immunofluorescence analyses. The proliferation of RCC cells was analyzed using MTT and EdU incorporation assays. The migration of RCC cells was evaluated by wound healing and Transwell migration assays. Extracellular acidification was examined using Seahorse technology. Protein stability was determined following treatment with protein synthesis inhibitor cycloheximide and proteasome inhibitor MG132. Mass spectrometry, immunoprecipitation, and immunoblotting were employed to examine protein-protein interactions. Results: RCC tumors from patients showed downregulation of HSPA12A, which was associated with advanced tumor node metastasis stage. Intriguingly, overexpression of HSPA12A in RCC cells inhibited migration, whereas HSPA12A knockdown had the opposite effect. Lactate export, glycolysis rate, and CD147 protein abundance were also inhibited by HSPA12A overexpression but promoted by HSPA12A knockdown. An interaction of HSPA12A with HRD1 ubiquitin E3 ligase was detected in RCC cells. Further studies demonstrated that CD147 ubiquitination and proteasomal degradation were promoted by HSPA12A overexpression whereas inhibited by HSPA12A knockdown. Notably, the HSPA12A overexpression-induced inhibition of lactate export and migration were abolished by CD147 overexpression. Conclusion: Human RCC shows downregulation of HSPA12A. Overexpression of HSPA12A in RCC cells unstabilizes CD147 through increasing its ubiquitin-proteasome degradation, thereby inhibits lactate export and glycolysis, and ultimately suppresses RCC cell migration. Our results demonstrate that overexpression of HSPA12A might represent a viable strategy for managing RCC metastasis.
Collapse
|
4
|
van der Beek JN, Geller JI, de Krijger RR, Graf N, Pritchard-Jones K, Drost J, Verschuur AC, Murphy D, Ray S, Spreafico F, Dzhuma K, Littooij AS, Selle B, Tytgat GAM, van den Heuvel-Eibrink MM. Characteristics and Outcome of Children with Renal Cell Carcinoma: A Narrative Review. Cancers (Basel) 2020; 12:E1776. [PMID: 32635225 PMCID: PMC7407101 DOI: 10.3390/cancers12071776] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
Pediatric renal cell carcinoma (RCC) is a rare type of kidney cancer, most commonly occurring in teenagers and young adolescents. Few relatively large series of pediatric RCC have been reported. Knowledge of clinical characteristics, outcome and treatment strategies are often based on the more frequently occurring adult types of RCC. However, published pediatric data suggest that clinical, molecular and histological characteristics of pediatric RCC differ from adult RCC. This paper summarizes reported series consisting of ≥10 RCC pediatric patients in order to create an up-to-date overview of the clinical and histopathological characteristics, treatment and outcome of pediatric RCC patients.
Collapse
Affiliation(s)
- Justine N. van der Beek
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - James I. Geller
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45229, USA;
| | - Ronald R. de Krijger
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University Medical Center and Saarland University Faculty of Medicine, D-66421 Homburg, Germany;
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (K.P.-J.); (K.D.)
| | - Jarno Drost
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Arnauld C. Verschuur
- Department of Pediatric Oncology, Hôpital d’Enfants de la Timone, APHM, 13005 Marseille, France;
| | - Dermot Murphy
- Department of Paediatric Oncology, Royal Hospital for Children, Glasgow G51 4TF, Scotland; (D.M.); (S.R.)
| | - Satyajit Ray
- Department of Paediatric Oncology, Royal Hospital for Children, Glasgow G51 4TF, Scotland; (D.M.); (S.R.)
| | - Filippo Spreafico
- Pediatric Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy;
| | - Kristina Dzhuma
- UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK; (K.P.-J.); (K.D.)
| | - Annemieke S. Littooij
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht/Wilhelmina Children’s Hospital, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Barbara Selle
- Department of Pediatric Hematology and Oncology, St. Annastift Children’s Hospital, 67065 Ludwigshafen, Germany;
| | - Godelieve A. M. Tytgat
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
| | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (R.R.d.K.); (J.D.); (A.S.L.); (G.A.M.T.); (M.M.v.d.H.-E.)
| |
Collapse
|
5
|
Seles M, Hutterer GC, Foßelteder J, Svoboda M, Resel M, Barth DA, Pichler R, Bauernhofer T, Zigeuner RE, Pummer K, Slaby O, Klec C, Pichler M. Long Non-Coding RNA PANTR1 is Associated with Poor Prognosis and Influences Angiogenesis and Apoptosis in Clear-Cell Renal Cell Cancer. Cancers (Basel) 2020; 12:E1200. [PMID: 32397610 PMCID: PMC7281347 DOI: 10.3390/cancers12051200] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
POU3F3 adjacent non-coding transcript 1 (PANTR1) is an oncogenic long non-coding RNA with significant influence on numerous cellular features in different types of cancer. No characterization of its role in renal cell carcinoma (RCC) is yet available. In this study, PANTR1 expression was confined to human brain and kidney tissue and was found significantly up-regulated in clear-cell renal cell carcinoma tissue (ccRCC) compared to non-cancerous kidney tissue in two independent cohorts (p < 0.001 for both cohorts). In uni- and multivariate Cox regression analysis, ccRCC patients with higher levels of PANTR1 showed significantly poorer disease-free survival in our own respective cohort (n = 175, hazard ratio: 4.3, 95% confidence interval: 1.45-12.75, p = 0.008) in accordance with significantly poorer overall survival in a large The Cancer Genome Atlas database (TCGA) cohort (n = 530, hazard ratio: 2.19, 95% confidence interval: 1.59-3.03, p ≤ 0.001). To study the underlying cellular mechanisms mediated by varying levels of PANTR1 in kidney cancer cells, we applied siRNA-mediated knock-down experiments in three independent ccRCC cell lines (RCC-FG, RCC-MF, 769-P). A decrease in PANTR1 levels led to significantly reduced cellular growth through activation of apoptosis in all tested cell lines. Moreover, as angiogenesis is a critical driver in ccRCC pathogenesis, we identified that PANTR1 expression is critical for in vitro tube formation and endothelial cell migration (p < 0.05). On the molecular level, knock-down of PANTR1 led to a decrease in Vascular Endothelial growth factor A (VEGF-A) and cell adhesion molecule laminin subunit gamma-2 (LAMC2) expression, corroborated by a positive correlation in RCC tissue (for VEGF-A R = 0.19, p < 0.0001, for LAMC2 R = 0.13, p = 0.0028). In conclusion, this study provides first evidence that PANTR1 has a relevant role in human RCC by influencing apoptosis and angiogenesis.
Collapse
Affiliation(s)
- Maximilian Seles
- Department of Urology, Medical University of Graz, 8036 Graz, Austria; (M.S.); (G.C.H.); (R.E.Z.); (K.P.)
| | - Georg C. Hutterer
- Department of Urology, Medical University of Graz, 8036 Graz, Austria; (M.S.); (G.C.H.); (R.E.Z.); (K.P.)
| | - Johannes Foßelteder
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (J.F.); (M.R.); (D.A.B.); (T.B.); (M.P.)
- “Non-coding RNAs and Genome Editing in Cancer” Research Unit, Medical University of Graz, 8036 Graz, Austria
| | - Marek Svoboda
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic; (M.S.); (O.S.)
| | - Margit Resel
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (J.F.); (M.R.); (D.A.B.); (T.B.); (M.P.)
- “Non-coding RNAs and Genome Editing in Cancer” Research Unit, Medical University of Graz, 8036 Graz, Austria
| | - Dominik A. Barth
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (J.F.); (M.R.); (D.A.B.); (T.B.); (M.P.)
- “Non-coding RNAs and Genome Editing in Cancer” Research Unit, Medical University of Graz, 8036 Graz, Austria
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Renate Pichler
- Department of Urology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Thomas Bauernhofer
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (J.F.); (M.R.); (D.A.B.); (T.B.); (M.P.)
| | - Richard E. Zigeuner
- Department of Urology, Medical University of Graz, 8036 Graz, Austria; (M.S.); (G.C.H.); (R.E.Z.); (K.P.)
| | - Karl Pummer
- Department of Urology, Medical University of Graz, 8036 Graz, Austria; (M.S.); (G.C.H.); (R.E.Z.); (K.P.)
| | - Ondrej Slaby
- Central European Institute of Technology (CEITEC), Masaryk University, 62500 Brno, Czech Republic; (M.S.); (O.S.)
- Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Christiane Klec
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (J.F.); (M.R.); (D.A.B.); (T.B.); (M.P.)
- “Non-coding RNAs and Genome Editing in Cancer” Research Unit, Medical University of Graz, 8036 Graz, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria; (J.F.); (M.R.); (D.A.B.); (T.B.); (M.P.)
- “Non-coding RNAs and Genome Editing in Cancer” Research Unit, Medical University of Graz, 8036 Graz, Austria
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
6
|
Taneja K, Williamson SR. Updates in Pathologic Staging and Histologic Grading of Renal Cell Carcinoma. Surg Pathol Clin 2018; 11:797-812. [PMID: 30447842 DOI: 10.1016/j.path.2018.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The most important prognostic parameter in renal cell carcinoma is tumor stage. Although pathologic primary tumor (pT) categories are influenced by tumor size (pT1-pT2), critical elements (≥pT3) are dictated by invasion of structures, including renal sinus, perinephric fat, and the renal vein or segmental branches. Because this invasion can be subtle, awareness of the unique characteristics of renal cell carcinoma is critical for the pathologist to aid in clinical decision making. This review addresses challenges in pathologic stage and grade reporting and updates to the World Health Organization and American Joint Commission on Cancer classification schemes.
Collapse
Affiliation(s)
- Kanika Taneja
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Henry Ford Hospital, K6, W615, 2799 West Grand Boulevard, Detroit, MI 48202, USA
| | - Sean R Williamson
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Henry Ford Hospital, K6, W615, 2799 West Grand Boulevard, Detroit, MI 48202, USA; Department of Pathology and Laboratory Medicine, Wayne State University School of Medicine, 2799 West Grand Boulevard, Detroit, MI 48202, USA.
| |
Collapse
|
7
|
Seles M, Hutterer GC, Kiesslich T, Pummer K, Berindan-Neagoe I, Perakis S, Schwarzenbacher D, Stotz M, Gerger A, Pichler M. Current Insights into Long Non-Coding RNAs in Renal Cell Carcinoma. Int J Mol Sci 2016; 17:573. [PMID: 27092491 PMCID: PMC4849029 DOI: 10.3390/ijms17040573] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/08/2016] [Accepted: 04/12/2016] [Indexed: 02/07/2023] Open
Abstract
Renal cell carcinoma (RCC) represents a deadly disease with rising mortality despite intensive therapeutic efforts. It comprises several subtypes in terms of distinct histopathological features and different clinical presentations. Long non-coding RNAs (lncRNAs) are non-protein-coding transcripts in the genome which vary in expression levels and length and perform diverse functions. They are involved in the inititation, evolution and progression of primary cancer, as well as in the development and spread of metastases. Recently, several lncRNAs were described in RCC. This review emphasises the rising importance of lncRNAs in RCC. Moreover, it provides an outlook on their therapeutic potential in the future.
Collapse
Affiliation(s)
- Maximilian Seles
- Department of Urology, Medical University of Graz, A-8036 Graz, Austria.
| | - Georg C Hutterer
- Department of Urology, Medical University of Graz, A-8036 Graz, Austria.
| | - Tobias Kiesslich
- Department of Internal Medicine I, Salzburger Landeskliniken (SALK), Paracelsus Medical University, A-5020 Salzburg, Austria.
- Laboratory for Tumour Biology and Experimental Therapies, Institute of Physiology and Pathophysiology, Paracelsus Medical University, A-5020 Salzburg, Austria.
| | - Karl Pummer
- Department of Urology, Medical University of Graz, A-8036 Graz, Austria.
| | - Ioana Berindan-Neagoe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
- Research Center of Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
- Department of Experimental Pathology, The Oncology Institute Ion Chiricuta, 400015 Cluj-Napoca, Romania.
| | - Samantha Perakis
- Institute of Human Genetics, Medical University of Graz, A-8036 Graz, Austria.
| | - Daniela Schwarzenbacher
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
| | - Michael Stotz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
- Center for Biomarker Research in Medicine, Medical University of Graz, A-8036 Graz, Austria.
| | - Martin Pichler
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, A-8036 Graz, Austria.
| |
Collapse
|
8
|
Lin YL, Gui SL, Guo H, Ma JG, Li WP. Protocadherin17 Promoter Methylation is a Potential Predictive Biomarker in Clear Cell Renal Cell Carcinoma. Med Sci Monit 2015; 21:2870-6. [PMID: 26404644 PMCID: PMC4588677 DOI: 10.12659/msm.895603] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Protocadherin17 (PCDH17) is a tumor suppressor gene, and is frequently silenced by promoter methylation in human cancers, including clear cell renal cell carcinoma (ccRCC). However, the clinical significance of PCDH17 methylation in ccRCC remains largely unclear. The aim of the present study was to investigate the methylation status of PCDH17 in ccRCC and its potential relevance to clinicopathological parameters and prognosis. Material/Methods Methylation-specific PCR was used to examine the methylation status of PCDH17 in 191 ccRCC tumors and matched paired adjacent noncancerous tissues. Subsequently, the associations between PCDH17 methylation and clinicopathological parameters and prognosis of patients with ccRCC were analyzed. Results PCDH17 methylation occurred in 66.5% of ccRCC tumors, but in only 12.1% of adjacent noncancerous tissues. PCDH17 methylation is significantly correlated with advanced stage, higher grade, and lymph node metastasis in ccRCC. Moreover, it is an independent prognostic factor for progression-free survival and overall survival of patients with ccRCC. Conclusions PCDH17 methylation occurred more frequently and was associated with malignant clinicopathological characteristics and poor prognosis in ccRCC patients. Thus, PCDH17 methylation may be used as a novel biomarker to predict the prognosis of patients with ccRCC.
Collapse
Affiliation(s)
- Ying-Li Lin
- Department of Urology, Affiliated Xuzhou Hospital of Jiangsu University (Xuzhou Cancer Hospital), Xuzhou, Jiangsu, China (mainland)
| | - Shi-Liang Gui
- Department of Urology, First Hospital of Jiamusi University, Jiamusi, Heilongjiang, China (mainland)
| | - Hong Guo
- Department of Pharmacy, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Jian-Guo Ma
- Department of Urology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Wen-Ping Li
- Department of Urology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
9
|
Fernandes DS, Lopes JM. Pathology, therapy and prognosis of papillary renal carcinoma. Future Oncol 2015; 11:121-32. [PMID: 25572787 DOI: 10.2217/fon.14.133] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Papillary renal cell carcinoma (pRCC) accounts for approximately 10% of renal parenchymal tumors. There are two pRCC subtypes reported in several studies, but at present, there is limited molecular evidence to validate this pRCC subtyping in the daily routine. The utility of subtyping pRCC is based on reports describing that pRCC subtype is an independent predictor of outcome, with type 1 tumors showing significantly better survival than type 2 tumors. In this article, we summarize the relevant knowledge on pRCC regarding tumor features: clinical presentation, histopathology, electron microscopy, immunohistochemistry, cytogenetics, genetic/molecular and prognosis. We present an overview of the currently available pRCC treatment options and some of the new promising agents.
Collapse
|
10
|
Abstract
Renal cell carcinoma (RCC) represents the sixth-leading cancer-specific cause of death worldwide. This is mainly caused by metastatic or locally advanced RCC. Approximately 25-30% of patients present with metastasis during the initial diagnosis. Furthermore, 20-30% of patients develop metastatic disease following initial curative surgery. Metastatic RCC is characterized by a poor prognosis with a median overall survival of less than 2 years. Today, targeted therapies such as VEGF receptor inhibitors and antagonists as well as mTOR inhibitors represent the standard of care in metastatic RCC. Conventional chemotherapies or cytokine-based medications have been abandoned due to inferior clinical efficacy compared with targeted therapies. In Germany, sunitinib, pazopanib, temsirolimus, and bevacizumab have been approved for first-line treatment and sorafenib, axitinib, and everolimus for second-line treatment. Prognostic models, assessing individual risk profiles, have been developed in the last 15 years, which are crucial for the design of trials, patient counseling, and initiation of goal-directed therapies.
Collapse
|
11
|
Lin YL, Wang YL, Fu XL, Ma JG. Aberrant methylation of PCDH8 is a potential prognostic biomarker for patients with clear cell renal cell carcinoma. Med Sci Monit 2014; 20:2380-5. [PMID: 25416427 PMCID: PMC4251547 DOI: 10.12659/msm.892433] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/02/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND PCDH8 is a tumor suppressor that regulates cell adhesin, proliferation, and migration. It is often inactivated by aberrant promoter methylation in several human cancers, including clear cell renal cell carcinoma (CCRCC). The clinical significance of PCDH8 methylation in CCRCC remains unclear. The aim of this study was to investigate the relationship between PCDH8 methylation and clinicopathological characteristics as well as outcome of patients with CCRCC. MATERIAL/METHODS The methylation status of PCDH8 in 153 CCRCC tissues and 97 paired adjacent normal renal tissues were examined using methylation-specific PCR (MSP). Then the relationships between PCDH8 methylation and clinicopathological features as well as progression-free survival of CCRCC patients were evaluated. RESULTS PCDH8 methylation was significantly more frequent in CCRCC tissues compared with normal renal tissues. Moreover, PCDH8 methylation was significantly correlated with advanced clinical stage (P=0.0141), higher grade (P=0.0190), and lymph node metastasis (P=0.0098). In addition, multivariate analysis showed that PCDH8 methylation was independently associated with poor progression-free survival (P=0.0316). CONCLUSIONS PCDH8 methylation is a frequent event in CCRCC and is correlated with unfavorable clinicopathological features. Moreover, PCDH8 methylation may be a useful biomarker to predict the progression of CCRCC.
Collapse
Affiliation(s)
- Ying-Li Lin
- Department of Urology, Affiliated Xuzhou Hospital of Jiangsu University (Xuzhou Cancer Hospital), Xuzhou, Jiangsu, China
| | - Yan-Ling Wang
- Department of Anesthesiology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xing-Li Fu
- Health Sciences Center, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jian-Guo Ma
- Department of Urology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
12
|
Su B, Zhao W, Shi B, Zhang Z, Yu X, Xie F, Guo Z, Zhang X, Liu J, Shen Q, Wang J, Li X, Zhang Z, Zhou L. Let-7d suppresses growth, metastasis, and tumor macrophage infiltration in renal cell carcinoma by targeting COL3A1 and CCL7. Mol Cancer 2014; 13:206. [PMID: 25193015 PMCID: PMC4168121 DOI: 10.1186/1476-4598-13-206] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/26/2014] [Indexed: 01/10/2023] Open
Abstract
Background MicroRNAs are endogenous small noncoding RNAs that are functionally involved in numerous critical cellular processes including tumorigenesis. Data mining using a microRNA array database suggested that let-7d microRNA may be associated with renal cell carcinoma (RCC) malignant progression. Here, we performed further analyses to determine whether let-7d is functionally linked to RCC malignancy. Methods Quantitative real-time PCR was used to determine the level of mature let-7d in RCC clinical specimens and its correlation with clinicopathological data. Immunohistochemical staining was conducted to characterize the stroma of RCC. Let-7d overexpressing RCC cell lines combined with mouse models bearing cell-derived xenografts and patient-derived xenografts were used to assess the functional role of let-7d in vitro and in vivo. Results Downregulation of let-7d in clinical RCC samples was associated with advanced tumor grade and T stage and increased vascular invasion. An inverse relationship between let-7d expression and macrophage infiltration was found in clinical RCC samples. Functional studies indicated that ectopic expression of let-7d significantly inhibited RCC cell proliferation, migration, and peripheral blood monocyte (PBMC) recruitment in vitro, as well as tumor growth, metastasis, and tumor macrophage infiltration in vivo. In silico analysis and subsequent experimental validation confirmed collagen, type III, alpha 1 (COL3A1) and C-C subfamily chemokine member CCL7 as direct let-7d target genes. The addition of COL3A1 and CCL7 counteracted the inhibitory effects of let-7d on RCC cell proliferation, migration, and PBMC recruitment. The inhibition of let-7d increased cell proliferation, migration, and PBMC recruitment by the enhanced expression of COL3A1 and CCL7 genes in vitro. The mRNA levels of COL3A1 and CCL7 were inversely correlated with let-7d level in RCC clinical specimens. Conclusions These results suggest that let-7d may suppress RCC growth, metastasis, and tumor macrophage infiltration at least partially through targeting COL3A1 and CCL7. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-206) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Zhiqian Zhang
- Department of Urology, Peking University First Hospital & the Institute of Urology, Peking University, Beijing 100034, China.
| | | |
Collapse
|