1
|
Zhang Y, Liu Y, Hu X, Song F, Zheng S, Zheng X, Sun J, Li L, Huang P. Stemness-associated senescence genes as potential novel risk factors for papillary renal cell carcinoma. Transl Androl Urol 2022; 10:4241-4252. [PMID: 34984189 PMCID: PMC8661265 DOI: 10.21037/tau-21-913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/10/2021] [Indexed: 11/06/2022] Open
Abstract
Background Papillary renal cell carcinoma (PRCC) is the 2nd most common type of renal carcinoma; however, there is limited data about PRCC, and strategies for the diagnosis and treatment of PRCC need to be identified. Methods In this study, the stemness-associated senescence (SAS) phenotype of PRCC was obtained by a bioinformatics analysis. We acquired the gene expression profiles of patients with PRCC and calculated the PRCC messenger ribonucleic acid stemness index (mRNAsi). We then screened the SAS genes from the GenAge database. A least absolute shrinkage and selection operator-Cox regression was conducted to examine correlations between risk signatures and the abundance of the SAS genes in the PRCC samples. Functional enrichment analyses were then performed via molecular co-expression studies of mRNAsi, and the risk scores of PRCC patients were calculated. Results We identified the following 8 SAS signatures that were strongly associated with prognosis in PRCC patients: cyclin-dependent kinase 1, heat shock protein family D member 1, platelet-derived growth factor receptor A, cyclin-dependent kinase inhibitor 2B, pyrroline-5-carboxylate reductase 1, sequestosome-1, sirtuin-3, and cyclin-dependent kinase inhibitor 1A. The SAS signatures were significantly associated with the stage and type of PRCC. The calculated risk scores can be used to divide PRCC patients into low- and high-risk groups, and provide guidance in determining treatment plans. Conclusions We have developed a reliable prognostic tool to predict the clinical outcomes of PRCC patients. This tool could improve treatment decisions regarding drug therapy, surgery, and conservative options.
Collapse
Affiliation(s)
- Yiwen Zhang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| | - Yujia Liu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoping Hu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Feifeng Song
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shuilian Zheng
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaowei Zheng
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiao Sun
- Department of Pharmacy, Institute of Cancer Research and Basic Medical Sciences of the Chinese Academy of Sciences, Cancer Hospital of the University of the Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Li
- Department of Pharmacy, The First People's Hospital of Chun An, Hangzhou, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.,Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
2
|
Huang R, Meens J, Yuzwa S, Ailles L, Ohh M, Robinson CM. Side population analysis in clear cell renal cell carcinoma. Biochem Biophys Res Commun 2021; 585:196-202. [PMID: 34813980 DOI: 10.1016/j.bbrc.2021.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 10/19/2022]
Abstract
Cancer stem cells have an important role in tumour biology. While their identity in haematological malignancies is clearly defined, stem cell identity remains elusive in some solid tumours. Clear cell renal cell carcinoma (ccRCC) represents the most common form of kidney cancer, but the identity or existence of ccRCC stem cells remains unknown. We aimed to discern their existence using the widely utilised side population approach in ccRCC cell lines. In all cells tested, a well-defined side population was identified, and cell-based assays suggested stem-like properties. However, limiting dilution assays revealed comparable tumour initiating abilities and tumour histology of side and non-side populations, and single cell RNA-sequencing revealed minimal differences between these populations. The results indicate that the side population approach is not sufficient for cancer stem cell discovery in ccRCC.
Collapse
Affiliation(s)
- Richard Huang
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5G 1M1, Canada.
| | - Jalna Meens
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 1L7, Canada.
| | - Scott Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5G 1M1, Canada.
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M5G 1L7, Canada.
| | - Michael Ohh
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5G 1M1, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, M5G 1M1, Canada.
| | - Claire M Robinson
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5G 1M1, Canada; Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, Galway, H91 FD82, Ireland.
| |
Collapse
|
3
|
Liu H, Liu C, Qu Y. The effect and molecular mechanism of hypoxia on proliferation and apoptosis of CD133+ renal stem cells. Bosn J Basic Med Sci 2021; 21:313-322. [PMID: 32767964 PMCID: PMC8112556 DOI: 10.17305/bjbms.2020.4887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/30/2020] [Indexed: 11/24/2022] Open
Abstract
Congenital hydronephrosis caused by ureteropelvic junction obstruction (UPJO) eventually leads to renal interstitial fibrosis and atrophy, after a series of pathophysiological problems. Renal repair after injury depends on renal stem cells. This study aimed to determine the expression of renal stem cell marker CD133 in children of different ages and the regulatory effect of stem cell microenvironment. Renal stem cells from children of different ages were identified and screened out by flow cytometry in the study. Children with hydronephrosis were divided into neonates, infants, preschool age, school age, and adolescents groups. A hypoxic cell model prepared with CoCl2 was developed to detect the effect of hypoxia on the proliferation and apoptosis of renal stem cells. The effect and molecular mechanism of hypoxia-inducible factor 1-alpha (HIF-1α) on the proliferation and apoptosis of renal stem cells were also explored. Both hypoxia and HIF-1α significantly promoted the proliferation of renal stem cells and inhibited cell apoptosis. HIF-1α could bind to the promoter region of proliferating cell nuclear antigen (PCNA) and PROM1 (CD133) to mediate their transcription and expression. The content of CD133+ renal stem cells was the highest in the neonatal group and it decreased with the increase of age. Taken together, this study clarified the effect of age on the content of human renal stem cells and determined the regulatory mechanism of hypoxia on renal stem cells. We expect our results to provide a research basis for the treatment and clinical application of renal stem cells.
Collapse
Affiliation(s)
- Hong Liu
- Department of Pediatric General Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Cui Liu
- Department of Pediatric General Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yan Qu
- Department of Pediatric General Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
4
|
Singh D. Current updates and future perspectives on the management of renal cell carcinoma. Life Sci 2020; 264:118632. [PMID: 33115605 DOI: 10.1016/j.lfs.2020.118632] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) refers to renal-epithelial cancer, which represents over 90% of kidney cancer and is a cause for cancer related deaths in the world. Studies suggested somatic VHL mutations to be the cause for the occurrence of cancer, but with the time, more latest genomic and biological studies have detected variation in epigenetic regulatory genes and showed significant heterogeneity of the intratumor that may lead to strategies of diagnostic, predictive, and therapeutic importance. Immune dysfunction is responsible for almost all types of renal cancer, and angiogenesis and immunosuppression function together in the tumor microenvironment of renal cell carcinoma (RCC). Over the past few years, advancement in the management of the RCC has finally revolutionized with the arrival of the entrapped immune inhibitors which particularly concentrated on the receptor (programmed cell death-1) and focus on the new generation receptor i.e. TKRI (tyrosine-kinase receptor inhibitors). The present review deals with the comprehensive review of RCC and emphasizes on its types, pathogenesis and advancement in these diseases. This review also overviews the role of innate and adaptive immune response-related mechanism, the function of cancer stem cell in this diseases, therapeutic targeted drugs and hormonal signaling pathways as an emerging strategy in the management of the renal cancer.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad 211007, Uttar Pradesh, India.
| |
Collapse
|
5
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
6
|
Corrò C, Healy ME, Engler S, Bodenmiller B, Li Z, Schraml P, Weber A, Frew IJ, Rechsteiner M, Moch H. IL-8 and CXCR1 expression is associated with cancer stem cell-like properties of clear cell renal cancer. J Pathol 2019; 248:377-389. [PMID: 30883740 PMCID: PMC6618115 DOI: 10.1002/path.5267] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/25/2019] [Accepted: 03/13/2019] [Indexed: 12/28/2022]
Abstract
Recent studies suggest that clear cell renal cell carcinoma (ccRCC) possesses a rare population of cancer stem cells (CSCs) that might contribute to tumor heterogeneity, metastasis and therapeutic resistance. Nevertheless, their relevance for renal cancer is still unclear. In this study, we successfully isolated CSCs from established human ccRCC cell lines. CSCs displayed high expression of the chemokine IL‐8 and its receptor CXCR1. While recombinant IL‐8 significantly increased CSC number and properties in vitro, CXCR1 inhibition using an anti‐CXCR1 antibody or repertaxin significantly reduced these features. After injection into immune‐deficient mice, CSCs formed primary tumors that metastasized to the lung and liver. All xenografted tumors in mice expressed high levels of IL‐8 and CXCR1. Furthermore, IL‐8/CXCR1 expression significantly correlated with decreased overall survival in ccRCC patients. These results suggest that the IL‐8/CXCR1 phenotype is associated with CSC‐like properties in renal cancer. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Claudia Corrò
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland.,Life Science Zurich Graduate School, ETH and University of Zurich, Zurich, Switzerland
| | - Marc E Healy
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Stefanie Engler
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Zhe Li
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Schraml
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Ian J Frew
- Clinic of Internal Medicine I, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Markus Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Gao L, Fan YZ, Zhang TH, Xu HQ, Zeng XL, Hou T, Dan WC, Zeng J, An RF. Biocompatible carbon-doped MoSe2 nanoparticles as a highly efficient targeted agent for human renal cell carcinoma. RSC Adv 2019; 9:11567-11575. [PMID: 35520231 PMCID: PMC9063525 DOI: 10.1039/c9ra01029g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022] Open
Abstract
MoSe2 is a typical transition-metal dichalcogenide material, and many researches have been focused on using its property of near infrared strong absorption for laser mediated photothermal cancer treatment. However, the anti-canter effect of MoSe2 and its possible mechanism in renal cell carcinoma (RCC) is still unclear. RCC has high incidence of metastasis, which is known as one of the most lethal malignancies in the urological system. This study revealed that the carbon-doped MoSe2 particles can obviously inhibit proliferation for 786-O and ACHN cells. Meanwhile, the carbon-doped MoSe2 nanoparticles have little impact on the viability of KH-2 cells in vitro. The mechanism analysis revealed that the carbon-doped MoSe2 particles have hydrogen bond effect in aqueous solution, and the particle aggregation effect caused the KH-2 cells to have high viability. The carbon-doped MoSe2 nanoparticles with minimal toxicity may be a potential therapeutic candidate against RCC. HK-2 cells have weak cellular uptake efficiency leading to high viability with carbon-doped MoSe2 nanoparticles.![]()
Collapse
Affiliation(s)
- Li Gao
- Department of Gynaecology and Obstetrics
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Yi-zeng Fan
- Department of Urology
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Tao-hong Zhang
- Department of Gynaecology and Obstetrics
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Hui-qiu Xu
- Department of Gynaecology and Obstetrics
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Xian-ling Zeng
- Department of Gynaecology and Obstetrics
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Tao Hou
- Department of Urology
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Wei-chao Dan
- Department of Urology
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Jin Zeng
- Department of Urology
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| | - Rui-fang An
- Department of Gynaecology and Obstetrics
- The First Affiliated Hospital of Xi'an Jiaotong University
- Health Science Center
- Xi'an Jiaotong University
- Xi'an
| |
Collapse
|
8
|
Ge Y, Weygant N, Qu D, May R, Berry WL, Yao J, Chandrakesan P, Zheng W, Zhao L, Zhao KL, Drake M, Vega KJ, Bronze MS, Tomasek JJ, An G, Houchen CW. Alternative splice variants of DCLK1 mark cancer stem cells, promote self-renewal and drug-resistance, and can be targeted to inhibit tumorigenesis in kidney cancer. Int J Cancer 2018; 143:1162-1175. [PMID: 29577277 DOI: 10.1002/ijc.31400] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/17/2018] [Accepted: 03/08/2018] [Indexed: 12/21/2022]
Abstract
Renal cell carcinoma (RCC) is a common and devastating disease characterized by a hypoxic microenvironment, epithelial-mesenchymal transition and potent resistance to therapy evidencing the presence of cancer stem cells (CSCs). Various CSC markers have been studied in RCC, but overall there is limited data on their role and most markers studied have been relatively nonspecific. Doublecortin-like kinase 1 (DCLK1) is a validated CSC marker in the gastrointestinal tract and evidence for an equivalent role in other cancers is accumulating. We used bioinformatics, immunohistochemistry, flow cytometry, spheroid self-renewal and chemoresistance assays in combination with overexpression and siRNA-knockdown to study the stem cell-supportive role of DCLK1 alternative splice variants (DCLK1 ASVs) in RCC. To target tumor cells expressing DCLK1 ASVs directly, we developed a novel monoclonal antibody (CBT-15) and delivered it systemically to RCC tumor xenografts. DCLK1 ASVs were overexpressed, enriched together with CSC markers and predictive of overall and recurrence-free survival in RCC patients. In vitro, DCLK1 ASVs were able to directly stimulate essential molecular and functional characteristics of renal CSCs including expression of aldehyde dehydrogenase, self-renewal and resistance to FDA-approved receptor tyrosine kinase and mTOR inhibitors, while targeted downregulation of DCLK1 reversed these characteristics. Finally, targeting DCLK1 ASV-positive cells with the novel CBT-15 monoclonal antibody blocked RCC tumorigenesis in vivo. These findings establish DCLK1 as a CSC marker with implications for therapy, disease progression and survival in RCC and demonstrate the therapeutic value of DCLK1-targeted monoclonal antibodies against renal CSCs.
Collapse
Affiliation(s)
- Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Nathaniel Weygant
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,COARE Biotechnology Inc., Oklahoma City, OK
| | - Dongfeng Qu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,COARE Biotechnology Inc., Oklahoma City, OK.,The Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK.,United States Department of Veterans Affairs Medical Center, Oklahoma City, OK
| | - Randal May
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,COARE Biotechnology Inc., Oklahoma City, OK.,United States Department of Veterans Affairs Medical Center, Oklahoma City, OK
| | - William L Berry
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Jiannan Yao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Parthasarathy Chandrakesan
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,COARE Biotechnology Inc., Oklahoma City, OK.,The Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK
| | - Wei Zheng
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Lichao Zhao
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Karena L Zhao
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Michael Drake
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Kenneth J Vega
- Department of Medicine, National Jewish Health, Denver, CO
| | - Michael S Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - James J Tomasek
- Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,COARE Biotechnology Inc., Oklahoma City, OK.,The Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK.,United States Department of Veterans Affairs Medical Center, Oklahoma City, OK
| |
Collapse
|
9
|
Zhao CL, Amin A, Hui Y, Yang D, Cao W. TGR5 expression in normal kidney and renal neoplasms. Diagn Pathol 2018; 13:22. [PMID: 29606134 PMCID: PMC5880016 DOI: 10.1186/s13000-018-0700-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/20/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The G protein-coupled bile acid receptor (TGR5) is a cell surface receptor which induces the production of intracellular cAMP and promotes epithelial-mesenchymal transition in gastric cancer cell lines. TGR5 is found in a wide variety of tissues including the kidney. However, the patterns of TGR5 expression have not been well characterized in physiologic kidney or renal neoplasms. We explore the expression of TGR5 in benign renal tissue and renal neoplasms and assess its utility as a diagnostic marker. METHODS Sixty-one renal cortical neoplasms from 2000 to 2014 were retrieved. TGR5 protein expression was examined by immunohistochemistry. TGR5 mRNA was also measured by real-time PCR. RESULTS In normal renal tissue, TGR5 was strongly positive in collecting ducts, distal convoluted tubules and thin loop of Henle. Proximal convoluted tubules showed absent or focal weak staining. In clear cell renal cell carcinomas (RCCs), 25 of 27 cases (92%) were negative for TGR5 (p < 0.001). TGR5 mRNA was also significantly decreased in clear cell RCCs, suggesting that decreased TGR5 protein expression may be attributable to the downregulation of TGR5 mRNA in these tumors. All 11 papillary RCCs expressed TGR5 with 45% (5/11) exhibiting moderate to strong staining. All chromophobe RCCs and oncocytomas were positive for TGR5 with weak to moderate staining. TGR5 mRNA expression in these tumors was similar to normal kidney. All urothelial carcinomas of the renal pelvis strongly expressed TGR5 including a poorly differentiated urothelial carcinoma with sarcomatoid features. CONCLUSION TGR5 is strongly expressed in collecting ducts, distal convoluted tubules and thin loop of Henle. TGR5 protein and mRNA expression were notably decreased in clear cell RCCs and may be helpful in differentiating these tumors from other RCCs.
Collapse
Affiliation(s)
- Chaohui Lisa Zhao
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Ali Amin
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Yiang Hui
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Dongfang Yang
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA
| | - Weibiao Cao
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 12, Providence, RI, 02903, USA. .,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| |
Collapse
|
10
|
Song L, Ye W, Cui Y, Lu J, Zhang Y, Ding N, Hu W, Pei H, Yue Z, Zhou G. Ecto-5'-nucleotidase (CD73) is a biomarker for clear cell renal carcinoma stem-like cells. Oncotarget 2018; 8:31977-31992. [PMID: 28404888 PMCID: PMC5458263 DOI: 10.18632/oncotarget.16667] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 03/17/2017] [Indexed: 12/16/2022] Open
Abstract
Identification of a specific biomarker for cancer stem cells (CSCs) is of potential applications in the development of effective therapeutic strategies for renal cell carcinoma (RCC). In this study, both the RCC cell line 786-O and surgically removed clear cell RCC (ccRCC) tissues were implemented to grew as spheroids in serum-free medium supplemented with mitogens. This subpopulation possessed key characteristics defining CSCs. We also identified that surgically removed ccRCC tissues were heterogenic and there was a subpopulation of cells that was highly stained with rhodamine-123. Based on membrane-proteomic analyses, CD73 was identified as a candidate biomarker. We further found that CD73high cells were highly tumorigenic. As few as 100 CD73high cells were capable of forming xenograft tumors in non obese diabetic/severe combined immunodeficiency disease mice, whereas 1 × 105 CD73low cells did not initiate tumor formation. During successive culture, the CD73high population regenerated both CD73high and CD73low cells, whereas the CD73low population remained low expression level of CD73. Furthermore, the CD73high cells were more resistant to radiation and DNA-damaging agents than the CD73low cells, and expressed a panel of 'stemness' genes at a higher level than the CD73low cells. These findings suggest that a high level of CD73 expression is a bona fide biomarker of ccRCC stem-like cells. Future research will aim at the elucidation of the underlying mechanisms of CD73 in RCC development and the distinct aspects of ccRCC stem-like cells from other tumor types.
Collapse
Affiliation(s)
- Lei Song
- Medical College, Northwest Minzu University, Lanzhou 730030, China.,Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Wenling Ye
- Medical College, Henan University, Kaifeng 475001, China.,Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yong Cui
- Department of Urology Surgery, Shuyang Hospital of Traditional Chinese Medicine, Suqian 223600, China.,Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jianzhong Lu
- Institute of Urology, Department of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Yanan Zhang
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Nan Ding
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modem Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Wentao Hu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Hailong Pei
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Zhongjin Yue
- Institute of Urology, Department of Urology, Gansu Nephro-Urological Clinical Center, Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Guangming Zhou
- School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| |
Collapse
|
11
|
Hasmim M, Bruno S, Azzi S, Gallerne C, Michel JG, Chiabotto G, Lecoz V, Romei C, Spaggiari GM, Pezzolo A, Pistoia V, Angevin E, Gad S, Ferlicot S, Messai Y, Kieda C, Clay D, Sabatini F, Escudier B, Camussi G, Eid P, Azzarone B, Chouaib S. Isolation and characterization of renal cancer stem cells from patient-derived xenografts. Oncotarget 2017; 7:15507-24. [PMID: 26551931 PMCID: PMC4941257 DOI: 10.18632/oncotarget.6266] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 01/06/2023] Open
Abstract
As rapidly developing patient-derived xenografts (PDX) could represent potential sources of cancer stem cells (CSC), we selected and characterized non-cultured PDX cell suspensions from four different renal carcinomas (RCC). Only the cell suspensions from the serial xenografts (PDX-1 and PDX-2) of an undifferentiated RCC (RCC-41) adapted to the selective CSC medium. The cell suspension derived from the original tumor specimen (RCC-41-P-0) did not adapt to the selective medium and strongly expressed CSC-like markers (CD133 and CD105) together with the non-CSC tumor marker E-cadherin. In comparison, PDX-1 and PDX-2 cells exhibited evolution in their phenotype since PDX-1 cells were CD133high/CD105-/Ecadlow and PDX-2 cells were CD133low/CD105-/Ecad-. Both PDX subsets expressed additional stem cell markers (CD146/CD29/OCT4/NANOG/Nestin) but still contained non-CSC tumor cells. Therefore, using different cell sorting strategies, we characterized 3 different putative CSC subsets (RCC-41-PDX-1/CD132+, RCC-41-PDX-2/CD133-/EpCAMlow and RCC-41-PDX-2/CD133+/EpCAMbright). In addition, transcriptomic analysis showed that RCC-41-PDX-2/CD133− over-expressed the pluripotency gene ERBB4, while RCC-41-PDX-2/CD133+ over-expressed several tumor suppressor genes. These three CSC subsets displayed ALDH activity, formed serial spheroids and developed serial tumors in SCID mice, although RCC-41-PDX-1/CD132+ and RCC-41-PDX-2/CD133+ displayed less efficiently the above CSC properties. RCC-41-PDX-1/CD132+ tumors showed vessels of human origin with CSC displaying peri-vascular distribution. By contrast, RCC-41-PDX-2 originated tumors exhibiting only vessels of mouse origin without CSC peri-vascular distribution. Altogether, our results indicate that PDX murine microenvironment promotes a continuous redesign of CSC phenotype, unmasking CSC subsets potentially present in a single RCC or generating ex novo different CSC-like subsets.
Collapse
Affiliation(s)
- Meriem Hasmim
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France.,INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France
| | - Stefania Bruno
- Department of Molecular Biotechnology and Healthy Science, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Sandy Azzi
- INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France
| | - Cindy Gallerne
- INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France
| | - Julien Giron Michel
- INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France
| | - Giulia Chiabotto
- Department of Medical Science, University of Torino, Medical School, Torino, Italy
| | - Vincent Lecoz
- INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France
| | | | | | | | - Vito Pistoia
- Laboratory of Oncology Giannina Gaslini Institute, Genoa, Italy
| | - Eric Angevin
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France.,Medical Oncology Department, Gustave Roussy Campus, Villejuif, France
| | - Sophie Gad
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France.,Laboratoire de Génétique Oncologique EPHE, Ecole Pratique des Hautes Etudes, Paris, France
| | - Sophie Ferlicot
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France.,Université Paris-Sud, Assistance Publique-Hôpitaux de Paris, Service d'Anatomo-Pathologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Yosra Messai
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans, France
| | - Denis Clay
- INSERM UMR 972, Paul Brousse Hospital, Villejuif, France
| | - Federica Sabatini
- Stem Cell and Cell Therapy Laboratory, Istituto G. Gaslini, Genoa, Italy
| | - Bernard Escudier
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France.,Medical Oncology Department, Gustave Roussy Campus, Villejuif, France
| | - Giovanni Camussi
- Department of Medical Science, University of Torino, Medical School, Torino, Italy
| | - Pierre Eid
- INSERM UMR 1014, Lavoisier Building, Paul Brousse Hospital, Villejuif, France
| | | | - Salem Chouaib
- INSERM U 1186, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy Campus, Villejuif, France
| |
Collapse
|
12
|
Zhang XF, Weng DS, Pan K, Zhou ZQ, Pan QZ, Zhao JJ, Tang Y, Jiang SS, Chen CL, Li YQ, Zhang HX, Chang AE, Wicha MS, Zeng YX, Li Q, Xia JC. Dendritic-cell-based immunotherapy evokes potent anti-tumor immune responses in CD105+ human renal cancer stem cells. Mol Carcinog 2017; 56:2499-2511. [DOI: 10.1002/mc.22697] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Xiao-Fei Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - De-sheng Weng
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Ke Pan
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Zi-Qi Zhou
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Qiu-zhong Pan
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Yan Tang
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Shan-Shan Jiang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Chang-Long Chen
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Yong-Qiang Li
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Hong-Xia Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| | - Alfred E. Chang
- University of Michigan Comprehensive Cancer Center; Ann Arbor Michigan
| | - Max S. Wicha
- University of Michigan Comprehensive Cancer Center; Ann Arbor Michigan
| | | | - Qiao Li
- University of Michigan Comprehensive Cancer Center; Ann Arbor Michigan
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center; Guangzhou People's Republic of China
- Department of Biotherapy; Sun Yat-Sen University Cancer Center; Guangzhou People's Republic of China
| |
Collapse
|
13
|
Progression of Human Renal Cell Carcinoma via Inhibition of RhoA-ROCK Axis by PARG1. Transl Oncol 2017; 10:142-152. [PMID: 28131798 PMCID: PMC5284488 DOI: 10.1016/j.tranon.2016.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 01/20/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most lethal urological malignancy with high risk of recurrence; thus, new prognostic biomarkers are needed. In this study, a new RCC antigen, PTPL1 associated RhoGAP1 (PARG1), was identified by using serological identification of recombinant cDNA expression cloning with sera from RCC patients. PARG1 protein was found to be differentially expressed in RCC cells among patients. High PARG1 expression is significantly correlated with various clinicopathological factors relating to cancer cell proliferation and invasion, including G3 percentage (P = .0046), Ki-67 score (p expression is also correlated with high recurrence of N0M0 patients (P = .0084) and poor prognosis in RCC patients (P = .0345). Multivariate analysis has revealed that high PARG1 expression is an independent factor for recurrence (P = .0149) of N0M0 RCC patients. In in vitro studies, depletion of PARG1by siRNA in human RCC cell lines inhibited their proliferation through inducing G1 cell cycle arrest via upregulation of p53 and subsequent p21Cip1/Waf1, which are mediated by increased RhoA-ROCK activities. Similarly, PARG1 depletion cells inhibited invasion ability via increasing RhoA-ROCK activities in the RCC cell lines. Conversely, overexpression of PARG1 on human embryonic kidney cell line HEK293T promotes its cell proliferation and invasion. These results indicate that PARG1 plays crucial roles in progression of human RCC in increasing cell proliferation and invasion ability via inhibition of the RhoA-ROCK axis, and PARG1 is a poor prognostic marker, particularly for high recurrence of N0M0 RCC patients.
Collapse
|
14
|
Tumour necrosis factor receptor-associated factor-1 (TRAF-1) expression is increased in renal cell carcinoma patient serum but decreased in cancer tissue compared with normal: potential biomarker significance. Pathology 2016; 46:518-22. [PMID: 25158810 DOI: 10.1097/pat.0000000000000145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Renal cell carcinoma (RCC) generally has a poor prognosis because of late diagnosis and metastasis. We have previously described decreased tumour necrosis factor receptor-associated factor-1 (TRAF-1) in RCC compared with paired normal kidney in a patient cohort in Australia. In the present study, TRAF-1 expression in clear cell RCC (ccRCC) and normal kidney was again compared, but in a cohort from University Malaya Medical Centre. Serum TRAF-1 was also evaluated in RCC and normal samples.Immunohistochemistry with automated batch staining and Aperio ImageScope morphometry was used to compare TRAF-1 in 61 ccRCC with paired normal kidney tissue. Serum from 15 newly diagnosed and untreated ccRCC and 15 healthy people was tested for TRAF-1 using ELISA.In this cohort, TRAF-1 was highly expressed in proximal tubular epithelium of normal kidney, and significantly decreased in ccRCC tissue (p < 0.001). Conversely, TRAF-1 in serum from ccRCC patients was significantly increased over control serum (132 ± 30 versus 54 ± 14 pg/mL, respectively; p = 0.013).Decreased TRAF-1 in RCC tissue, reported previously, was confirmed. This, along with significantly increased serum TRAF-1 may indicate the protein is actively secreted during development and progression of ccRCC. Therefore, the increased serum TRAF-1 may be a useful non-invasive indicator of RCC development.
Collapse
|
15
|
Xu S, Yang Z, Fan Y, Guan B, Jia J, Gao Y, Wang K, Wu K, Wang X, Zheng P, He D, Guo P. Curcumin enhances temsirolimus-induced apoptosis in human renal carcinoma cells through upregulation of YAP/p53. Oncol Lett 2016; 12:4999-5006. [PMID: 28105206 PMCID: PMC5228528 DOI: 10.3892/ol.2016.5376] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/04/2016] [Indexed: 02/07/2023] Open
Abstract
Curcumin has frequently been used as a therapeutic agent in the treatment of various types of disease and is known to enhance the drug sensitivity of cells. In the present study, the combined effect of curcumin and temsirolimus treatment on apoptosis in human renal cell carcinoma (RCC) cells was investigated. Temsirolimus is an inhibitor of the mechanistic target of rapamycin signaling pathway and used in the first-line treatment of metastatic RCC. It was demonstrated that curcumin combined with temsirolimus markedly induced apoptosis in RCC cells, however this effect was not observed following curcumin or temsirolimus treatment alone. Co-treatment with temsirolimus and curcumin led to the activation of cleaved poly ADP-ribose polymerase and caspase 3, upregulation of p53 expression and nuclear translocation, and downregulation of B-cell lymphoma 2 protein expression. Furthermore, curcumin treatment was demonstrated to increase Yes-associated protein (YAP) expression in a time-dependent manner, which was concurrent with the curcumin-induced expression pattern of p53 after 2 h. In addition, knockdown of YAP by small interfering RNA caused the attenuation of curcumin-induced increased p53 expression in RCC cells. In conclusion, the present results indicate that combined curcumin and temsirolimus treatment has a synergistic effect on apoptosis in human RCC cells, through the activation of p53. Mechanistically, YAP is essential in the induction of p53 expression by curcumin. Furthermore, the results suggest that pre-treatment or co-treatment of cells with low concentration curcumin enhances the response to targeted drugs, and this presents a potentially novel and efficient strategy to overcome drug resistance in human RCC.
Collapse
Affiliation(s)
- Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Zheng Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Yizeng Fan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Bing Guan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Jia
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Yang Gao
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Kaijie Wu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Pengsheng Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital, Xi'an Jiaotong University Medical School, Xi'an, Shaanxi 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China; Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
16
|
Prognostic Significance of CD24 in Clear Cell Renal Cell Carcinoma. Pathol Oncol Res 2016; 23:409-416. [DOI: 10.1007/s12253-016-0128-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 10/05/2016] [Indexed: 12/12/2022]
|
17
|
Myszczyszyn A, Czarnecka AM, Matak D, Szymanski L, Lian F, Kornakiewicz A, Bartnik E, Kukwa W, Kieda C, Szczylik C. The Role of Hypoxia and Cancer Stem Cells in Renal Cell Carcinoma Pathogenesis. Stem Cell Rev Rep 2016. [PMID: 26210994 PMCID: PMC4653234 DOI: 10.1007/s12015-015-9611-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cancer stem cell (CSC) model has recently been approached also in renal cell carcinoma (RCC). A few populations of putative renal tumor-initiating cells (TICs) were identified, but they are indifferently understood; however, the first and most thoroughly investigated are CD105-positive CSCs. The article presents a detailed comparison of all renal CSC-like populations identified by now as well as their presumable origin. Hypoxic activation of hypoxia-inducible factors (HIFs) contributes to tumor aggressiveness by multiple molecular pathways, including the governance of immature stem cell-like phenotype and related epithelial-to-mesenchymal transition (EMT)/de-differentiation, and, as a result, poor prognosis. Due to intrinsic von Hippel-Lindau protein (pVHL) loss of function, clear-cell RCC (ccRCC) develops unique pathological intra-cellular pseudo-hypoxic phenotype with a constant HIF activation, regardless of oxygen level. Despite satisfactory evidence concerning pseudo-hypoxia importance in RCC biology, its influence on putative renal CSC-like largely remains unknown. Thus, the article discusses a current knowledge of HIF-1α/2α signaling pathways in the promotion of undifferentiated tumor phenotype in general, including some experimental findings specific for pseudo-hypoxic ccRCC, mostly dependent from HIF-2α oncogenic functions. Existing gaps in understanding both putative renal CSCs and their potential connection with hypoxia need to be filled in order to propose breakthrough strategies for RCC treatment.
Collapse
Affiliation(s)
- Adam Myszczyszyn
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| | - Damian Matak
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Lukasz Szymanski
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Fei Lian
- Emory School of Medicine, Atlanta, GA, USA
| | - Anna Kornakiewicz
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.,Department of General Surgery and Transplantology, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Wojciech Kukwa
- Department of Otolaryngology, Czerniakowski Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Claudine Kieda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Orléans, France
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| |
Collapse
|
18
|
Zhou D, Kannappan V, Chen X, Li J, Leng X, Zhang J, Xuan S. RBP2 induces stem-like cancer cells by promoting EMT and is a prognostic marker for renal cell carcinoma. Exp Mol Med 2016; 48:e238. [PMID: 27282106 PMCID: PMC4929691 DOI: 10.1038/emm.2016.37] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/30/2015] [Accepted: 01/06/2016] [Indexed: 12/13/2022] Open
Abstract
Renal cell carcinoma (RCC), one of the most common kidney cancers, has a poor prognosis. Epithelial to mesenchymal transition (EMT) is a hallmark of carcinoma invasion and metastasis. Several studies have examined the molecular regulation of EMT, but the relationship between histone demethylases and EMT is little understood. In this study, we investigated the role of retinoblastoma-binding protein-2 (RBP2), a histone demethylase that is highly expressed in RCC and is positively correlated with poor RCC prognosis in the regulation of EMT. We found that ectopic overexpression of RBP2 can induce cancer stem cell-like (CSC) phenotypes through EMT in RCC cells by converting them to a more mesenchymal phenotype. This results in increased resistance to apoptosis, which leads to enhanced tumor growth in xenograft models. Together, our data show that RBP2 is an epigenetic regulator that has an important role in the initiation of CSC phenotypes through EMT, leading to tumor progression. RBP2 is also a novel biomolecule for RCC diagnosis, and prognosis and may be a therapeutic target.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Carcinogenesis/metabolism
- Carcinogenesis/pathology
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Line, Tumor
- Epithelial-Mesenchymal Transition
- Follow-Up Studies
- Humans
- Kidney/metabolism
- Kidney/pathology
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mice, Inbred BALB C
- Mice, Nude
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Prognosis
- Retinol-Binding Proteins, Cellular/analysis
- Retinol-Binding Proteins, Cellular/metabolism
Collapse
Affiliation(s)
- Dahai Zhou
- College of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Department of Urology, 401 Hospital of PLA, Qingdao, China
| | - Vinodh Kannappan
- Research Institute in Healthcare Science, Faculty of Science & Engineering, University of Wolverhampton, Wolverhampton, UK
| | | | - Jingqin Li
- Department of Urology, 401 Hospital of PLA, Qingdao, China
| | - Xuefeng Leng
- Department of Urology, 401 Hospital of PLA, Qingdao, China
| | - Jinping Zhang
- Department of Urology, 401 Hospital of PLA, Qingdao, China
| | - Shiying Xuan
- College of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
19
|
Renal Cancer Stem Cells: Characterization and Targeted Therapies. Stem Cells Int 2016; 2016:8342625. [PMID: 27293448 PMCID: PMC4884584 DOI: 10.1155/2016/8342625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/24/2016] [Indexed: 02/08/2023] Open
Abstract
Renal cell carcinoma (RCC) is a major neoplasm with high incidence in western countries. Tumors are heterogeneous and are composed of differentiated cancer cells, stromal cells, and cancer stem cells (CSCs). CSCs possess two main properties: self-renewal and proliferation. Additionally, they can generate new tumors once transplanted into immunodeficient mice. Several approaches have been described to identify them, through the expression of cell markers, functional assays, or a combination of both. As CSCs are involved in the resistance mechanisms to radio- and chemotherapies, several new strategies have been proposed to directly target CSCs in RCC. One approach drives CSCs to differentiate into cancer cells sensitive to conventional treatments, while the other proposes to eradicate them selectively. A series of innovative therapies aiming at eliminating CSCs have been designed to treat other types of cancer and have not been experimented with on RCC yet, but they reveal themselves to be promising. In conclusion, CSCs are an important player in carcinogenesis and represent a valid target for therapy in RCC patients.
Collapse
|
20
|
Yuan H, Meng X, Guo W, Cai P, Li W, Li Q, Wang W, Sun Y, Xu Q, Gu Y. Transmembrane-Bound IL-15-Promoted Epithelial-Mesenchymal Transition in Renal Cancer Cells Requires the Src-Dependent Akt/GSK-3β/β-Catenin Pathway. Neoplasia 2016; 17:410-20. [PMID: 26025664 PMCID: PMC4468369 DOI: 10.1016/j.neo.2015.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 03/27/2015] [Accepted: 04/09/2015] [Indexed: 11/25/2022]
Abstract
Intrarenal interleukin-15 (IL-15) plays a major role controlling epithelial survival and polarization both in physiological and pathologic conditions. Herein, we confirmed that human renal cell carcinomas (RCCs) express a membrane-bound IL-15 isoform displaying an unusual molecular weight of 27 kDa. Its stimulation with soluble IL-15 receptor α chain (s-IL-15Rα) triggers epithelial-mesenchymal transition (EMT) process as shown by the down-regulation of E-cadherin and zona occludens 1 and the up-regulation of vimentin and N-cadherin and promotes the migratory and invasive properties of RCC. S-IL-15Rα treatment triggered the Src/PI3K/Akt/GSK-3β pathway and promoted β-catenin nuclei translocation. Deactivation of this pathway by using Src-specific inhibitor PP2, PI3K inhibitor LY294002, and AKT inhibitor MK2206 hampered β-catenin nuclei translocation and suppressed EMT, migration, and invasion of RCC. S-IL-15Rα treatment also enhanced Src-dependent phosphorylation of focal adhesion kinase (FAK) and extracellular signal-regulated kinase (Erk1/2). FAK knockdown significantly decreased the migration and invasion of RCC, which suggest that Src-FAK signaling was involved in s-IL-15Rα-favored migration and invasion of RCC. At the same time, inhibitors of Erk1/2 also significantly decreased the migration and invasion of RCC but could not reverse s-IL-15Rα-induced EMT. Taken together, our results reveal that Src-dependent PI3K/Akt/GSK3b/β-catenin pathway is required for s-IL-15Ra-dependent induction of EMT in RCC, while Src-FAK and Src-Erk1/2 signaling were involved in s-IL-15Rα-promoted migration and invasion properties of RCC. Our study provides a better understanding of IL-15 signaling in RCC tumor progression, which may lead to novel targeted therapies and provide some suggestions when using IL-15 in clinic.
Collapse
Affiliation(s)
- Huaqin Yuan
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xiaoxin Meng
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Peifen Cai
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Wanshuai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qian Li
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Weicheng Wang
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
21
|
Mahalingaiah PKS, Ponnusamy L, Singh KP. Chronic oxidative stress leads to malignant transformation along with acquisition of stem cell characteristics, and epithelial to mesenchymal transition in human renal epithelial cells. J Cell Physiol 2015; 230:1916-28. [PMID: 25546616 DOI: 10.1002/jcp.24922] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/24/2014] [Accepted: 12/18/2014] [Indexed: 12/30/2022]
Abstract
Oxidative injury to cellular macromolecules has been suggested as a common pathway shared by multiple etiological factors for kidney cancer. Whether the chronic oxidative stress alone is sufficient to induce malignant transformation in human kidney cells is not clear. Therefore, the objective of this study was to evaluate the effect of H2O2-induced chronic oxidative stress on growth, and malignant transformation of HK-2 normal kidney epithelial cells. This study revealed that chronic oxidative stress causes increased growth and neoplastic transformation in normal kidney epithelial cells at non-cytotoxic dose and increased adaptation to cytotoxic level. This was confirmed by gene expression changes, cell cycle analysis, anchorage independent growth assay and in vivo tumorigenicity in nude mice. Stem cells characteristics as revealed by up-regulation of stem cell marker genes, and morphological changes indicative of EMT with up regulation of mesenchymal markers were also observed in cells exposed to chronic oxidative stress. Antioxidant NAC did not reverse the chronic oxidative stress-induced growth, and adaptation suggesting that perturbed biological function in these cells are permanent. Partial reversal of oxidative stress-induced growth, and adaptation by silencing of Oct 4 and Snail1, respectively, suggest that these changes are mediated by acquisition of stem cell and EMT characteristics. In summary, this study for the first time suggests that chronic exposure to elevated levels of oxidative stress is sufficient to induce malignant transformation in kidney epithelial cells through acquisition of stem cell characteristics. Additionally, the EMT plays an important role in increased adaptive response of renal cells to oxidative stress.
Collapse
Affiliation(s)
- Prathap Kumar S Mahalingaiah
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH), Texas Tech University, Lubbock, Texas
| | | | | |
Collapse
|
22
|
Lombardi D, Becherucci F, Romagnani P. How much can the tubule regenerate and who does it? An open question. Nephrol Dial Transplant 2015; 31:1243-50. [PMID: 26175143 PMCID: PMC4967725 DOI: 10.1093/ndt/gfv262] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/22/2015] [Indexed: 01/09/2023] Open
Abstract
The tubular compartment of the kidney is the primary site of a wide range of insults that can result in acute kidney injury (AKI), a condition associated with high mortality and an increased risk to develop end-stage renal disease. Nevertheless, kidney function is often quickly recovered after tubular injury. How this happens has only partially been unveiled. Indeed, although it has clearly been demonstrated that regenerated epithelial cells arise from survived intratubular cells, the true entity, as well as the cellular source of this regenerative process, remains mostly unknown. Is whichever proximal tubular epithelial cell able to dedifferentiate and divide to replace neighboring lost tubular cells, thus suggesting an extreme regenerative ability of residual tubular epithelium, or is the regenerative potential of tubular epithelium limited, and mostly related to a preexisting population of intratubular scattered progenitor cells which are more resistant to death? Gaining insights on how this process takes place is essential for developing new therapeutic strategies to prevent AKI, as well as AKI-related chronic kidney disease. The aim of this review is to discuss why the answers to these questions are still open, and how further investigations are needed to understand which is the true regenerative potential of the tubule and who are the players that allow functional recovery after AKI.
Collapse
Affiliation(s)
- Duccio Lombardi
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
| | - Francesca Becherucci
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
23
|
Garattini E, Tavani A. Is �Bad Luck' an Important Determinant of Cancer Incidence and Does This Concept Apply to Kidney Tumors? Nephron Clin Pract 2015; 129:219-22. [DOI: 10.1159/000381196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/09/2015] [Indexed: 11/19/2022] Open
|
24
|
Kornakiewicz A, Solarek W, Bielecka ZF, Lian F, Szczylik C, Czarnecka AM. Mammalian Target of Rapamycin Inhibitors Resistance Mechanisms in Clear Cell Renal Cell Carcinoma. CURRENT SIGNAL TRANSDUCTION THERAPY 2014; 8:210-218. [PMID: 25152703 PMCID: PMC4141323 DOI: 10.2174/1574362409666140206222746] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/21/2014] [Accepted: 01/29/2014] [Indexed: 11/22/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a kinase protein involved in PI3K/AKT signaling with a central role in the processes of cell growth, survival and angiogenesis. Frequent mutations of this pathway make upstream and downstream components novel targets for tailored therapy design. Two mTOR inhibitors - everolimus and temsirolimus - enable an increase in overall survival (OS) or progression-free survival (PFS) time in a treatment of renal cancer. Despite recent advances in renal cancer treatment, resistance to targeted therapy is common. Understanding of molecular mechanisms is the basis of drug resistance which can facilitate prediction of success or failure in combinational or sequential targeted therapy. The article provides current knowledge on the mTOR signaling network and gives insight into the mechanisms of resistance to mTOR inhibitors from the complex perspective of RCC biology. The mechanisms of resistance developed not only by cancer cells, but also by interactions with tumor microenvironment are analyzed to emphasize the role of angiogenesis in ccRCC pathogenesis. As recent studies have shown the role of PI3K/AKT-mTOR pathway in proliferation and differentiation of cancer stem cells, we discuss cancer stem cell hypothesis and its possible contribution to ccRCC resistance. In the context of drug resistance, we also elaborate on a new approach considering ccRCC as a metabolic disease. In conclusion we speculate on future developments in agents targeting the mTOR pathway taking into consideration the singular biology of ccRCC.
Collapse
Affiliation(s)
- Anna Kornakiewicz
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
- I Faculty of Medicine, Medical University of Warsaw,Poland
- Collegium Invisibile, Warsaw,Poland
| | - Wojciech Solarek
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw,Poland
| | - Zofia F. Bielecka
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw,Poland
| | - Fei Lian
- Department of Urology, Emory School of Medicine, Atlanta, GA ,USA
| | - Cezary Szczylik
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
| | - Anna M. Czarnecka
- Oncology Department, Laboratory of Molecular Oncology, Military Institute of Medicine, Warsaw,Poland
| |
Collapse
|
25
|
Report on ISN Forefronts, Florence, Italy, 12-15 September 2013: Stem cells and kidney regeneration. Kidney Int 2014; 86:23-7. [PMID: 24897031 DOI: 10.1038/ki.2014.32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 01/02/2014] [Accepted: 01/09/2014] [Indexed: 02/07/2023]
Abstract
In recent years it has become clear that most organs and tissues, including kidney, contain resident stem/progenitor cells. Stem cells are undifferentiated, long-lived cells that are unique in their ability to produce differentiated daughter cells and to retain their stem cell identity by self-renewal. A primary goal of this meeting was to review the current understanding of kidney stem cells and mechanisms of kidney regeneration in both lower vertebrates and mammals. Presenters covered a broad range of topics including stem cell quiescence, epigenetics, transcriptional control circuits, dedifferentiation, pluripotent stem cells, renal progenitors, and novel imaging approaches in kidney regeneration. By the end of this highly interactive conference it was clear we are entering into very exciting times for regenerative medicine and the kidney.
Collapse
|
26
|
Kusaba T, Humphreys BD. Controversies on the origin of proliferating epithelial cells after kidney injury. Pediatr Nephrol 2014; 29:673-9. [PMID: 24322596 PMCID: PMC3944107 DOI: 10.1007/s00467-013-2669-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/15/2013] [Indexed: 02/07/2023]
Abstract
The kidney possesses the capacity to repair after an acute insult, even one that causes complete organ failure. This regenerative response is characterized by robust proliferation of epithelial cells, principally those located in the proximal tubule. Because defining the origin of these reparative cells has important consequences for stem cell and regenerative approaches to treating kidney injury, this area has been the subject of intense investigation and debate. While progress has been made in narrowing the possible origin of these cells to an intratubular source, there has been no consensus between the possibility of a pre-existing intratubular stem or progenitor cell versus the possibility that fully differentiated epithelial cells re-enter the cell cycle after injury and generate new proximal tubule cells through self-duplication. This review will summarize the evidence on both sides of this active controversy and provide support for the notion that no pre-existing proximal tubule stem cell population exists, but rather all differentiated proximal tubule epithelia have the capacity to proliferate during repair by a mechanism of dedifferentiation and self-duplication.
Collapse
Affiliation(s)
- Tetsuro Kusaba
- Renal Division, Brigham and Women’s Hospital, Boston, MA,Harvard Medical School, Boston, Massachusetts
| | - Benjamin D. Humphreys
- Renal Division, Brigham and Women’s Hospital, Boston, MA,Harvard Medical School, Boston, Massachusetts,Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
27
|
Abstract
Thyroid cancer cells were believed to be generated by multi-step carcinogenesis, in which cancer cells are derived from thyrocytes, via multiple incidences of damage to their genome, especially in oncogenes or anti-oncogenes that accelerate proliferation or foster malignant phenotypes, such as the ability to invade the surrounding tissue or metastasize to distant organs, until a new hypothesis, fetal cell carcinogenesis, was presented. In fetal cell carcinogenesis, thyroid tumor cells are assumed to be derived from three types of fetal thyroid cell which only exist in fetuses or young children, namely, thyroid stem cells (TSCs), thyroblasts and prothyrocytes, by proliferation without differentiation. Genomic alternations, such as RET/PTC and PAX8-PPARγ1 rearrangements and a mutation in the BRAF gene, play an oncogenic role by preventing thyroid fetal cells from differentiating. Fetal cell carcinogenesis effectively explains recent molecular and clinical evidence regarding thyroid cancer, including thyroid cancer initiating cells (TCICs), and it underscores the importance of identifying a stem cells and clarifying the molecular mechanism of organ development in cancer research. It introduces three important concepts, the reverse approach, stem cell crisis and mature and immature cancers. Further, it implies that analysis of a small population of cells in a cancer tissue will be a key technique in establishing future laboratory tests. In the contrary, mass analysis such as gene expression profiling, whole genomic scan, and proteomics analysis may have definite limitations since they can only provide information based on many cells.
Collapse
Affiliation(s)
- Toru Takano
- Department of Laboratory Medicine, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| |
Collapse
|
28
|
Herrera M, Mirotsou M. Stem cells: potential and challenges for kidney repair. Am J Physiol Renal Physiol 2013; 306:F12-23. [PMID: 24197069 DOI: 10.1152/ajprenal.00238.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal damage resulting from acute and chronic kidney injury poses an important problem to public health. Currently, patients with end-stage renal disease rely solely on kidney transplantation or dialysis for survival. Emerging therapies aiming to prevent and reverse kidney damage are thus in urgent need. Although the kidney was initially thought to lack the capacity for self-repair, several studies have indicated that this might not be the case; progenitor and stem cells appear to play important roles in kidney repair under various pathological conditions. In this review, we summarize recent findings on the role of progenitor/stem cells on kidney repair as well as discuss their potential as a therapeutic approach for kidney diseases.
Collapse
Affiliation(s)
- Marcela Herrera
- Division of Cardiology, Genome Research Bldg. II, Rm. 4022, 210 Research Drive, Duke Univ. Medical Center, Durham, NC 27710.
| | | |
Collapse
|
29
|
Romagnani P, Lasagni L, Remuzzi G. Renal progenitors: an evolutionary conserved strategy for kidney regeneration. Nat Rev Nephrol 2013; 9:137-46. [PMID: 23338209 DOI: 10.1038/nrneph.2012.290] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Following kidney injury, repair can result in functional tissue becoming a patch of cells and disorganized extracellular matrix--a scar--or it can recapitulate the original tissue architecture through the process of regeneration. Regeneration can potentially occur in all animal species and humans. Indeed, the repair of portions of the existing nephron after tubular damage, a response that has been designated classically as cellular regeneration, is conserved in all animal species from the ancestral phases of evolution. By contrast, another type of regenerative response--nephron neogenesis--has been described in lower branches of the animal kingdom, but does not occur in adult mammals. Converging evidence suggests that a renal progenitor system is present in the adult kidney across different stages of evolution, with renal progenitors having been identified as the main drivers of kidney regenerative responses in fish, insects, rodents and humans. In this Review, we describe similarities and differences between the renal progenitor systems through evolution, and propose explanations for how progressive kidney adaptation to environmental changes both required and permitted neonephrogenesis to be given up and for cellular regeneration to be retained as the main regenerative strategy. Understanding the mechanisms that drive renal progenitor growth and differentiation represent the key step for modulating this potential for therapeutic purposes.
Collapse
Affiliation(s)
- Paola Romagnani
- Pediatric Nephrology Unit, Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, 50139 Florence, Italy.
| | | | | |
Collapse
|