1
|
Zhou H, Li X, Liu D. Inhibition of Renal Cell Carcinoma Growth by 1,3-thiazin-6-one Through Targeting the Inflammatory Reaction. DOKL BIOCHEM BIOPHYS 2025:10.1134/S1607672924601008. [PMID: 39847292 DOI: 10.1134/s1607672924601008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 01/24/2025]
Abstract
The current study examined the underlying mechanism and the effect of 1,3-thiazin-6-one on the growth of renal cancer. The findings showed that 1,3-thiazin-6-one treatment inhibited the growth of xenograft tumors in a dose-dependent manner in mice model of renal cancer. Furthermore, when 1,3-thiazin-6-one was administered in a dose-dependent manner to mice with renal cancer, the expression of the proteins p-PI3K and p-Akt significantly decreased. In mice model of renal cancer, 1,3-thiazin-6-one treatment also inhibited p-mTOR expression. In a model of renal cancer in mice, the 1,3-thiazin-6-one therapy specifically targeted the expression of nuclear factor κB (NF κB) and signal transducer and activator of transcription 3 (STAT3). Renal cancer cells' vitality was significantly (p < 0.05) reduced in a dose-dependent manner upon exposure to 1,3-thiazin-6-one. It also prevents invasiveness of the renal cancer cells in addition to suppression of colony forming potential. In summary, the 1,3-thiazin-6-one blocks the growth of kidney cancer by focusing on the pathways that trigger the inflammatory cascade. Therefore, 1,3-thiazin-6-one might be developed as a significant medicinal agent to cure renal cancer.
Collapse
Affiliation(s)
- Hongmei Zhou
- Nephrology Department, Zhongxian People's Hospital of Chongqing, Zhongxian County, 404300, Chongqing, China
| | - Xin Li
- Nephrology Department, Zhongxian People's Hospital of Chongqing, Zhongxian County, 404300, Chongqing, China
| | - Dongju Liu
- Nephrology Department, Liangping Hospital, Liangping District People's Hospital of Chongqing, 405299, Chongqing, China.
| |
Collapse
|
2
|
Wang Y, Xu X, Fang Y, Yang S, Wang Q, Liu W, Zhang J, Liang D, Zhai W, Qian K. Self-Assembled Hyperbranched Gold Nanoarrays Decode Serum United Urine Metabolic Fingerprints for Kidney Tumor Diagnosis. ACS NANO 2024; 18:2409-2420. [PMID: 38190455 DOI: 10.1021/acsnano.3c10717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Serum united urine metabolic analysis comprehensively reveals the disease status for kidney diseases in particular. Thus, the precise and convenient acquisition of metabolic molecular information from united biofluids is vitally important for clinical disease diagnosis and biomarker discovery. Laser desorption/ionization mass spectrometry (LDI-MS) presents various advantages in metabolic analysis; however, there remain challenges in ionization efficiency and MS signal reproducibility. Herein, we constructed a self-assembled hyperbranched black gold nanoarray (HyBrAuNA) assisted LDI-MS platform to profile serum united urine metabolic fingerprints (S-UMFs) for diagnosis of early stage renal cell carcinoma (RCC). The closely packed HyBrAuNA afforded strong electromagnetic field enhancement and high photothermal conversion efficacy, enabling effective ionization of low abundant metabolites for S-UMF collection. With a uniform nanoarray, the platform presented excellent reproducibility to ensure the accuracy of S-UMFs obtained in seconds. When it was combined with automated machine learning analysis of S-UMFs, early stage RCC patients were discriminated from the healthy controls with an area under the curve (AUC) > 0.99. Furthermore, we screened out a panel of 9 metabolites (4 from serum and 5 from urine) and related pathways toward early stage kidney tumor. In view of its high-throughput, fast analytical speed, and low sample consumption, our platform possesses potential in metabolic profiling of united biofluids for disease diagnosis and pathogenic mechanism exploration.
Collapse
Affiliation(s)
- Yuning Wang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Xiaoyu Xu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Yuzheng Fang
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, People's Republic of China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Qirui Wang
- Health Management Center, Renji Hospital of Medical School of Shanghai Jiao Tong University, Shanghai 200127, People's Republic of China
| | - Wanshan Liu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Juxiang Zhang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Dingyitai Liang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine in Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, People's Republic of China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China
| |
Collapse
|
3
|
Wang TB, Geng M, Jin H, Tang AG, Sun H, Zhou LZ, Chen BH, Shen G, Sun Q. SREBP1 site 1 protease inhibitor PF-429242 suppresses renal cell carcinoma cell growth. Cell Death Dis 2021; 12:717. [PMID: 34285190 PMCID: PMC8292369 DOI: 10.1038/s41419-021-03999-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022]
Abstract
Renal cell carcinoma (RCC) cells have increased lipogenesis and cholesterol synthesis. Sterol regulatory element-binding protein-1 (SREBP1) is cleaved by site 1 protease (S1P) to release the transcriptionally active amino-terminal domain. PF-429242 is a potent and competitive S1P inhibitor. We here tested its activity in RCC cells. In established and primary human RCC cells, PF-429242 potently inhibited cell proliferation, migration, and invasion. The S1P inhibitor provoked apoptosis activation in RCC cells. Furthermore, shRNA-mediated S1P silencing or CRISPR/Cas9-induced S1P knockout led to RCC cell growth inhibition and apoptosis activation. Conversely, ectopic overexpression of SREBP1 or S1P augmented RCC cell proliferation and migration. Daily i.v. injection of a single dose of PF-429242 robustly inhibited RCC xenograft growth in severe combined immunodeficiency mice. Additionally, intratumoral injection of S1P shRNA lentivirus inhibited RCC xenograft growth in mice. SREBP1, S1P, and its target gene low density lipoprotein receptor (LDLR) were significantly elevated in human RCC tissues. These results suggest that targeting S1P by PF-429242 inhibited RCC cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Tong-Bing Wang
- Department of Urology, People's Hospital of Yangzhong City, Yangzhong, China
| | - Mei Geng
- Department of Oncology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Jin
- The Child Health Care Department, Suzhou Ninth People's Hospital, Suzhou, China
| | - Ai-Guo Tang
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hao Sun
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Liu-Zheng Zhou
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Bin-Hai Chen
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China.
| | - Qiang Sun
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
4
|
Senturk A, Sahin AT, Armutlu A, Kiremit MC, Acar O, Erdem S, Bagbudar S, Esen T, Tuncbag N, Ozlu N. Quantitative Proteomics Identifies Secreted Diagnostic Biomarkers as well as Tumor-Dependent Prognostic Targets for Clear Cell Renal Cell Carcinoma. Mol Cancer Res 2021; 19:1322-1337. [PMID: 33975903 DOI: 10.1158/1541-7786.mcr-21-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/12/2021] [Accepted: 04/30/2021] [Indexed: 11/16/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third most common and most malignant urological cancer, with a 5-year survival rate of 10% for patients with advanced tumors. Here, we identified 10,160 unique proteins by in-depth quantitative proteomics, of which 955 proteins were significantly regulated between tumor and normal adjacent tissues. We verified four putatively secreted biomarker candidates, namely, PLOD2, FERMT3, SPARC, and SIRPα, as highly expressed proteins that are not affected by intratumor and intertumor heterogeneity. Moreover, SPARC displayed a significant increase in urine samples of patients with ccRCC, making it a promising marker for the detection of the disease in body fluids. Furthermore, based on molecular expression profiles, we propose a biomarker panel for the robust classification of ccRCC tumors into two main clusters, which significantly differed in patient outcome with an almost three times higher risk of death for cluster 1 tumors compared with cluster 2 tumors. Moreover, among the most significant clustering proteins, 13 were targets of repurposed inhibitory FDA-approved drugs. Our rigorous proteomics approach identified promising diagnostic and tumor-discriminative biomarker candidates which can serve as therapeutic targets for the treatment of ccRCC. IMPLICATIONS: Our in-depth quantitative proteomics analysis of ccRCC tissues identifies the putatively secreted protein SPARC as a promising urine biomarker and reveals two molecular tumor phenotypes.
Collapse
Affiliation(s)
- Aydanur Senturk
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Ayse T Sahin
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Ayse Armutlu
- Department of Pathology, Koc University School of Medicine, Istanbul, Turkey
| | - Murat C Kiremit
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Omer Acar
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Selcuk Erdem
- Department of Urology, Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Sidar Bagbudar
- Department of Pathology, Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Tarik Esen
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey.,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey. .,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| |
Collapse
|
5
|
Zheng B, Sun X, Chen XF, Chen Z, Zhu WL, Zhu H, Gu DH. Dual inhibition of DNA-PKcs and mTOR by CC-115 potently inhibits human renal cell carcinoma cell growth. Aging (Albany NY) 2020; 12:20445-20456. [PMID: 33109772 PMCID: PMC7655216 DOI: 10.18632/aging.103847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/20/2020] [Indexed: 01/06/2023]
Abstract
CC-115 is a dual inhibitor of DNA-PKcs and mTOR, both are valuable therapeutic targets for renal cell carcinoma (RCC). Our results showed that CC-115 inhibited survival and proliferation of established RCC cell lines (786-O and A489) and primary human RCC cells. The dual inhibitor induced selective apoptosis activation in RCC cells, as compared to no cytotoxicity nor apoptotic effects toward normal renal epithelial cells. CC-115 inhibited DNA-PKcs and mTORC1/2 activation in RCC cells. It was however ineffective in DNA-PKcs-mTOR double knockout (DKO) 786-O cells. CC-115 induced feedback autophagy activation in RCC cells. Autophagy inhibitors or Beclin-1/Light chain 3 (LC3) silencing potentiated CC-115-induced anti-RCC cell activity. Conversely, ectopic overexpression of Beclin-1 inhibited CC-115-induced cytotoxicity. At last CC-115 oral administration inhibited 786-O subcutaneous xenograft growth in nude mice. Taken together, dual inhibition of DNA-PKcs and mTOR by CC-115 potently inhibited RCC cell growth.
Collapse
Affiliation(s)
- Bing Zheng
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xu Sun
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People’s Hospital, Taizhou, China
| | - Xin-Feng Chen
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Zhan Chen
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Wei-Li Zhu
- Port Clinic, Changshu Customs, Changshu, China
| | - Hua Zhu
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Dong-Hua Gu
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
6
|
BRD4 inhibition sensitizes renal cell carcinoma cells to the PI3K/mTOR dual inhibitor VS-5584. Aging (Albany NY) 2020; 12:19147-19158. [PMID: 33051401 PMCID: PMC7732329 DOI: 10.18632/aging.103723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023]
Abstract
Activation of the PI3K/AKT/mTOR pathway promotes the progression of renal cell carcinoma (RCC). This study tested the anti-RCC cell activity of the PI3K/mTOR dual inhibitor, VS-5584. We show that VS-5584 inhibited PI3K/AKT/mTORC1/2 activation in established (786-O and A498 lines) and primary RCC cells, thereby suppressing cell survival, proliferation, migration and cell cycle progression. VS-5584 induced significant apoptosis in RCC cells. A daily single oral dose of VS-5584 (20 mg/kg) significantly inhibited 786-O tumor growth in vivo. VS-5584 treatment of 786-O tumor xenografts and RCC cells resulted in feedback upregulation of bromodomain-containing protein 4 (BRD4). Furthermore, BRD4 inhibition (by JQ1 and CPI203), knockdown or complete knockout potentiated VS-5584-induced RCC cell death and apoptosis. Conversely, forced overexpression of BRD4 attenuated the cytotoxicity of VS-5584 in 786-O cells. Collectively, VS-5584 potently inhibits RCC cell proliferation and survival. Its anti-tumor activity is further enhanced by the targeted inhibition of BRD4.
Collapse
|
7
|
Ye X, Ruan JW, Huang H, Huang WP, Zhang Y, Zhang F. PI3K-Akt-mTOR inhibition by GNE-477 inhibits renal cell carcinoma cell growth in vitro and in vivo. Aging (Albany NY) 2020; 12:9489-9499. [PMID: 32421688 PMCID: PMC7288912 DOI: 10.18632/aging.103221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/31/2020] [Indexed: 02/05/2023]
Abstract
Sustained activation of PI3K-Akt-mTOR cascade is important for renal cell carcinoma (RCC) cell progression. GNE-477 is a novel and efficacious PI3K-mTOR dual inhibitor. The current study tested its anti-RCC cell activity. In the primary cultured human RCC cells, GNE-477 potently inhibited cell growth, viability and proliferation, as well as cell cycle progression, migration and invasion. Furthermore, it induced robust apoptosis activation in primary RCC cells, but being non-cytotoxic to HK-2 epithelial cells and primary human renal epithelial cells. In the primary RCC cells GNE-477 inactivated PI3K-Akt-mTOR cascade by blocking phosphorylation of p85, Akt1, p70S6K1 and S6. Restoring Akt-mTOR activation by a constitutively-active Akt1 reversed GNE-477-induced anti-RCC cell activity. In nude mice intraperitoneal injection of GNE-477 potently suppressed RCC xenograft tumor growth. Collectively, targeting PI3K-Akt-mTOR cascade by GNE-477 inhibits RCC cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Xueting Ye
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian-Wei Ruan
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, China
| | - Hang Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei-Ping Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Zhang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Fangyi Zhang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
M A S, Delma CR, Manickam K, Mohan S, Habib SL, Natarajan M. Heterozygous Tsc2 (Tsc2+/–) mouse model to study induced renal cancer in response to ionizing radiation at low doses. Carcinogenesis 2019; 40:782-790. [DOI: 10.1093/carcin/bgy172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Abstract
Kidneys are one of the main dose-limiting organs in radiotherapeutic procedures of lower abdomen. Likewise, the threat of exposure of radiosensitive organs such as kidneys in warfare or radiation accidents among military personal or due to terrorist activities in general public is of increasing concern. These events warrant the need for appropriate animal models to study the acute and chronic effects of low- and high-dose rate radiation exposures. In this study, for the first time, we validated Tsc2+/– mouse model to study whether radiation accelerates carcinogenesis in kidneys. Tsc2+/– mice at increasing age groups at 8 and 10 months were exposed to repeated doses of gamma radiation (0.4 Gy × 5) and assessed for aggravated kidney tumor formation at 2 months post-irradiation. Animals from irradiated group showed a significant increase in numbers of bilateral, multifocal tumors compared with mock-irradiated animals. Intra-glomerular reactive oxygen species (ROS) levels measured by dihydroethidium florescence showed significant increases in ROS production in irradiated Tsc2+/– mice compared with non-irradiated animals. Similarly, selective hematological parameters and glomerular filtration rate were further reduced significantly in irradiated Tsc2+/– mice. Tsc2 protein, tuberin in irradiated mice, however, remains at the same reduced levels as that of the mock-irradiated heterozygous Tsc2 mice. The results indicate that radiation alters kidney homeostatic function and influences high spontaneous incidence of renal cell carcinoma in this rodent model. Repurposing of Tsc2+/– mice model will, therefore, provide a unique opportunity to study acute and delayed effects of radiation in the development of kidney cancers.
Collapse
Affiliation(s)
- Sureshkumar M A
- Department of Pathology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Caroline R Delma
- Department of Pathology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Krishnan Manickam
- Department of Pathology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Sumathy Mohan
- Department of Pathology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Samy L Habib
- Department of Cell System and Anatomy, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - Mohan Natarajan
- Department of Pathology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, USA
| |
Collapse
|
9
|
Ye XT, Huang H, Huang WP, Hu WL. LncRNA THOR promotes human renal cell carcinoma cell growth. Biochem Biophys Res Commun 2019; 501:661-667. [PMID: 29752937 DOI: 10.1016/j.bbrc.2018.05.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recent studies have characterized a novel but extremely conserved long non-coding RNA (LncRNA) THOR. THOR directly associates with insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) to promote mRNA stabilization of key pro-cancerous genes. RESULTS Here, we show that THOR is expressed in human renal cell carcinoma (RCC) tissues and established/primary human RCC cells. It was not detected in normal renal tissues nor in HK-2 and primary human renal epithelial cells. THOR silencing (by targeted siRNAs) or CRISPR/Cas9 knockout inhibited RCC cell growth, viability and proliferation in vitro. Reversely, forced over-expression of THOR promoted RCC cell survival and proliferation. IGF2BP1-regulated genes, including IGF2, GLI1 and Myc, were downregulated by THOR silencing or knockout, but they were upregulated after THOR over-expression. In vivo, THOR-knockout 786-O tumors grew significantly slower than the control tumors in nude mice. CONCLUSION THOR expression promotes RCC cell growth in vitro and in vivo. THOR could be a novel and important therapeutic target for human RCC.
Collapse
Affiliation(s)
- Xue-Ting Ye
- Graduate School, Southern Medical University, Guangzhou, China; Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hang Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei-Ping Huang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei-Lie Hu
- Graduate School, Southern Medical University, Guangzhou, China; Department of Urology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, China.
| |
Collapse
|
10
|
Inhibiting tryptophan metabolism enhances interferon therapy in kidney cancer. Oncotarget 2018; 7:66540-66557. [PMID: 27572319 PMCID: PMC5341819 DOI: 10.18632/oncotarget.11658] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/01/2016] [Indexed: 12/28/2022] Open
Abstract
Renal cell carcinoma (RCC) is increasing in incidence, and a complete cure remains elusive. While immune-checkpoint antibodies are promising, interferon-based immunotherapy has been disappointing. Tryptophan metabolism, which produces immunosuppressive metabolites, is enhanced in RCC. Here we show indolamine-2,3-dioxygenase-1 (IDO1) expression, a kynurenine pathway enzyme, is increased not only in tumor cells but also in the microenvironment of human RCC compared to normal kidney tissues. Neither kynurenine metabolites nor IDO inhibitors affected the survival or proliferation of human RCC or murine renal cell adenocarcinoma (RENCA) cells in vitro. However, interferon-gamma (IFNγ) induced high levels of IDO1 in both RCC and RENCA cells, concomitant with enhanced kynurenine levels in conditioned media. Induction of IDO1 by IFNα was weaker than by IFNγ. Neither the IDO1 inhibitor methyl-thiohydantoin-DL-tryptophan (MTH-trp) nor IFNα alone inhibited RENCA tumor growth, however the combination of MTH-trp and IFNα reduced tumor growth compared to IFNα. Thus, the failure of IFNα therapy for human RCC is likely due to its inability to overcome the immunosuppressive environment created by increased IDO1. Based on our data, and given that IDO inhibitors are already in clinical trials for other malignancies, IFNα therapy with an IDO inhibitor should be revisited for RCC.
Collapse
|
11
|
Abu Aboud O, Chen CH, Senapedis W, Baloglu E, Argueta C, Weiss RH. Dual and Specific Inhibition of NAMPT and PAK4 By KPT-9274 Decreases Kidney Cancer Growth. Mol Cancer Ther 2016; 15:2119-29. [PMID: 27390344 PMCID: PMC5010932 DOI: 10.1158/1535-7163.mct-16-0197] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/27/2016] [Indexed: 11/16/2022]
Abstract
Kidney cancer (or renal cell carcinoma, RCC) is the sixth most common malignancy in the United States and one of the relatively few whose incidence is increasing. Because of the near universal resistance which occurs with the use of current treatment regimens, reprogrammed metabolic pathways are being investigated as potential targets for novel therapies of this disease. Borrowing from studies on other malignancies, we have identified the PAK4 and NAD biosynthetic pathways as being essential for RCC growth. We now show, using the dual PAK4/NAMPT inhibitor KPT-9274, that interference with these signaling pathways results in reduction of G2-M transit as well as induction of apoptosis and decrease in cell invasion and migration in several human RCC cell lines. Mechanistic studies demonstrate that inhibition of the PAK4 pathway by KPT-9274 attenuates nuclear β-catenin as well as the Wnt/β-catenin targets cyclin D1 and c-Myc. Furthermore, NAPRT1 downregulation, which we show occurs in all RCC cell lines tested, makes this tumor highly dependent on NAMPT for its NAD requirements, such that inhibition of NAMPT by KPT-9274 leads to decreased survival of these rapidly proliferating cells. When KPT-9274 was administered in vivo to a 786-O (VHL-mut) human RCC xenograft model, there was dose-dependent inhibition of tumor growth with no apparent toxicity; KPT-9274 demonstrated the expected on-target effects in this mouse model. KPT-9274 is being evaluated in a phase I human clinical trial in solid tumors and lymphomas, which will allow this data to be rapidly translated into the clinic for the treatment of RCC. Mol Cancer Ther; 15(9); 2119-29. ©2016 AACR.
Collapse
Affiliation(s)
- Omran Abu Aboud
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California
| | - Ching-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California
| | | | | | | | - Robert H Weiss
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California. Cancer Center, University of California, Davis, Davis, California. Medical Service, Sacramento VA Medical Center, Sacramento, California.
| |
Collapse
|
12
|
Hu SL, Chang A, Perazella MA, Okusa MD, Jaimes EA, Weiss RH. The Nephrologist's Tumor: Basic Biology and Management of Renal Cell Carcinoma. J Am Soc Nephrol 2016; 27:2227-37. [PMID: 26961346 PMCID: PMC4978061 DOI: 10.1681/asn.2015121335] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Kidney cancer, or renal cell carcinoma (RCC), is a disease of increasing incidence that is commonly seen in the general practice of nephrology. However, RCC is under-recognized by the nephrology community, such that its presence in curricula and research by this group is lacking. In the most common form of RCC, clear cell renal cell carcinoma (ccRCC), inactivation of the von Hippel-Lindau tumor suppressor is nearly universal; thus, the biology of ccRCC is characterized by activation of hypoxia-relevant pathways that lead to the associated paraneoplastic syndromes. Therefore, RCC is labeled the internist's tumor. In light of this characterization and multiple other metabolic abnormalities recently associated with ccRCC, it can now be viewed as a metabolic disease. In this review, we discuss the basic biology, pathology, and approaches for treatment of RCC. It is important to distinguish between kidney confinement and distant spread of RCC, because this difference affects diagnostic and therapeutic approaches and patient survival, and it is important to recognize the key interplay between RCC, RCC therapy, and CKD. Better understanding of all aspects of this disease will lead to optimal patient care and more recognition of an increasingly prevalent nephrologic disease, which we now appropriately label the nephrologist's tumor.
Collapse
Affiliation(s)
- Susie L Hu
- Division of Kidney Disease and Hypertension, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Anthony Chang
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Mark A Perazella
- Division of Nephrology, Yale University, New Haven, Connecticut; Medical Service Veterans Affairs Connecticut, West Haven, Connecticut
| | - Mark D Okusa
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Edgar A Jaimes
- Renal Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Renal Division, Weill-Cornell Medical College, New York, New York
| | - Robert H Weiss
- Division of Nephrology and Cancer Center, University of California, Davis, California; and Medical Service, Veterans Affairs Northern California Health Care System, Sacramento, California
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW This review summarizes recent metabolomics studies of renal disease, outlining some of the limitations of the literature to date. RECENT FINDINGS The application of metabolomics in nephrology research has expanded from the initial analyses of uremia to include both cross-sectional and longitudinal studies of earlier stages of kidney disease. Although these studies have nominated several potential markers of incident chronic kidney disease (CKD) and CKD progression, a lack of overlap in metabolite coverage has limited the ability to synthesize results across groups. Furthermore, direct examination of renal metabolite handling has underscored the substantial impact kidney function has on these potential markers (and many other circulating metabolites). In experimental studies, metabolomics has been used to identify a signature of decreased mitochondrial function in diabetic nephropathy and a preference for aerobic glucose metabolism in polycystic kidney disease. In each case, these studies have outlined novel therapeutic opportunities. Finally, as a complement to the longstanding interest in renal metabolite clearance, the microbiome has been increasingly recognized as the source of many plasma metabolites, including some with potential functional relevance to CKD and its complications. SUMMARY The high-throughput, high-resolution phenotyping enabled by metabolomics technologies has begun to provide insight on renal disease in clinical, physiologic, and experimental contexts.
Collapse
|
14
|
Kosti A, Harry Chen HI, Mohan S, Liang S, Chen Y, Habib SL. Microarray profile of human kidney from diabetes, renal cell carcinoma and renal cell carcinoma with diabetes. Genes Cancer 2015; 6:62-70. [PMID: 25821562 PMCID: PMC4362485 DOI: 10.18632/genesandcancer.51] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/13/2015] [Indexed: 11/25/2022] Open
Abstract
Recent study from our laboratory showed that patients with diabetes are at a higher risk of developing kidney cancer. In the current study, we have screened whole human DNA genome from healthy control, patients with diabetes or renal cell carcinoma (RCC) or RCC+diabetes. We found that 883 genes gain/163 genes loss of copy number in RCC+diabetes group, 669 genes gain/307 genes loss in RCC group and 458 genes gain/38 genes loss of copy number in diabetes group, after removing gain/loss genes obtained from healthy control group. Data analyzed for functional annotation enrichment pathways showed that control group had the highest number (280) of enriched pathways, 191 in diabetes+RCC group, 148 in RCC group, and 81 in diabetes group. The overlap GO pathways between RCC+diabetes and RCC groups showed that nine were enriched, between RCC+diabetes and diabetes groups was four and between diabetes and RCC groups was eight GO pathways. Overall, we observed majority of DNA alterations in patients from RCC+diabetes group. Interestingly, insulin receptor (INSR) is highly expressed and had gains in copy number in RCC+diabetes and diabetes groups. The changes in INSR copy number may use as a biomarker for predicting RCC development in diabetic patients.
Collapse
Affiliation(s)
- Adam Kosti
- Department of Cellular & Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Hung-I Harry Chen
- Department of Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas
| | - Sumathy Mohan
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Sitai Liang
- Department of Cellular & Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| | - Yidong Chen
- Department of Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas ; Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio, Texas
| | - Samy L Habib
- Geriatric Research, Education and Clinical Center, South Texas Veterans Healthcare System, University of Texas Health Science Center, San Antonio, Texas ; Department of Cellular & Structural Biology, University of Texas Health Science Center, San Antonio, Texas
| |
Collapse
|
15
|
Wettersten HI, Landesman Y, Friedlander S, Shacham S, Kauffman M, Weiss RH. Specific inhibition of the nuclear exporter exportin-1 attenuates kidney cancer growth. PLoS One 2014; 9:e113867. [PMID: 25461627 PMCID: PMC4252068 DOI: 10.1371/journal.pone.0113867] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 10/31/2014] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Despite the advent of FDA-approved therapeutics to a limited number of available targets (kinases and mTOR), PFS of kidney cancer (RCC) has been extended only one to two years due to the development of drug resistance. Here, we evaluate a novel therapeutic for RCC which targets the exportin-1 (XPO1) inhibitor. MATERIALS AND METHODS RCC cells were treated with the orally available XPO1 inhibitor, KPT-330, and cell viability and Annexin V (apoptosis) assays, and cell cycle analyses were performed to evaluate the efficacy of KPT-330 in two RCC cell lines. Immunoblotting and immunofluorescence analysis were performed to validate mechanisms of XPO1 inhibition. The efficacy and on-target effects of KPT-330 were further analyzed in vivo in RCC xenograft mice, and KPT-330-resistant cells were established to evaluate potential mechanisms of KPT-330 resistance. RESULTS KPT-330 attenuated RCC viability through growth inhibition and apoptosis induction both in vitro and in vivo, a process in which increased nuclear localization of p21 by XPO1 inhibition played a major role. In addition, KPT-330 resistant cells remained sensitive to the currently approved for RCC multi-kinase inhibitors (sunitinib, sorafenib) and mTOR inhibitors (everolimus, temsirolimus), suggesting that these targeted therapeutics would remain useful as second line therapeutics following KPT-330 treatment. CONCLUSION The orally-available XPO1 inhibitor, KPT-330, represents a novel target for RCC whose in vivo efficacy approaches that of sunitinib. In addition, cells resistant to KPT-330 retain their ability to respond to available RCC therapeutics suggesting a novel approach for treatment in KPT-330-naïve as well as -resistant RCC patients.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Administration, Oral
- Animals
- Apoptosis/drug effects
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Drug Approval
- Drug Resistance, Neoplasm/drug effects
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Humans
- Hydrazines/administration & dosage
- Hydrazines/pharmacology
- Karyopherins/antagonists & inhibitors
- Karyopherins/metabolism
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- Mice, Nude
- RNA, Small Interfering/metabolism
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
- Triazoles/administration & dosage
- Triazoles/pharmacology
- United States
- United States Food and Drug Administration
- Xenograft Model Antitumor Assays
- Exportin 1 Protein
Collapse
Affiliation(s)
- Hiromi I. Wettersten
- Division of Nephrology, Dept. of Internal Medicine, University of California Davis, Davis, California, United States of America
| | - Yosef Landesman
- Karyopharm Therapeutics Inc., Natick, Massachusetts, United States of America
| | - Sharon Friedlander
- Karyopharm Therapeutics Inc., Natick, Massachusetts, United States of America
| | - Sharon Shacham
- Karyopharm Therapeutics Inc., Natick, Massachusetts, United States of America
| | - Michael Kauffman
- Karyopharm Therapeutics Inc., Natick, Massachusetts, United States of America
| | - Robert H. Weiss
- Division of Nephrology, Dept. of Internal Medicine, University of California Davis, Davis, California, United States of America
- Cancer Center, University of California Davis, Davis, California, United States of America
- Medical Service, Sacramento VA Medical Center, Sacramento, California, United States of America
| |
Collapse
|
16
|
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
|
17
|
Dias F, Teixeira AL, Santos JI, Gomes M, Nogueira A, Assis J, Medeiros R. Renal cell carcinoma development and miRNAs: a possible link to the EGFR pathway. Pharmacogenomics 2014; 14:1793-803. [PMID: 24192126 DOI: 10.2217/pgs.13.184] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most common solid cancer of the adult kidney and the majority of RCC cases are detected accidentally. This reality and the nonexistence of a standard screening test contribute to the fact that one third of patients are diagnosed with local invasive disease or metastatic disease. miRNAs are a family of small ncRNAs that regulate gene expression and have been identified as key regulators in many biological processes including cell development, differentiation, apoptosis and proliferation. The EGF receptor signaling pathway is usually deregulated in cancer and it is suggested to have an important role in RCC. Further studies are needed to characterize deregulation of this pathway during RCC development. In this review we highlight some potential miRNAs that could be involved in the modulation of the EGF receptor pathway and consequently in RCC development.
Collapse
Affiliation(s)
- Francisca Dias
- Molecular Oncology Group, Portuguese Institute of Oncology, Rua Dr António Bernardino de Almeida, 4200-072 Porto, Portugal
| | | | | | | | | | | | | |
Collapse
|
18
|
Jones EE, Powers TW, Neely BA, Cazares LH, Troyer DA, Parker AS, Drake RR. MALDI imaging mass spectrometry profiling of proteins and lipids in clear cell renal cell carcinoma. Proteomics 2014; 14:924-35. [PMID: 24497498 DOI: 10.1002/pmic.201300434] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 12/05/2013] [Accepted: 12/21/2013] [Indexed: 01/08/2023]
Abstract
Reducing the incidence and mortality rates for clear cell renal cell carcinoma (ccRCC) remains a significant clinical challenge with poor 5-year survival rates. A unique tissue cohort was assembled of matched ccRCC and distal nontumor tissues (n = 20) associated with moderate risk of disease progression, half of these from individuals who progressed to metastatic disease and the other half who remained disease free. These tissues were used for MALDI imaging MS profiling of proteins in the 2-20 kDa range, resulting in a panel of 108 proteins that had potential disease-specific expression patterns. Protein lysates from the same tissues were analyzed by MS/MS, resulting in identification of 56 proteins of less than 20 kDa molecular weight. The same tissues were also used for global lipid profiling analysis by MALDI-FT-ICR MS. From the cumulative protein and lipid expression profile data, a refined panel of 26 proteins and 39 lipid species was identified that could either distinguish tumor from nontumor tissues, or tissues from recurrent disease progressors from nonrecurrent disease individuals. This approach has the potential to not only improve prognostic assessment and enhance postoperative surveillance, but also to inform on the underlying biology of ccRCC progression.
Collapse
Affiliation(s)
- Elizabeth Ellen Jones
- Department of Cell and Molecular Pharmacology, MUSC Proteomics Center, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Afreen S, Dermime S. The immunoinhibitory B7-H1 molecule as a potential target in cancer: Killing many birds with one stone. Hematol Oncol Stem Cell Ther 2014; 7:1-17. [DOI: 10.1016/j.hemonc.2013.09.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 09/06/2013] [Indexed: 02/06/2023] Open
|