101
|
A Predictive Model Based on Pyroptosis-Related Gene Features Can Effectively Predict Clear Cell Renal Cell Carcinoma Prognosis and May Be an Underlying Target for Immunotherapy. DISEASE MARKERS 2022; 2022:6402599. [PMID: 35845137 PMCID: PMC9286942 DOI: 10.1155/2022/6402599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/06/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Methods The clinical information and RNA-seq data of ccRCC patients were collected from the TCGA dataset to first explore differential pyroptosis-related genes (PRGs). Univariate Cox regression and consensus clustering were applied to identify ccRCC subtypes. The prognostic PRGs were subjected to LASSO regression analysis to establish a prognostic model and to investigate its value and function. Finally, the relationship of the model immunity checkpoints and immunity infiltration was assessed. Results The receiver operating characteristic (ROC) showed that the 1-year, 3-year, and 5-year prediction rates of the prognostic model were 0.715, 0.693, and 0.732, respectively. The high-risk group had lower overall survival and higher stage than the low-risk group. Functional enrichment analysis showed that PRGs were significantly enriched mainly in the PPAR pathway, inflammatory pathway, and immune activity. ccRCC patient prognosis correlates with immune components in the microenvironment, and immune checkpoint molecules are significantly expressed in the high-risk group. Immunotherapy may be effective in the high-risk group. Conclusion Pyroptosis-related gene has an important impact on the progression of ccRCC and can be used as an independent predictor of patient prognosis. In addition, immune checkpoint molecules are significantly upregulated in high-risk populations, which may be a potential target for immunotherapy.
Collapse
|
102
|
Hu F, Ren Y, Wang Z, Zhou H, Luo Y, Wang M, Tian F, Zheng J, Du J, Pang G. Bioinformatics analysis of KLF2 as a potential prognostic factor in ccRCC and association with epithelial‑mesenchymal transition. Exp Ther Med 2022; 24:561. [PMID: 35978925 PMCID: PMC9366276 DOI: 10.3892/etm.2022.11498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a primary pathological subtype of RCC and has poor clinical outcome. Krüppel-like factors (KLFs), which are zinc-finger proteins, may be involved in ccRCC development and progression. KLFs belong to the zinc-finger family of DNA-binding transcription factors and regulate transcription of downstream target genes. KLFs are involved in cancer development. The present study aimed to investigate the role of KLFs in ccRCC prognosis. The Cancer Genome Atlas database and multifactorial analysis showed that KLFs were widely expressed in pan-cancers and KLF2 was an independent protective factor for ccRCC prognosis. Patients with low KLF2 expression had a low survival probability and expression of KLF2 was downregulated in patients with ccRCC with high pathological grade (II + III vs. I). In addition, western blot and reverse transcription-quantitative PCR revealed that KLF2 was expressed at low levels in ccRCC cell lines and overexpression of KLF2 inhibited cell migration. In addition, KLF2 expression was negatively correlated with methylation. KLF2 expression was elevated following treatment of ccRCC cells with DNA methyltransferase inhibitor. A prognostic risk index prediction model was constructed based on multiple Cox regression. The receiver operating characteristic curve was 0.780 (area under curve >0.5). Furthermore, Gene Ontology enrichment analysis showed that ‘cell adhesion’ and ‘junction’ were negatively correlated with KLF2 and that high-risk group exhibited significantly activated ‘epithelial-mesenchymal transition’. Western blot analysis showed that overexpression of KLF2 increased expression of E-cadherin, while decreasing levels of N-cadherin and vimentin. The present study highlighted the role of KLFs in ccRCC prognosis prediction and provides a research base for the search of validated prognostic biological markers for ccRCC.
Collapse
Affiliation(s)
- Fangfang Hu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan Ren
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zunyun Wang
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Hui Zhou
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yumei Luo
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Minghua Wang
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Faqing Tian
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Jian Zheng
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| | - Juan Du
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Gang Pang
- Department of Human Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
103
|
Liu J, Xu J, Zhang T, Xu K, Bao P, Zhang Z, Xue K, He R, Ma L, Wang Y. Decoding the Immune Microenvironment of Clear Cell Renal Cell Carcinoma by Single-Cell Profiling to Aid Immunotherapy. Front Immunol 2022; 13:791158. [PMID: 35812372 PMCID: PMC9263726 DOI: 10.3389/fimmu.2022.791158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 05/23/2022] [Indexed: 01/09/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer, and it is the major cause of kidney cancer death. Understanding tumor immune microenvironments (TMEs) is critical in cancer immunotherapies. Here, we studied the immune characterization at single-cell resolution by integrating public data of ccRCC across different tissue types, and comparing the transcriptome features and tumor TME differences in tumors, normal adjacent tissue, and peripheral blood. A total of 16 different types of cell components of ccRCC were identified. We revealed that there is an overall increase in T-cell and myeloid populations in tumor-infiltrated immune cells compared to normal renal tissue, and the B-cell population in the tumor showed a sharp decrease, which indicates that the cells in tumor tissue undergo strong immune stress. In addition, the cell-cell communication analysis revealed specific or conserved signals in different tissue types, which may aid to uncover the distinct immune response. By combining and analyzing publicly available ccRCC bulk RNA-seq datasets, 10 genes were identified as marker genes in specific cell types, which were significantly associated with poor prognosis. Of note, UBE2C, which may be a good indicator of tumor proliferation, is positively associated with reductions in overall survival and highly associated with tumor grade. Our integrated analysis provides single-cell transcriptomic profiling of ccRCC and their TME, and it unmasked new correlations between gene expression, survival outcomes, and immune cell-type components, enabling us to dissect the dynamic variables in the tumor development process. This resource provides deeper insight into the transcriptome features and immune response of ccRCC and will be helpful in kidney cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Jiangfan Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Tong Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Kailong Xu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Peihua Bao
- College of Intelligent Systems Science and Engineering, Harbin Engineering University, Harbin, China
| | - Zhibo Zhang
- Department of Cardiothoracic Surgery, The 78th Group Army Hospital of Chinese People's Liberation Army, Mudanjiang, China
| | - Kaiwen Xue
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan, China
| | - Ruyi He
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Lixin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China,*Correspondence: Yang Wang, ; Lixin Ma,
| | - Yang Wang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China,*Correspondence: Yang Wang, ; Lixin Ma,
| |
Collapse
|
104
|
Zhu Z, Lei Z, Qian J, Zhang C, Gong Y, Yin G, Li Y, Li X, Lin J, Zhou L. The Ion Channel-Related Gene Signatures Correlated With Diagnosis, Prognosis, and Individualized Treatment in Patients With Clear Cell Renal Cell Carcinoma. Front Pharmacol 2022; 13:889142. [PMID: 35721115 PMCID: PMC9198310 DOI: 10.3389/fphar.2022.889142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
Background: Early detection and precise prognostic evaluation of clear cell renal cell carcinoma (ccRCC) are crucial for patient life expectancy. Ion channel-related genes (ICRGs) are of great diagnostic and prognostic value as components that maintain the normal structure of the kidney. Therefore, we systematically explored the diagnostic, prognostic, and therapeutic value of ICRGs in ccRCC using the multi-database. Methods: RNA transcriptome profiles and clinical data of ccRCC patients were extracted and integrated from public databases including The Cancer Genome Atlas, ICGC, GEO, and E-MTAB databases. Ion channel-related genes were obtained from the literature collection. The diagnostic signature was performed using the LASSO and SVM-REF analyses. Meanwhile, the prognostic signature was conducted using the LASSO analyses. Molecular subtyping was performed using the ConsensusClusterPlus and the corresponding therapeutic targets were evaluated using the pRRophetic package. In addition, a prognostic nomogram was constructed based on the results of cox regression analyses. Results: We successfully constructed diagnostic signatures for five ICRGs and prognostic signatures for 10 ICRGs with AUC values greater than 0.7, showing good predictive performance. Based on the median risk score, we found that high-risk patients had a significantly worse prognosis. We also divided ccRCC patients into two clusters according to prognostic ICRGs, and there was a significant survival outcome between the two clusters and different sensitivity to diverse clinical therapeutic strategies. Meanwhile, we constructed a nomogram based on clinical molecules and signatures, and its predictive efficacy was better than the signature or the present tumor-node-metastasis staging system. Conclusion: In this study, we established useful signatures for early detection, prognosis evaluation, and individualized treatment for ccRCC. Moreover, KCNJ16 deserves to be explored comprehensively in the future.
Collapse
Affiliation(s)
- Zhenpeng Zhu
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhenchuan Lei
- School of Biomedical Sciences, Heart and Vascular Institute and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jinqin Qian
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Cuijian Zhang
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yanqing Gong
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Guicao Yin
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yifan Li
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xuesong Li
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jian Lin
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Liqun Zhou
- National Urological Cancer Center, Department of Urology, Institute of Urology, Clinical Research Cooperation Network of Urology of the Peking University First Hospital, The Peking University First Hospital, Peking University, Beijing and the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| |
Collapse
|
105
|
Qi X, Che X, Li Q, Wang Q, Wu G. Potential Application of Pyroptosis in Kidney Renal Clear Cell Carcinoma Immunotherapy and Targeted Therapy. Front Pharmacol 2022; 13:918647. [PMID: 35795559 PMCID: PMC9252305 DOI: 10.3389/fphar.2022.918647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/30/2022] [Indexed: 12/21/2022] Open
Abstract
Renal cell carcinoma (RCC) is a type of cancer with an increasing rate of morbidity and mortality and is a serious threat to human health. The treatment of RCC, especially kidney renal clear cell carcinoma (KIRC), has always been the focus of clinical treatment. Using The Cancer Genome Atlas (TCGA) database as a starting point, we explored the feasibility of applying the pyroptosis mechanism to KIRC treatment by searching for cancer markers associated with pyroptosis and cancer treatment signatures. The obtained samples were clustered using unsupervised clustering analysis to define the different KIRC subtypes with different pyroptosis expression levels. Based on this, a gene expression analysis was performed to explore the carcinogenic mechanism that is markedly related to pyroptosis. The Genomics of Drug Sensitivity in Cancer database and single sample gene set enrichment analysis (ssGSEA) algorithm were used to analyze the different treatment methods of the current prominent KIRC to determine whether pyroptosis plays a role. Finally, LASSO regression was used to screen for related genes and construct a model to predict patient prognosis. The expression levels of GSDME, CASP3, CASP4, CASP5, CHMP3, and CHMP4C were incorporated into the model construction. After verification, the prediction accuracy of the 3-, 5-, 7- and 10 years survival rates of our prognostic model were 0.66, 0.701, 0.719, and 0.728, respectively. Through the above analysis, we demonstrated the feasibility of pyroptosis in the clinical treatment of KIRC and provided novel ideas and suggestions for the clinical treatment of KIRC.
Collapse
Affiliation(s)
| | | | | | - Qifei Wang
- *Correspondence: Guangzhen Wu, ; Qifei Wang,
| | | |
Collapse
|
106
|
Liu C, Zhang X, Hu C, Liang X, Cao X, Wang D. Systematic Construction and Validation of a Novel Macrophage Differentiation–Associated Prognostic Model for Clear Cell Renal Cell Carcinoma. Front Genet 2022; 13:877656. [PMID: 35774505 PMCID: PMC9237391 DOI: 10.3389/fgene.2022.877656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/09/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is a malignant tumor of the human urinary system. Macrophage differentiation is associated with tumorigenesis. Therefore, exploring the prognostic value of macrophage differentiation–associated genes (MDGs) may contribute to better clinical management of ccRCC patients.Methods: The RNA sequence data of ccRCC were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed MDGs were unveiled in ccRCC and normal samples. The prognostic model was established according to the univariate and multivariate Cox regression analyses. By combining clinico-pathological features and prognostic genes, a nomogram was established to predict individual survival probability. The Tumor Immune Estimation Resource (TIMER) database was utilized to analyze the correlation between prognostic genes and immune infiltrating cells. Eventually, the mRNA and protein expression levels of prognostic genes were verified.Results: A total of 52 differentially expressed prognosis-related MDGs were identified in ccRCC. Afterward, a six-gene prognostic model (ABCG1, KDF1, KITLG, TGFA, HAVCR2, and CD14) was constructed through the Cox analysis. The overall survival in the high-risk group was relatively poor. Moreover, the risk score was identified as an independent prognostic factor. We constructed a prognostic nomogram with a well-fitted calibration curve based on risk score and clinical data. Furthermore, the prognostic genes were significantly related to the level of immune cell infiltration including B cells, CD8+T cells, CD4+T cells, macrophages, neutrophils, and dendritic cells. Finally, the mRNA expression of prognostic genes in clinical ccRCC tissues showed that the ABCG1, HAVCR2, CD14, and TGFA mRNA in tumor samples were increased compared with the adjacent control tissue samples, while KDF1 and KITLG were decreased, which was consistent with the verification results in the GSE53757.Conclusion: In conclusion, this study identified and validated a macrophage differentiation–associated prognostic model for ccRCC that could be used to predict the outcomes of the ccRCC patients.
Collapse
Affiliation(s)
- Chen Liu
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuhui Zhang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Caoyang Hu
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuezhi Liang
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoming Cao
- Department of Urology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Dongwen Wang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of Urology, Cancer Hospital Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- *Correspondence: Dongwen Wang,
| |
Collapse
|
107
|
Chai D, Shi SY, Sobhani N, Ding J, Zhang Z, Jiang N, Wang G, Li M, Li H, Zheng J, Bai J. IFI35 Promotes Renal Cancer Progression by Inhibiting pSTAT1/pSTAT6-Dependent Autophagy. Cancers (Basel) 2022; 14:cancers14122861. [PMID: 35740527 PMCID: PMC9221357 DOI: 10.3390/cancers14122861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Interferon-induced protein 35 (IFI35), is currently acknowledged to govern the virus-related immune inflammatory responses. However, the biological significance and function of IFI35 in renal cell cancer (RCC) is still not well understood. Here, IFI35 expression and function were investigated in RCC tissues, renal cancer cells, and animal models. The results showed that IFI35 expression was significantly increased in 200 specimens of RCC patients. We found that higher IFI35 levels were significantly correlated with poor RCC prognosis. In human cell lines, the knockdown of IFI35 suppressed the malignant behavior of renal cancer cells. Similarly, the IFI35 knockdown resulted in significant inhibition of tumor progression in the subcutaneous or lung metastasis mouse model. Furthermore, the knockdown of IFI35 promoted the induction of autophagy by enhancing the autophagy-related gene expression (LC3-II, Beclin-1, and ATG-5). Additionally, blockade of STAT1/STAT6 phosphorylation (pSTAT1/pSTAT6) abrogated the induced autophagy by IFI35 knockdown in renal cancer cells. The autophagy inhibitor 3-MA also abolished the prevention of tumor growth by deleting IFI35 in renal cancer models. The above results suggest that the knockdown of IFI35 suppressed tumor progression of renal cancer by pSTAT1/pSTAT6-dependent autophagy. Our research revealed that IFI35 may serve as a potential diagnosis and therapeutic target for RCC.
Collapse
Affiliation(s)
- Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Shang Yuchen Shi
- Department of Stereotactic Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China;
| | - Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jiage Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Zichun Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China;
| | - Nan Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China;
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Hailong Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China;
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
- Correspondence: (J.Z.); (J.B.)
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
- Correspondence: (J.Z.); (J.B.)
| |
Collapse
|
108
|
Safaei S, Sajed R, Saeednejad Zanjani L, Rahimi M, Fattahi F, Ensieh Kazemi-Sefat G, Razmi M, Dorafshan S, Eini L, Madjd Z, Ghods R. Overexpression of cytoplasmic dynamin 2 is associated with worse outcomes in patients with clear cell renal cell carcinoma. Cancer Biomark 2022; 35:27-45. [DOI: 10.3233/cbm-210514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Dynamin 2 (DNM2) involved in tumor progression in various malignancies. OBJECTIVE: For the first time, we evaluated DNM2 expression pattern, its association with clinicopathological characteristics and survival outcomes in RCC subtypes. METHODS: We evaluated the DNM2 expression pattern in RCC tissues as well as adjacent normal tissue using immunohistochemistry on tissue microarray (TMA) slides. RESULTS: Our findings revealed increased DNM2 expression in RCC samples rather than in adjacent normal tissues. The results indicated that there was a statistically significant difference between cytoplasmic expression of DNM2 among subtypes of RCC in terms of intensity of staining, percentage of positive tumor cells, and H-score (P= 0.024, 0.049, and 0.009, respectively). The analysis revealed that increased cytoplasmic expression of DNM2 in ccRCC is associated with worse OS (log rank: P= 0.045), DSS (P= 0.049), and PFS (P= 0.041). Furthermore, cytoplasmic expression of DNM2 was found as an independent prognostic factor affecting DSS and PFS in multivariate analysis. CONCLUSIONS: Our results indicated that DNM2 cytoplasmic expression is associated with tumor aggressiveness and poor outcomes. DNM2 could serve as a promising prognostic biomarker and therapeutic target in patients with ccRCC.
Collapse
Affiliation(s)
- Sadegh Safaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Roya Sajed
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Mandana Rahimi
- Hasheminejad Kidney Center, Pathology department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fahimeh Fattahi
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Golnaz Ensieh Kazemi-Sefat
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Razmi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Shima Dorafshan
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leila Eini
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Division of Histology, Department of Basic Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Madjd
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Roya Ghods
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
109
|
Extracellular Vesicles—A New Potential Player in the Immunology of Renal Cell Carcinoma. J Pers Med 2022; 12:jpm12050772. [PMID: 35629194 PMCID: PMC9144962 DOI: 10.3390/jpm12050772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
The incidence of renal cell carcinoma (RCC) has doubled in the developed world within the last fifty years, and now it is responsible for 2–3% of diagnosed cancers. The delay in diagnosis and the not fully understood pathogenesis are the main challenges that have to be overcome. It seems that extracellular vesicles (EVs) are one of the key players in tumor development since they ensure a proper microenvironment for the tumor cells. The stimulation of angiogenesis and immunosuppression is mediated by molecules contained in EVs. It was shown that EVs derived from cancer cells can inhibit T cell proliferation, natural killer lymphocyte activation, and dendritic cell maturation by this mechanism. Moreover, EVs may be a biomarker for the response to anti-cancer treatment. In this review, we sum up the knowledge about the role of EVs in RCC pathogenesis and show their future perspectives in this field.
Collapse
|
110
|
Yang J, Zhou Y, Li Y, Hu W, Yuan C, Chen S, Ye G, Chen Y, Wu Y, Liu J, Wang Y, Du J, Tong X. Functional deficiency of succinate dehydrogenase promotes tumorigenesis and development of clear cell renal cell carcinoma through weakening of ferroptosis. Bioengineered 2022; 13:11187-11207. [PMID: 35510387 PMCID: PMC9278435 DOI: 10.1080/21655979.2022.2062537] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal carcinomas, with high mortality and poor prognoses worldwide. Succinate dehydrogenase (SDH) consists of four nuclear-encoded subunits and it is the only complex involved in both the tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS). Previous studies have shown decreased SDH activity in ccRCC. However, the role and underlying molecular mechanisms of SDH in ccRCC initiation and development remain unclear. In the present study, pan-cancer analysis of SDH gene expression was analyzed and the relationship between SDH gene expression and clinicopathological parameters was assessed using different databases. cBioPortal, UACLAN, and Tumor Immune Estimation Resource (TIMER) were subsequently utilized to analyze genetic alterations, methylation, and immune cell infiltration of SDH genes in ccRCC patients. We found SDHs were significantly downregulated in ccRCC tissues and correlated with ccRCC progression. Increased methylation and high SDH promoter mutation rates may be the cause of reduced expression of SDHs in ccRCC. Moreover, the interaction network showed that SDH genes were correlated with ferroptosis-related genes. We further demonstrated that SDH inhibition dampened oxidative phosphorylation, reduced ferroptotic events, and restored ferroptotic cell death, characterized by eliminated mitochondrial ROS levels, decreased cellular ROS and diminished peroxide accumulation. Collectively, this study provides new insights into the regulatory role of SDH in the carcinogenesis and progression of ccRCC, introducing a potential target for advanced antitumor therapy through ferroptosis.
Collapse
Affiliation(s)
- Jing Yang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yi Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wanye Hu
- Graduate School, Bengbu Medical College, Bengbu, Anhui, China
| | - Chen Yuan
- Graduate School, Bengbu Medical College, Bengbu, Anhui, China
| | - Shida Chen
- Graduate School, Bengbu Medical College, Bengbu, Anhui, China
| | - Gaoqi Ye
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Yuzhou Chen
- Pittsburgh Institute, Sichuan University, Chengdu, Sichuan, China
| | - Yunyi Wu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Jing Liu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ying Wang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.,Department of Central Laboratory, Affiliated Hangzhou first people's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Du
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Xiangmin Tong
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Graduate School, Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
111
|
Causey J, Stubblefield J, Qualls J, Fowler J, Cai L, Walker K, Guan Y, Huang X. An Ensemble of U-Net Models for Kidney Tumor Segmentation With CT Images. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1387-1392. [PMID: 34061750 PMCID: PMC9210325 DOI: 10.1109/tcbb.2021.3085608] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
We present here the Arkansas AI-Campus solution method for the 2019 Kidney Tumor Segmentation Challenge (KiTS19). Our Arkansas AI-Campus team participated the KiTS19 Challenge for four months, from March to July of 2019. This paper provides a summary of our methods, training, testing and validation results for this grand challenge in biomedical imaging analysis. Our deep learning model is an ensemble of U-Net models developed after testing many model variations. Our model has consistent performance on the local test dataset and the final competition independent test dataset. The model achieved local test Dice scores of 0.949 for kidney and tumor segmentation, and 0.601 for tumor segmentation, and the final competition test earned Dice scores 0.9470 and 0.6099 respectively. The Arkansas AI-Campus team solution with a composite DICE score of 0.7784 has achieved a final ranking of top fifty worldwide, and top five among the United States teams in the KiTS19 Competition.
Collapse
|
112
|
Qi Q, Sun Y, Yang Y, Liu Y. Circ_0000274 contributes to renal cell carcinoma progression by regulating miR-338-3p/NUCB2 axis and JAK1/STAT3 pathway. Transpl Immunol 2022; 74:101626. [DOI: 10.1016/j.trim.2022.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 10/18/2022]
|
113
|
Chae MS, Shim JW, Choi H, Hong SH, Lee JY, Jeong W, Lee B, Kim E, Hong SH. Effects of Multimodal Bundle with Remote Ischemic Preconditioning and Intrathecal Analgesia on Early Recovery of Estimated Glomerular Filtration Rate after Robot-Assisted Laparoscopic Partial Nephrectomy for Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14081985. [PMID: 35454891 PMCID: PMC9032668 DOI: 10.3390/cancers14081985] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This study suggested that robot-assisted laparoscopic partial nephrectomy (RALPN) may have benefits with regard to the preservation of renal function and few complications postoperatively in patients with renal cell carcinoma (RCC). However, a reduction in the estimated glomerular filtration rate may be unavoidable. Our results suggested that the preservation of renal function may be enhanced by combining robot-assisted nephron-sparing surgery with an intraoperative bundle strategy consisting of remote ischemic preconditioning (RIPC) and an intrathecal morphine block (ITMB), to protect against ischemia–reperfusion injury and the pain-related stress induced by renal artery clamping and surgical insults. It is important to adjust modifiable variables related to the progression of renal impairment in a timely and appropriate manner for the recovery of renal function after RALPN. Together with surgical and pharmacological methods to minimize irreversible injury, RIPC and ITMB combined bundle therapy may relieve ischemia–reperfusion- and pain-induced stress and serve as a safe and efficient method for improving renal outcomes of RALPN in patients with RCC. Abstract We investigated the effects of multimodal combined bundle therapy, consisting of remote ischemic preconditioning (RIPC) and intrathecal morphine block (ITMB), on the early recovery of kidney function after robot-assisted laparoscopic partial nephrectomy (RALPN) in patients with renal cell carcinoma (RCC). In addition, we compared the surgical and analgesic outcomes between patients with and without bundle treatment. This prospective randomized double-blind controlled trial was performed in a cohort of 80 patients with RCC, who were divided into two groups: a bundle group (n = 40) and non-bundle group (n = 40). The primary outcome was postoperative kidney function, defined as the lowest estimated glomerular filtration rate (eGFR) on postoperative day (POD) 2. Surgical complications, pain, and length of hospital stay were assessed as secondary outcomes. The eGFR immediately after surgery was significantly lower in the bundle group compared to the preoperative baseline, but serial levels on PODs 1 and 2 and at three and six months after surgery were comparable to the preoperative baseline. The eGFR level immediately after surgery was lower in the non-bundle than bundle group, and serial levels on PODs 1 and 2 and at three months after surgery remained below the baseline. The eGFR level immediately after surgery was higher in the bundle group than in the non-bundle group. The eGFR changes immediately after surgery, and on POD 1, were smaller in the bundle than in the non-bundle group. The non-bundle group had longer hospital stays and more severe pain than the bundle group, but there were no severe surgical complications in either group. The combined RIPC and ITMB bundle may relieve ischemia–reperfusion- and pain-induced stress, as a safe and efficient means of improving renal outcomes following RALPN in patients with RCC.
Collapse
Affiliation(s)
- Min Suk Chae
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.S.C.); (J.-W.S.); (H.C.)
| | - Jung-Woo Shim
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.S.C.); (J.-W.S.); (H.C.)
| | - Hoon Choi
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.S.C.); (J.-W.S.); (H.C.)
| | - Sung Hoo Hong
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.H.H.); (J.Y.L.)
| | - Ji Youl Lee
- Department of Urology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (S.H.H.); (J.Y.L.)
| | - Woohyung Jeong
- Department of Anesthesiology and Pain Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Bongsung Lee
- Department of Anesthesiology and Pain Medicine, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Eunji Kim
- Department of Anesthesiology and Pain Medicine, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Sang Hyun Hong
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (M.S.C.); (J.-W.S.); (H.C.)
- Correspondence: ; Tel.: +82-2-2258-6150; Fax: +82-2-537-1951
| |
Collapse
|
114
|
De Alwis R, Hansson J, Lindgren D, Schoch S, Tejera A, Scholtz B, Elfving P, Möller C, Nilsson H, Johansson M, Axelson H. Size‑based isolation and detection of renal carcinoma cells from whole blood. Mol Clin Oncol 2022; 16:101. [PMID: 35463211 PMCID: PMC9022084 DOI: 10.3892/mco.2022.2534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/18/2022] [Indexed: 11/25/2022] Open
Abstract
Renal cell carcinoma (RCC) is a tumour type with an indolent growth pattern and rather vague symptoms. The present study developed a platform for liquid biopsy of RCC based upon the isolation of circulating tumour cells (CTCs). Founded on the observation that RCC tumour cells are considerably larger than leucocytes, the present study employed a microfluidics-based system for isolation of RCC CTCs from whole blood. Using this system, it was revealed that 66% of spiked-in RCC tumour cells could be retrieved using this approach. Furthermore, it was demonstrated that these cells could be molecularly detected with digital PCR using RCC-specific genes down to one tumour cell, whilst avoiding detection in samples lacking tumour cells. Finally, subtype specific transcripts were identified to distinguish the different subtypes of RCC, which were then validated in patient tumours. The present study established a novel workflow for the isolation of RCC CTCs from whole blood, with the potential to detect these cells irrespective of subtype.
Collapse
Affiliation(s)
- Roger De Alwis
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Jennifer Hansson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - David Lindgren
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Sarah Schoch
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Alexander Tejera
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Bianca Scholtz
- Department of Urology, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Peter Elfving
- Department of Urology, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Christina Möller
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Helén Nilsson
- Center for Molecular Pathology, Department of Translational Medicine, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Martin Johansson
- Center for Molecular Pathology, Department of Translational Medicine, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Håkan Axelson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| |
Collapse
|
115
|
Jin J, Zhou TJ, Ren GL, Cai L, Meng XM. Novel insights into NOD-like receptors in renal diseases. Acta Pharmacol Sin 2022; 43:2789-2806. [PMID: 35365780 PMCID: PMC8972670 DOI: 10.1038/s41401-022-00886-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 11/09/2022] Open
Abstract
Nucleotide-binding oligomerization domain-like receptors (NLRs), including NLRAs, NLRBs (also known as NAIPs), NLRCs, and NLRPs, are a major subfamily of pattern recognition receptors (PRRs). Owing to a recent surge in research, NLRs have gained considerable attention due to their involvement in mediating the innate immune response and perpetuating inflammatory pathways, which is a central phenomenon in the pathogenesis of multiple diseases, including renal diseases. NLRs are expressed in different renal tissues during pathological conditions, which suggest that these receptors play roles in acute kidney injury, obstructive nephropathy, diabetic nephropathy, IgA nephropathy, lupus nephritis, crystal nephropathy, uric acid nephropathy, and renal cell carcinoma, among others. This review summarises recent progress on the functions of NLRs and their mechanisms in the pathophysiological processes of different types of renal diseases to help us better understand the role of NLRs in the kidney and provide a theoretical basis for NLR-targeted therapy for renal diseases.
Collapse
|
116
|
Jiang W, Zeng H, Liu Z, Jin K, Hu B, Chang Y, Liu L, Zhu Y, Xu L, Wang Z, Guo J, Xu J. Immune inactivation by CD47 expression predicts clinical outcomes and therapeutic responses in clear cell renal cell carcinoma patients. Urol Oncol 2022; 40:166.e15-166.e25. [PMID: 34998671 DOI: 10.1016/j.urolonc.2021.11.024] [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: 08/11/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND CD47 has been identified as a phagocytosis checkpoint conferring poor clinical outcomes in various cancer types. A flurry of clinical trials designed to evaluate agents that block CD47 have been initiated. We aimed to explore the clinical significance of CD47 and its correlation with immune infiltration and molecular features in clear cell renal cell carcinoma (ccRCC). METHODS 235 tumor tissue microarray specimens of ccRCC patients from Zhongshan Hospital, 530 ccRCC patients from The Cancer Genome Atlas and 726 ccRCC patients from JAVELIN Renal 101 study were analyzed. CD47 expression and immune contexture were examined by immunohistochemistry and CIBERSORT algorithm. Survival analyses were conducted through Kaplan-Meier curves and Cox regression model. RESULTS We demonstrated that ccRCC patients with high CD47 expression exhibited inferior overall survival and recurrence-free survival. CD47 expression associated with heavily immune infiltrated but immunosuppressed microenvironment. CD8+ T cells infiltration had discordant prognostic value based on CD47 expression, where high CD8+ T cell infiltration was associated with worse clinical outcome in CD47hi patients and with favorable prognosis in CD47lo patients. Patients with mutated PBRM1 and SETD2 correlated with decreased CD47 mRNA expression. Patients with higher CD47 expression possessed improved PFS in ICI + VEGFR TKI combination therapy. CONCLUSIONS CD47 expression was an independent prognosticator of clinical outcome for ccRCC patients. CD47 expression correlated with ccRCC molecular classification and response to combination therapy. The phagocytosis checkpoint CD47 could be applied as an attractive candidate for immunotherapeutic approach in ccRCC.
Collapse
Affiliation(s)
- Wenbin Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Han Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaopei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaifeng Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Baoying Hu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
117
|
Long Noncoding RNA MMP2-AS1 Contributes to Progression of Renal Cell Carcinoma by Modulating miR-34c-5p/MMP2 Axis. JOURNAL OF ONCOLOGY 2022; 2022:7346460. [PMID: 35342412 PMCID: PMC8942703 DOI: 10.1155/2022/7346460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/23/2022]
Abstract
Renal cell carcinoma (RCC) serves as a prevalent malignancy of urinary system and presents severe mortality and increasing incidence. Long noncoding RNAs (lncRNAs) have demonstrated critical roles in RCC development. Here, we were interested in the function of MMP2-AS1 during RCC progression. We observed that MP2-AS1 localized in both nucleus and cytoplasm of RCC cells using fluorescent in situ hybridization (FISH). The cell viability, proliferation, invasion, and migration of RCC cells were reduced by the depletion of MMP2-AS1. The MMP2-AS1 depletion-inhibited viability, proliferation, migration, and invasion of RCC cells were rescued by the overexpression of MMP2 in vitro. Consistently, the tumor growth of RCC cells was repressed by the depletion of MMP2-AS1 in the nude mice, while the overexpression of MMP2 could reverse this effect in vivo. Mechanically, we predicted the potential interaction of miR-34c-5p with both MMP2-AS1 and MMP2. The treatment of miR-34c-5p mimic reduced the luciferase activity of MMP2-AS1 and MMP2 3'UTR. The depletion of MMP2-AS1 enhanced miR-34c-5p expression and the expression of MMP2 was inhibited by miR-34c-5p in RCC cells. The protein levels of MMP2 were downregulated by MMP2-AS1 knockdown, while the inhibitor of miR-34c-5p rescued the expression of MMP2 in the cells. The treatment of miR-34c-5p mimic attenuated the cell viability, proliferation, invasion, and migration of RCC cells, in which MMP2 overexpression restored the phenotypes. MMP2-AS1 depletion-attenuated viability, proliferation, migration, and invasion of RCC cells were reversed by miR-34c-5p inhibitor. We concluded that MMP2-AS1 contributed to progression of renal cell carcinoma by modulating miR-34c-5p/MMP2 axis.
Collapse
|
118
|
Tanio M, Fukiage Y, Muramoto A, Yokoyama O, Kobayashi M. Proposal of sialyl Lewis x/a as prognostic biomarkers in clear cell renal cell carcinoma: A study on a cohort of 117 patients submitted to curative surgery. JOURNAL OF CLINICAL UROLOGY 2022. [DOI: 10.1177/20514158221082884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective: Metastatic recurrence has been reported to occur in 20–30% of patients with clear cell renal cell carcinoma (ccRCC). Although the prognosis of these patients is poor, no marker has been established to predict metastatic potential and/or prognosis. Therefore, we investigated membrane expression of sialyl Lewis x (sLex) and sialyl Lewis a (sLea), which is generally considered to be associated with cancer metastasis. Materials and methods: We enrolled 117 patients who underwent curative surgery for RCC and were pathologically diagnosed as ccRCC. Immunohistochemistry for sLex and sLea was performed to evaluate the signal intensity on the cell membrane. We statistically analysed whether membrane expression of sLex/sLea is correlated with clinicopathological parameters and prognosis. Results: Of the 117 patients, 72 were classified as sLex-positive and 44 as sLea-positive. The sLex-positive group had significantly shorter progression-free survival (PFS) and overall survival (OS) than the negative group. Similarly, the sLea-positive group had significantly shorter PFS than the negative group, and it showed a trend towards a reduction of OS, although it did not reach statistical significance, a fact that could be due to the small sample size. Conclusion: Both sLex and sLea could be possible future prognostic indicators in ccRCC. Level of evidence: Level 3
Collapse
Affiliation(s)
- Makoto Tanio
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Japan
- Department of Urology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Yusuke Fukiage
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Japan
- Department of Urology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Akifumi Muramoto
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Japan
- Division of Surgical Pathology, University of Fukui Hospital, Japan
| | - Osamu Yokoyama
- Department of Urology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Motohiro Kobayashi
- Department of Tumor Pathology, Faculty of Medical Sciences, University of Fukui, Japan
| |
Collapse
|
119
|
AC010973.2 promotes cell proliferation and is one of six stemness-related genes that predict overall survival of renal clear cell carcinoma. Sci Rep 2022; 12:4272. [PMID: 35277527 PMCID: PMC8917182 DOI: 10.1038/s41598-022-07070-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 02/02/2022] [Indexed: 11/09/2022] Open
Abstract
Extensive research indicates that tumor stemness promotes tumor progression. Nonetheless, the underlying roles of stemness-related genes in renal clear cell carcinoma (ccRCC) are unclear. Data used in bioinformatics analysis were downloaded from The Cancer Genome Atlas (TCGA) database. Moreover, the R software, SPSS, and GraphPad Prism 8 were used for mapping and statistical analysis. First, the stemness index of each patient was quantified using a machine learning algorithm. Subsequently, the differentially expressed genes between high and low stemness index were identified as stemness-related genes. Based on these genes, a stable and effective prognostic model was identified to predict the overall survival of patients using a random forest algorithm (Training cohort; 1-year AUC: 0.67; 3-year AUC: 0.79; 5-year AUC: 0.73; Validation cohort; 1-year AUC: 0.66; 3-year AUC: 0.71; 5-year AUC: 0.7). The model genes comprised AC010973.2, RNU6-125P, AP001209.2, Z98885.1, KDM5C-IT1, and AL021368.3. Due to its highest importance evaluated by randomforst analysis, the AC010973.2 gene was selected for further research. In vitro experiments demonstrated that AC010973.2 is highly expressed in ccRCC tissue and cell lines. Meanwhile, its knockdown could significantly inhibit the proliferation of ccRCC cells based on colony formation and CCK8 assays. In summary, our findings reveal that the stemness-related gene AC01097.3 is closely associated with the survival of patients. Besides, it remarkably promotes cell proliferation in ccRCC, hence a novel potential therapeutic target.
Collapse
|
120
|
Application of 18F Prostate-Specific Membrane Antigen Positron Emission Tomography/Computed Tomography in Monitoring Gastric Metastasis and Cancer Thrombi from Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5681463. [PMID: 35154318 PMCID: PMC8837453 DOI: 10.1155/2022/5681463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Background. Renal cell carcinoma (RCC) with gastric metastasis is rare, particularly accompanied by multiple cancer thrombi. Methods. We reported a 66-year-old man with a history of a right radical nephrectomy because of RCC. The patient underwent 18F prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) scanning after 6 months of targeted therapy because of gastric metastasis and cancer thrombi. We conducted a systematic review of the literature and identified 73 cases of RCC with gastric metastasis. We analyzed the clinicopathological characteristics, therapies, and outcomes of patients. Results. 18F-PSMA PET/CT showed a large mass in the gastric fundus and cancer thrombi in the right atrium, inferior vena cava, and splenic vein with intense tracer uptake. Other metastases with increased tracer uptake included multiple bones and abdominal lymph nodes. The majority of gastric metastasis of RCC were men (53/73, 72.6%), with a median age at presentation of 67 (from 48 to 87) years. Gastric metastasis of RCC was mainly metachronous, and presented with small polyps or mass appearance and often accompanied by multiple-site metastases and gastrointestinal symptoms. An overall median interval between nephrectomy and diagnosis of gastric metastasis was 6 (from 0.1 to 23) years, and an overall median survival time was 14 (from 0.25 to 72) months. The median interval time of solitary gastric metastasis was longer than gastric metastasis with multiple-site metastases (7 vs.5 years;
). Patients with gastric and multiple-site metastases had higher mortality than patients with solitary metastasis (17 vs.1;
). The patients with synchronous gastric metastasis had a shorter survival time than metachronous gastric metastasis (6 vs.17 months;
). Conclusions. Postoperative follow-up of multiple imaging modalities to monitor recurrence and metastasis is necessary and important. PSMA PET/CT can improve the detection sensitivity of RCC, especially in metastatic clear cell renal cell carcinoma (ccRCC), and could provide a basis for disease staging, restaging, and therapeutic efficacy evaluation.
Collapse
|
121
|
Huang J, Guo J, Pedrosa I, Fei B. Deep Learning-based Deformable Registration of Dynamic Contrast-Enhanced MR Images of the Kidney. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2022; 12034:1203410. [PMID: 36793654 PMCID: PMC9928502 DOI: 10.1117/12.2611768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Respiratory motion is a major contributor to bias in quantitative analysis of magnetic resonance imaging (MRI) acquisitions. Deformable registration of three-dimensional (3D) dynamic contrast-enhanced (DCE) MRI data improves estimation of kidney kinetic parameters. In this study, we proposed a deep learning approach with two steps: a convolutional neural network (CNN) based affine registration network, followed by a U-Net trained for deformable registration between two MR images. The proposed registration method was applied successively across consecutive dynamic phases of the 3D DCE-MRI dataset to reduce motion effects in the different kidney compartments (i.e., cortex, medulla). Successful reduction in the motion effects caused by patient respiration during image acquisition allows for improved kinetic analysis of the kidney. Original and registered images were analyzed and compared using dynamic intensity curves of the kidney compartments, target registration error of anatomical markers, image subtraction, and simple visual assessment. The proposed deep learning-based approach to correct motion effects in abdominal 3D DCE-MRI data can be applied to various kidney MR imaging applications.
Collapse
Affiliation(s)
- James Huang
- University of Texas at Dallas, Dept of Bioengineering, Richardson, TX
- University of Texas at Dallas, Center for Imaging and Surgical Innovation, Richardson, TX
| | - Junyu Guo
- University of Texas Southwestern Medical Center, Dept of Radiology, Dallas, TX
| | - Ivan Pedrosa
- University of Texas Southwestern Medical Center, Dept of Radiology, Dallas, TX
| | - Baowei Fei
- University of Texas at Dallas, Dept of Bioengineering, Richardson, TX
- University of Texas at Dallas, Center for Imaging and Surgical Innovation, Richardson, TX
- University of Texas Southwestern Medical Center, Dept of Radiology, Dallas, TX
| |
Collapse
|
122
|
Wang J, Zhanghuang C, Tan X, Mi T, Liu J, Jin L, Li M, Zhang Z, He D. Development and Validation of a Nomogram to Predict Distant Metastasis in Elderly Patients With Renal Cell Carcinoma. Front Public Health 2022; 9:831940. [PMID: 35155365 PMCID: PMC8831843 DOI: 10.3389/fpubh.2021.831940] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/24/2021] [Indexed: 12/09/2022] Open
Abstract
BackgroundRenal cell carcinoma (RCC) is the most common renal malignant tumor in elderly patients. The prognosis of renal cell carcinoma with distant metastasis is poor. We aim to construct a nomogram to predict the risk of distant metastasis in elderly patients with RCC to help doctors and patients with early intervention and improve the survival rate.MethodsThe clinicopathological information of patients was downloaded from SEER to identify all elderly patients with RCC over 65 years old from 2010 to 2018. Univariate and multivariate logistic regression analyzed the training cohort's independent risk factors for distant metastasis. A nomogram was established to predict the distant metastasis of elderly patients with RCC based on these risk factors. We used the consistency index (C-index), calibration curve, and area under the receiver operating curve (AUC) to evaluate the accuracy and discrimination of the prediction model. Decision curve analysis (DCA) was used to assess the clinical application value of the model.ResultsA total of 36,365 elderly patients with RCC were included in the study. They were randomly divided into the training cohort (N = 25,321) and the validation cohort (N = 11,044). In the training cohort, univariate and multivariate logistic regression analysis suggested that race, tumor histological type, histological grade, T stage, N stage, tumor size, surgery, radiotherapy, and chemotherapy were independent risk factors for distant metastasis elderly patients with RCC. A nomogram was constructed to predict the risk of distant metastasis in elderly patients with RCC. The training and validation cohort's C-indexes are 0.949 and 0.954, respectively, indicating that the nomogram has excellent accuracy. AUC of the training and validation cohorts indicated excellent predictive ability. DCA suggested that the nomogram had a better clinical application value than the traditional TN staging.ConclusionThis study constructed a new nomogram to predict the risk of distant metastasis in elderly patients with RCC. The nomogram has excellent accuracy and reliability, which can help doctors and patients actively monitor and follow up patients to prevent distant metastasis of tumors.
Collapse
Affiliation(s)
- Jinkui Wang
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Chenghao Zhanghuang
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Urology, Yunnan Key Laboratory of Children's Major Disease Research, Kunming Children's Hospital (Children's Hospital Affiliated to Kunming Medical University), Kunming, China
| | - Xiaojun Tan
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Urology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical University, Nanchong, China
| | - Tao Mi
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jiayan Liu
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liming Jin
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Mujie Li
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaoxia Zhang
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dawei He
- Department of Urology, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Dawei He
| |
Collapse
|
123
|
Huang Y, Chen S, Xiao L, Qin W, Li L, Wang Y, Ma L, Yuan X. A Novel Prognostic Signature for Survival Prediction and Immune Implication Based on SARS-CoV-2–Related Genes in Kidney Renal Clear Cell Carcinoma. Front Bioeng Biotechnol 2022; 9:744659. [PMID: 35141213 PMCID: PMC8819071 DOI: 10.3389/fbioe.2021.744659] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/31/2021] [Indexed: 12/28/2022] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) is a common aggressive malignancy of the urinary system. COVID-19, a highly infectious and severe disease caused by SARS-CoV-2, has become a significant challenge for global public health. Cancer patients have been reported to be more vulnerable to SARS-CoV-2 infection and have a higher risk for serious complications than the general population. However, the correlation between KIRC and COVID-19 remains incompletely elucidated. In this study, we comprehensively investigated the expression and prognostic significance of 333 SARS-CoV-2 infection–related genes in KIRC using the TCGA dataset and identified 31 SARS-CoV-2–related differently expressed genes between KIRC and normal renal tissues. Based on these genes, we constructed and validated a 5-gene prognostic signature (including ACADM, CENPF, KDELC1, PLOD2, and TRMT1) to distinguish low- and high-risk KIRC patients of poor survival in TCGA and E-MTAB-1980 cohorts. Gene set enrichment analysis (GSEA) showed that some inflammatory/immune-related pathways were significantly enriched in the high-risk group. The ESTIMATE analysis indicated that patients in the high-risk group had higher stromal and immune cell scores, therefore lower tumor purity. Moreover, they presented higher proportions of macrophages M0, regulatory T cells (Tregs), and T follicular helper cells and higher expression of immune checkpoints CTLA-4, LAG-3, TIGIT, and PDCD1 than low-risk patients. Besides, we also developed a nomogram to expand clinical applicability, which exhibits excellent predictive accuracy for survival. In conclusion, we identified a novel prognostic signature and nomogram based on SARS-CoV-2–related genes as reliable prognostic predictors for KIRC patients and provided potential therapeutic targets for KIRC and COVID-19.
Collapse
Affiliation(s)
- Yongbiao Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yali Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Xianglin Yuan,
| |
Collapse
|
124
|
Renner AM, Derichsweiler C, Ilyas S, Gessner I, Fries JWU, Mathur S. High efficiency capture of biomarker miRNA15a for noninvasive diagnosis of malignant kidney tumors. Biomater Sci 2022; 10:1113-1122. [PMID: 35048092 DOI: 10.1039/d1bm01737c] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To date, there are no preoperative and quantitative dynamics in clinical practice that can reliably differentiate between a benign and malignant renal cell carcinoma (RCC). For monitoring different analytes in body fluids, more than 40 different molecular biomarkers have been identified, however, they are associated with limited clinical sensitivity and/or non-optimal specificity due to their leaky nature. Previous work on RCC demonstrated the miRNA15a to be reliable and novel biomarker with 98.1% specificity and 100% sensitivity. Despite the high potential of miRNA15a biomarker, its clinical application is considerably hampered by the insensitive nature of the detection methods and low concentration of biomarker in samples that is aggravated by the high level of contamination due to other solutes present in body fluids. In this work, a non-invasive quantitative approach is demonstrated to overcome such diagnostics issues through biotin-streptavidin binding and fluorescence active magnetic nanocarriers that ensured prompt isolation, enrichment and purification of the biomarker miRNA15a from urine. The study demonstrates that detectable low levels of these miRNAs through miRNA capturing nanocarriers can potentially function as advanced diagnostic markers for the non-invasive investigation and early detection of renal cancer.
Collapse
Affiliation(s)
- Alexander M Renner
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Christina Derichsweiler
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Shaista Ilyas
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Isabel Gessner
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| | - Jochen W U Fries
- Institute of Urology/Pathology, University Hospital of Cologne, Kerpenerstr. 62, 50924 Cologne, Germany
| | - Sanjay Mathur
- Institute of Inorganic Chemistry, University of Cologne, Greinstr. 6, 50939 Cologne, Germany.
| |
Collapse
|
125
|
Konaka H. Editorial Comment to Novel cut-off values of time from diagnosis to systematic therapy predict the overall survival and the efficacy of targeted therapy in renal cell carcinoma: A long-term, follow-up, retrospective study. Int J Urol 2022; 29:220-221. [PMID: 35023233 DOI: 10.1111/iju.14794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/28/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Hiroyuki Konaka
- Department of Urology, Japanese Red Cross Society Kanazawa Hospital, Kanazawa, Ishikawa, Japan
| |
Collapse
|
126
|
Biostatistics of VHL-Gene Transfection in the Health Informatics Analysis of Renal Cell Carcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5297580. [PMID: 35035522 PMCID: PMC8759873 DOI: 10.1155/2022/5297580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/26/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022]
Abstract
Objective In this paper, we study the role of the VHL gene in regulating the proliferation and apoptosis of renal cell carcinoma, as well as the safety and transfection efficiency of ultrasound microbubble gene transfection technology. Method We use kidney cancer cell lines as an in vitro research object and apply ultrasound microbubble gene transfection technology to transfect the VHL gene into kidney cancer cell line (786-0). The proliferation and apoptosis of cells were measured to clarify the inhibitory effect of the VHL gene in renal cell carcinoma. After that, pEGFP-VHL was transfected using ultrasonic microbubble and liposome gene transfection techniques, respectively, and the transfection efficiency was measured by immunofluorescence. Results Compared with untreated and 786-0 cells that are transfected with empty vector, the expression level of VHL gene mRNA in 786-0 cells that are transfected with pcDNA3.1-VHL was significantly increased, and the cell growth inhibition rate was significantly higher. The rate of apoptosis increased significantly. Transfection efficiency of the pEGFP-VHL gene after transfection of 786-0 cells for 48 h: control group 0, liposome group (35.55 ± 2.77) %, ultrasound microbubble group (18.27 ± 2.83) %, and two transfection methods on cells. There is no significant difference in the impact of vitality. Conclusion VHL gene expression can significantly inhibit the proliferation ability of renal cancer cell line 786-0 and promote its apoptosis. VHL gene is a potential target for gene therapy of kidney cancer.
Collapse
|
127
|
Xi W, Hou Y, Hu X, Xia Y, Jiang S, Wang H, Bai Q, Hou J, Guo J. Prognostic significance of pseudocapsule status in patients with metastatic renal cell carcinoma treated with tyrosine kinase inhibitors. Transl Androl Urol 2022; 10:4132-4141. [PMID: 34984179 PMCID: PMC8661261 DOI: 10.21037/tau-21-429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/22/2021] [Indexed: 11/06/2022] Open
Abstract
Background We sought to determine whether pseudocapsule (PS) features have prognostic implications in patients with metastatic renal cell carcinoma (mRCC). Methods We retrospectively reviewed 231 patients diagnosed with mRCC and treated with tyrosine kinase inhibitors; 188 patients with data available regarding the tumor-parenchyma interfacial PS of the primary tumor were enrolled for analysis. PS status was evaluated as intact (grade 0), merely involved (grade 1), penetrated (grade 2), and absent (grade 3). We applied the Kaplan-Meier method and Cox regression model to assess the survival impact. Results Of the 188 patients, 19 (10.1%), 61 (32.4%), 96 (51.1%) and 12 (6.4%) had grade 0, 1, 2 and 3 PS, respectively. PS status was significantly associated with histology (P=0.0206), venous tumor embolus (P=0.0511), microvascular invasion (P=0.0108) and microsatellite formation (P=0.0097). Patients without a PS had the worst overall survival (OS), with a 3-year OS rate of 12.7%, whereas the OS rates for grades 0, 1 and 2 were 78.8%, 50.8% and 43.6%, respectively. Adjusted by other variables, grade 3 and grade 2 PS gave rise to a much higher risk of death across the cohort [hazard ratio (HR) =5.217, P=0.0182; HR =3.765, P=0.0281, respectively]. Sarcomatoid change was also an independent factor for OS (HR =2.932, P=0.0075). In contrast, microsatellite formation was not associated with survival in the cohort. Conclusions PS status has prognostic implications for OS in metastatic renal cancer. The absence of the PS and sarcomatoid change are two pathological features related to an extremely poor prognosis.
Collapse
Affiliation(s)
- Wei Xi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyi Hu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Xia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hang Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Bai
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
128
|
Chung HC, Kang TW, Lee JY, Hwang EC, Park HJ, Hwang JE, Chang KD, Kim YH, Jung JH. Tumor enucleation for the treatment of T1 renal tumors: A systematic review and meta-analysis. Investig Clin Urol 2022; 63:126-139. [PMID: 35244986 PMCID: PMC8902429 DOI: 10.4111/icu.20210361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/21/2021] [Accepted: 01/06/2022] [Indexed: 11/27/2022] Open
Abstract
Purpose To evaluate the clinical efficacy and safety of tumor enucleation (TE) compared with partial nephrectomy (PN) for T1 renal cell carcinoma. Materials and Methods According to protocol, we searched multiple data sources for published and unpublished randomized controlled trials and nonrandomized studies (NRSs) in any language. We performed systematic review and meta-analysis according to the Cochrane Handbook for Systematic Reviews of Interventions and rated the certainty of the evidence (CoE) using the GRADE framework. Results We are uncertain about the effects of TE on perioperative (mean difference [MD] 3.38, 95% CI 1.52 to 5.23; I2=68%; 4 NRSs; 942 participants; very low CoE) and long-term (MD 2.31, 95% CI -1.40 to 6.01; I2=57%; 4 NRSs; 542 participants; very low CoE) residual renal function. TE may result in little to no difference in short-term residual renal function (MD 1.04, 95% CI 0.25 to 1.83; I2=0%; 2 NRSs; 256 participants; low CoE). We are uncertain about the effects of TE on cancer-specific mortality (risk ratio [RR] 0.90, 95% CI: 0.11 to 7.28; I2=0%; 2 NRSs; 551 participants; very low CoE) and major adverse events (RR 0.48, 95% CI: 0.30 to 0.79; I2=0%; 10 NRS; 2,360 participants; very low CoE). Conclusions While TE appears to have similar effects on short term postoperative residual renal function, there were uncertainties on mortality and major adverse events. However, we need rigorous RCTs to elucidate the effects of TE as the evidence stems mostly from NRSs.
Collapse
Affiliation(s)
- Hyun Chul Chung
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Tae Wook Kang
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Joon Young Lee
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Korea
- Center of Evidence Based Medicine, Institute of Convergence Science, Yonsei University, Seoul, Korea
| | - Eu Chang Hwang
- Department of Urology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hong Jun Park
- Center of Evidence Based Medicine, Institute of Convergence Science, Yonsei University, Seoul, Korea
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jun Eul Hwang
- Department of Hematology-Oncology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Ki Don Chang
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Hwan Kim
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jae Hung Jung
- Department of Urology, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
129
|
Diseases of the Kidney. Fam Med 2022. [DOI: 10.1007/978-3-030-54441-6_104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
130
|
Dahle DO, Skauby M, Langberg CW, Brabrand K, Wessel N, Midtvedt K. Renal Cell Carcinoma and Kidney Transplantation: A Narrative Review. Transplantation 2022; 106:e52-e63. [PMID: 33741842 PMCID: PMC8667800 DOI: 10.1097/tp.0000000000003762] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/08/2021] [Accepted: 03/03/2021] [Indexed: 11/27/2022]
Abstract
Kidney transplant recipients (KTRs) are at increased risk of developing renal cell carcinoma (RCC). The cancer can be encountered at different steps in the transplant process. RCC found during work-up of a transplant candidate needs treatment and to limit the risk of recurrence usually a mandatory observation period before transplantation is recommended. An observation period may be omitted for candidates with incidentally discovered and excised small RCCs (<3 cm). Likewise, RCC in the donor organ may not always preclude usage if tumor is small (<2 to 4 cm) and removed with clear margins before transplantation. After transplantation, 90% of RCCs are detected in the native kidneys, particularly if acquired cystic kidney disease has developed during prolonged dialysis. Screening for RCC after transplantation has not been found cost-effective. Treatment of RCC in KTRs poses challenges with adjustments of immunosuppression and oncologic treatments. For localized RCC, excision or nephrectomy is often curative. For metastatic RCC, recent landmark trials in the nontransplanted population demonstrate that immunotherapy combinations improve survival. Dedicated trials in KTRs are lacking. Case series on immune checkpoint inhibitors in solid organ recipients with a range of cancer types indicate partial or complete tumor response in approximately one-third of the patients at the cost of rejection developing in ~40%.
Collapse
Affiliation(s)
- Dag Olav Dahle
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Morten Skauby
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Knut Brabrand
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Nicolai Wessel
- Department of Urology, Oslo University Hospital, Oslo, Norway
| | - Karsten Midtvedt
- Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
131
|
Shimizu T, Hongo F, Takahashi H, Fujihara A, Ukimura O. Double renal cell carcinoma with histological type of clear cell carcinoma and papillary carcinoma in the same kidney concurrently treated with robot-assisted partial nephrectomy. IJU Case Rep 2022; 5:62-65. [PMID: 35005477 PMCID: PMC8720717 DOI: 10.1002/iju5.12395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/08/2021] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The presence of two different histologic types of renal cell carcinoma in the same kidney is rare in clinical practice. This report describes a patient with ipsilateral renal cell carcinomas, consisting of a clear cell renal cell carcinoma and a papillary type 1 renal cell carcinoma, who was successfully treated by robot-assisted partial nephrectomy. CASE PRESENTATION A 70-year man was referred to our hospital for the treatment of two right mid-pole renal tumors, measuring 51 mm and 31 mm in diameter. The two tumors, which differed in contrast enhancement on computed tomography, were removed simultaneously by robot-assisted partial nephrectomy. Histopathological and immunohistochemical findings confirmed that one tumor was a clear cell renal cell carcinoma, pT1a, and the other was a papillary type1 renal cell carcinoma, pT1b. CONCLUSION This report describes a rare patient presenting with two ipsilateral renal cell carcinomas differing in histology. Robot-assisted partial nephrectomy was the safe and effective nephron-sparing surgery, even in patients with complex double renal tumors.
Collapse
Affiliation(s)
- Teruki Shimizu
- Department of UrologyKyoto Prefectural University of MedicineKyotoJapan
| | - Fumiya Hongo
- Department of UrologyKyoto Prefectural University of MedicineKyotoJapan
| | - Hikaru Takahashi
- Department of UrologyKyoto Prefectural University of MedicineKyotoJapan
| | - Atsuko Fujihara
- Department of UrologyKyoto Prefectural University of MedicineKyotoJapan
| | - Osamu Ukimura
- Department of UrologyKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
132
|
Ding J, Jiang N, Zheng Y, Wang J, Fang L, Li H, Yang J, Hu A, Xiao P, Zhang Q, Chai D, Zheng J, Wang G. Adenovirus vaccine therapy with CD137L promotes CD8 + DCs-mediated multifunctional CD8 + T cell immunity and elicits potent anti-tumor activity. Pharmacol Res 2022; 175:106034. [PMID: 34915126 DOI: 10.1016/j.phrs.2021.106034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/10/2021] [Indexed: 11/27/2022]
Abstract
Renal carcinoma progresses aggressively in patients with metastatic disease while curative strategies are limited. Here, we constructed a recombinant non-replicating adenovirus (Ad) vaccine encoding an immune activator, CD137L, and a tumor antigen, CAIX, for treating renal carcinoma. In a subcutaneous tumor model, tumor growth was significantly suppressed in the Ad-CD137L/CAIX vaccine group compared with the single vaccine group. The induction and maturity of CD11C+ and CD8+CD11C+ dendritic cell (DC) subsets were promoted in Ad-CD137L/CAIX co-immunized mice. Furthermore, the Ad-CD137L/CAIX vaccine elicited stronger tumor-specific multifunctional CD8+ T cell immune responses as demonstrated by increased proliferation and cytolytic function of CD8+ T cells. Notably, depletion of CD8+ T cells greatly compromised the effective protection provided by Ad-CD137L/CAIX vaccine, suggesting an irreplaceable role of CD8+ T cells for the immunopotency of the vaccine. In both lung metastatic and orthotopic models, Ad-CD137L/CAIX vaccine treatment significantly decreased tumor metastasis and progression and increased the induction of tumor-specific multifunctional CD8+ T cells, in contrast to treatment with the Ad-CAIX vaccine alone. The Ad-CD137L/CAIX vaccine also augmented the tumor-specific multifunctional CD8+ T cell immune response in both orthotopic and metastatic models. These results indicated that Ad-CD137L/CAIX vaccine elicited a potent anti-tumor activity by inducing CD8+DC-mediated multifunctional CD8+ T cell immune responses. The potential strategy of CD137L-based vaccine might be served as a novel treatment for renal carcinoma or other malignant tumors.
Collapse
Affiliation(s)
- Jiage Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Nan Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Yanyan Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Jiawei Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Jie Yang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Ankang Hu
- Center of Animal laboratory, Xuzhou Medical University, Xuzhou, Jiangsu 221002 PR China
| | - Pengli Xiao
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| |
Collapse
|
133
|
The Role of Circulating Biomarkers in the Oncological Management of Metastatic Renal Cell Carcinoma: Where Do We Stand Now? Biomedicines 2021; 10:biomedicines10010090. [PMID: 35052770 PMCID: PMC8773056 DOI: 10.3390/biomedicines10010090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/25/2021] [Accepted: 12/29/2021] [Indexed: 01/08/2023] Open
Abstract
Renal cell carcinoma (RCC) is an increasingly common malignancy that can progress to metastatic renal cell carcinoma (mRCC) in approximately one-third of RCC patients. The 5-year survival rate for mRCC is abysmally low, and, at the present time, there are sparingly few if any effective treatments. Current surgical and pharmacological treatments can have a long-lasting impact on renal function, as well. Thus, there is a compelling unmet need to discover novel biomarkers and surveillance methods to improve patient outcomes with more targeted therapies earlier in the course of the disease. Circulating biomarkers, such as circulating tumor DNA, noncoding RNA, proteins, extracellular vesicles, or cancer cells themselves potentially represent a minimally invasive tool to fill this gap and accelerate both diagnosis and treatment. Here, we discuss the clinical relevance of different circulating biomarkers in metastatic renal cell carcinoma by clarifying their potential role as novel biomarkers of response or resistance to treatments but also by guiding clinicians in novel therapeutic approaches.
Collapse
|
134
|
Chen J, Liao X, Cheng J, Su G, Yuan F, Zhang Z, Wu J, Mei H, Tan W. Targeted Methylation of the LncRNA NEAT1 Suppresses Malignancy of Renal Cell Carcinoma. Front Cell Dev Biol 2021; 9:777349. [PMID: 34957107 PMCID: PMC8696001 DOI: 10.3389/fcell.2021.777349] [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: 09/15/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
Long-chain non-coding RNA (LncRNA) has been found to play an important role in the regulation of the occurrence and progression of renal cell carcinoma (RCC). In this study, we demonstrated that LncRNA NEAT1 expression and m6A methylation level was decreased in RCC tissues. Further, the downregulated expression level of LncRNA NEAT1 was associated with poor prognosis for RCC patients. Then we used CRIPSR/dCas13b-METTL3 to methylate LncRNA NEAT1 in RCC cells. The results showed that the expression level of LncRNA NEAT1 was upregulated after methylated by dCas13b-METTL3 in RCC cells. And the proliferation and migration ability of RCC cells was decreased after methylated LncRNA NEAT1. Finally, we examined the effect of LncRNA NEAT1 hypermethylation on the transcriptome. We found differentially expressed genes in RCC cells were associated with “cGMP-PKG signaling pathway”, “Cell adhesion molecules” and “Pathways in cancer”. In conclusion, CRISPR/Cas13b-METTL3 targeting LncRNA NEAT1 m6A methylation activates LncRNA NEAT1 expression and provides a new target for treatment of RCC.
Collapse
Affiliation(s)
- Jieqing Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xinhui Liao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jianli Cheng
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Ganglin Su
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Fen Yuan
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhongfu Zhang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jianting Wu
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Hongbing Mei
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
135
|
Lv Z, Cui B, Huang X, Feng HY, Wang T, Wang HF, Xuan YD, Li HZ, Ma X, Huang Y, Zhang X. FGL1 as a Novel Mediator and Biomarker of Malignant Progression in Clear Cell Renal Cell Carcinoma. Front Oncol 2021; 11:756843. [PMID: 34956878 PMCID: PMC8695555 DOI: 10.3389/fonc.2021.756843] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), which is the most prevalent renal cell carcinoma subtype, has a poor prognosis. Emerging strategies for enhancing the immune response in ccRCC therapy are currently being investigated. Fibrinogen-like Protein 1(FGL1) is a novel mechanism that tumors may use to evade the immune system by binding LAG-3 and negatively regulating T cells. In this study, we aimed at investigating the underlying mechanism of FGL1 in ccRCC, and its expression and prognostic value. We found that FGL1 was upregulated in tumor tissues and plasma specimens of ccRCC patients. High FGL1 expression predicted a poor prognosis for ccRCC patients. We also discovered that overexpression of FGL1 enhances RCC cell migration, invasion, and metastasis by activating the epithelial-to-mesenchymal transition (EMT). Consistent with these results, we identified a significant positive correlation between expression of FGL1 and EMT-related genes through tissue microarray analysis. Gene-expression analysis revealed that FGL1-deficient ccRCC cell lines had altered transcriptional output in inflammatory response, cell-cell signaling, negative regulation of T cell activation, and intracellular signal transduction. Depletion of FGL1 significantly inhibited tumor growth and lung metastasis in orthotopic xenograft mouse model. Infiltration of myeloid-derived CD11b+ and Ly6G+ immune cells in tumor microenvironment (TME) was strikingly decreased when FGL1 expression reduced. Therefore, increased FGL1 expression in ccRCC is positively correlated with poor prognosis. Mechanistically, FGL1 facilitates the EMT process and modulates TME, which promotes ccRCC progression and metastasis. Consequently, targeting FGL1 can potentially improve clinical outcome of ccRCC patients.
Collapse
Affiliation(s)
- Zheng Lv
- School of Medicine, Nankai University, Tianjin, China
| | - Bo Cui
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xing Huang
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hua-Yi Feng
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Tao Wang
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Han-Feng Wang
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yun-Dong Xuan
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Zhao Li
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xin Ma
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- School of Medicine, Nankai University, Tianjin, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
136
|
Zhang Y, Dai J, Huang W, Chen Q, Chen W, He Q, Chen F, Zhang P. Identification of a competing endogenous RNA network related to immune signature in clear cell renal cell carcinoma. Aging (Albany NY) 2021; 13:25980-26002. [PMID: 34958632 PMCID: PMC8751601 DOI: 10.18632/aging.203784] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 12/08/2021] [Indexed: 02/05/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a fatal cancer of the urinary system. Long non-coding RNAs (lncRNAs) act as competitive endogenous RNAs (ceRNAs) involving the ccRCC progression. However, the relationship between the ceRNA network and immune signature is largely unknown. In this study, the ccRCC-related gene expression profiles retrieved from the TCGA database were used first to identify the differentially expressed genes through differential gene expression analysis and weighted gene co-expression network analysis. The interaction among differentially expressed lncRNAs, miRNAs, and mRNAs were matched using public databases. As a result, a ceRNA network was developed that contained 144 lncRNAs, 23 miRNAs, as well as 62 mRNAs. Four of 144 lncRNAs including LINC00943, SRD5A3-AS1, LINC02345, and U62317.3 were identified through LASSO regression and Cox regression analyses, and were used to create a prognostic risk model. Then, the ccRCC samples were divided into the high- and low-risk groups depending on their risk scores. ROC curves, Kaplan-Meier survival analysis, and the survival risk plots indicated that the predictive performance of our developed risk model was accurate. Moreover, the CIBERSORT algorithm was used to measure the infiltration levels of immune cells in the ccRCC samples. The further genomic analysis illustrated a positive correlation between most immune checkpoint blockade-related genes and the risk score. In conclusion, the present findings effectually contribute to the comprehensive understanding of the ccRCC pathogenesis, and may offer a reference for developing novel therapeutic and prognostic biomarkers.
Collapse
Affiliation(s)
- Yuke Zhang
- Department of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Jiangwen Dai
- Department of Oncology, Chengdu Fifth People's Hospital of Chengdu University of TCM, Chengdu, China
| | - Weifeng Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingsong Chen
- Department of Traumatology, Chongqing University Central Hospital, Chongqing, China
| | - Wei Chen
- Department of Hepatobiliary Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiying He
- Department of Urology, West China Hospital of Sichuan University, Chengdu, China
| | - Feng Chen
- Department of Integrated Care Management Center, West China Hospital of Sichuan University, Chengdu, China
| | - Peng Zhang
- Department of Urology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
137
|
Yang X, Liao HY, Zhang HH. Roles of MET in human cancer. Clin Chim Acta 2021; 525:69-83. [PMID: 34951962 DOI: 10.1016/j.cca.2021.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/18/2023]
Abstract
The MET proto-oncogene was first identified in osteosarcoma cells exposed to carcinogens. Although expressed in many normal cells, MET is overexpressed in many human cancers. MET is involved in the initiation and development of various human cancers and mediates proliferation, migration and invasion. Accordingly, MET has been successfully used as a biomarker for diagnosis and prognosis, survival, post-operative recurrence, risk assessment and pathologic grading, as well as a therapeutic target. In addition, recent work indicates that inhibition of MET expression and function has potential clinical benefit. This review summarizes the role, mechanism, and clinical significance of MET in the formation and development of human cancer.
Collapse
Affiliation(s)
- Xin Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
138
|
Fu W, Zhao H, Liu Y, Nie H, Gao B, Yin F, Wang B, Li T, Zhang T, Wang L, Wu X, Zhu M, Xia L. Exosomes Derived from Cancer-Associated Fibroblasts Regulate Cell Progression in Clear-Cell Renal-Cell Carcinoma. Nephron Clin Pract 2021; 146:383-392. [PMID: 34903693 DOI: 10.1159/000520304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/18/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUNDS Exosomes from multiple sources function as regulatory factors in progression of various tumors. However, studies on the impact of exosomes from cancer-associated fibroblasts (CAFs) on tumor-cell proliferation, migration, invasion, and cycle regulation in clear-cell renal-cell carcinoma (ccRCC) are still lacking. METHODS A Western blot assay was performed to test the exosome-related marker protein level in exosomes derived from CAFs and normal fibroblasts (NFs). A confocal microscope was utilized to observe the internalization of CAF- and NF-derived exosomes after coculturing with cancer cells. MTT, EdU, colony formation, and transwell assays were conducted to detect progression of cancer cells incubated with CAF-derived exosomes. A Western blot assay was also conducted to test expression levels of metastasis-associated proteins. Changes in cell apoptosis and cell cycle were measured by flow cytometry. RESULTS Expression of CAF-derived exosome-related marker proteins was higher than that from NFs. Exosomes derived from CAFs and NFs could enter into cancer cells smoothly and be internalized by cancer cells. After cancer cells were cocultured with CAF-derived exosomes, cell proliferation, migration, and invasion were notably enhanced, and cell apoptosis was reduced. Moreover, expression of fibronectin, N-cadherin, vimentin, MMP9, and MMP2 in cancer cells increased, while E-cadherin was decreased. Besides, the proportion of cancer cells in the S phase increased. CONCLUSION CAF-derived exosomes are internalized into ccRCC cells and promote the progression of ccRCC.
Collapse
Affiliation(s)
- Wenqiang Fu
- Department of Urology Surgery, Tangshan Central Hospital, Tangshan, China
| | - Haihong Zhao
- Department of Hemodialysis, Tangshan Central Hospital, Tangshan, China
| | - Yifei Liu
- Department of Urology, Tangshan Central Hospital, Tangshan, China
| | - Honglin Nie
- Department of Health Management, Tangshan Central Hospital, Tangshan, China
| | - Bin Gao
- Department of Urology, Tangshan Central Hospital, Tangshan, China,
| | - Feng Yin
- Department of Urology, Tangshan Central Hospital, Tangshan, China
| | - Baocun Wang
- Department of Urology, Tangshan Central Hospital, Tangshan, China
| | - Tengfei Li
- Department of Urology, Tangshan Central Hospital, Tangshan, China
| | - Tingting Zhang
- Department of Urology, Tangshan Central Hospital, Tangshan, China
| | - Lijuan Wang
- Department of Urology, Tangshan Central Hospital, Tangshan, China
| | - Xiaotang Wu
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China
| | - Mengjiao Zhu
- Shanghai Engineering Research Center of Pharmaceutical Translation, Shanghai, China
| | - Lianjie Xia
- Department of Urology Surgery, Tangshan Central Hospital, Tangshan, China
| |
Collapse
|
139
|
Ged Y, Voss MH. Novel emerging biomarkers to immunotherapy in kidney cancer. Ther Adv Med Oncol 2021; 13:17588359211059367. [PMID: 34868351 PMCID: PMC8640284 DOI: 10.1177/17588359211059367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022] Open
Abstract
The treatment of metastatic renal cell carcinoma has significantly evolved in recent years, particularly with the advent of novel immune checkpoint inhibitors (ICI). Despite the striking benefits observed on a population level, outcomes vary and some patients do not respond to ICI-based regimens, ultimately require salvage therapies. An ever deeper understanding of the disease biology mediated by the development of multiple high-throughput molecular omics has led to significant progress in biomarkers discovery. But despite growing insights into the molecular underpinnings of the tumor microenvironment, biomarkers have not been integrated successfully into clinical practice. In this review, we discuss some of the novel emerging predictive biomarkers to ICIs in metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Yasser Ged
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA
| |
Collapse
|
140
|
Ma H, Tan Y, Wen D, Qu N, Kong Q, Li K, Ma S, Zhang J. DC-CTL targeting carbonic anhydrase IX gene combined with iAPA therapy in the treatment of renal cell carcinoma. Hum Vaccin Immunother 2021; 17:4363-4373. [PMID: 34851805 DOI: 10.1080/21645515.2021.1955610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
INTRODUCTION To deliver specific antigens in tumor immunotherapy, tumor cell lysates are commonly used to sensitize dendritic cells (DCs). However, the lysates possess low immunogenicity and contain many types of non-tumor-related antigens, which may induce autoimmune diseases. Tumor antigen peptides can provide high specificity but are expensive and their short half-lives limit their clinical application. METHODS In this study, we used adenovirus to transfer the carbonic anhydrase IX (CA9) gene into DCs to generate specificity to renal cell carcinoma (RCC) which is the most common space-occupying lesion in humans. Inhibition of antigen presentation attenuators (iAPA) technology was also used to enhance the DC delivery capacity. Finally, DCs were co-cultured with cytotoxic T-lymphocytes (CTLs) and the anti-tumor effects were evaluated. RESULTS The results showed that the CA9-DC-CTLs possessed a high specificity to CA9-positive cells and showed stronger anti-tumor activity than GFP-DC-CTLs both in vitro and in vivo. DISCUSSION These findings may suggest a novel treatment option for RCC.
Collapse
Affiliation(s)
- Heran Ma
- Qilu Cell Therapy Technology Co., Ltd, Jinan, China.,Shandong Yinfeng Life Science Research Institute, Jinan, China
| | - Yi Tan
- Qilu Cell Therapy Technology Co., Ltd, Jinan, China.,Shandong Yinfeng Life Science Research Institute, Jinan, China
| | - Dingke Wen
- Qilu Cell Therapy Technology Co., Ltd, Jinan, China
| | - Na Qu
- Qilu Cell Therapy Technology Co., Ltd, Jinan, China
| | - Qunfang Kong
- Qilu Cell Therapy Technology Co., Ltd, Jinan, China
| | - Kun Li
- Department of Digestion, First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Suxia Ma
- Heze Municipal Hospital, Heze, Shandong, China
| | | |
Collapse
|
141
|
Assouad E, El Hage S, Safi S, El Kareh A, Mokled E, Salameh P. Kidney cancer trends and risk factors in Lebanon: a 12-year epidemiological study. Cancer Causes Control 2021; 33:303-312. [PMID: 34839395 DOI: 10.1007/s10552-021-01525-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 11/15/2021] [Indexed: 11/27/2022]
Abstract
PURPOSE Lebanon has witnessed an increase in the rates of several cancer subtypes over the last years. The aim of this study is to examine the incidence rates of kidney cancer in Lebanon over 12 years and to compare them to other countries. METHODS Data were collected from the Lebanese National Cancer Registry (NCR) for the time period 2005-2016 (inclusive). Data from other countries were retrieved from an online database "Cancer Incidence in Five Continents." The age specific and age-standardized rates (ASR) were calculated and analyzed using Joinpoint regression. RESULTS Kidney cancer ranked as the 10th commonest cancer among men and the 19th among women. An age-standardized rate of 3.54 (per 100,000) was obtained. The average ASR was 4.80 for men and 2.27 for women. Kidney cancer showed a significantly rising trend for both genders. Lebanon had the highest ASR for kidney cancer in men and the third highest in women among regional countries. CONCLUSION Lebanon presented an average-to-high ASR for kidney cancer compared to regional countries. However, compared to countries worldwide, Lebanon had a below-average ASR. Nonetheless, with the rising kidney cancer trends, it is important to study the associated risk factors in order to develop proper preventive and screening measures and therefore decrease the incidence rates.
Collapse
Affiliation(s)
- Elise Assouad
- Faculty of Medicine, Lebanese American University, Byblos, Lebanon
| | - Said El Hage
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- INSPECT-LB (Institut National de Santé Publique, Epidémiologie Clinique et Toxicologie), Beirut, Lebanon
| | - Steven Safi
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon.
| | - Antonio El Kareh
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Elie Mokled
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Pascale Salameh
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
- Faculty of Pharmacy, Lebanese University, Hadath, Lebanon
- INSPECT-LB (Institut National de Santé Publique, Epidémiologie Clinique et Toxicologie), Beirut, Lebanon
| |
Collapse
|
142
|
piRNA-31115 Promotes Cell Proliferation and Invasion via PI3K/AKT Pathway in Clear Cell Renal Carcinoma. DISEASE MARKERS 2021; 2021:6915329. [PMID: 34790278 PMCID: PMC8592738 DOI: 10.1155/2021/6915329] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 01/01/2023]
Abstract
PIWI-interacting RNAs (piRNAs) are small noncoding RNAs that play important roles in germline development and carcinogenesis. In this study, we used the deep sequencing of small RNA Transcriptome to explore the piRNA expression in six clear cell renal carcinoma (ccRCC) tissues and matched adjacent normal tissues and found that six piRNAs were upregulated and sixteen were downregulated in ccRCC tissues. Among them, piRNA-31115 (NCBI accession number: DQ571003) was the most upregulated piRNA in ccRCC tissues compared with matched adjacent normal tissues. Quantitative real-time PCR (qRT-PCR) was used to confirm piR-31115 expression in other ccRCC tissues (n = 40) and ccRCC cell lines. Besides, function analysis demonstrated that silencing of piR-31115 inhibited ccRCC cell proliferation, motility, and invasiveness. Mechanistic investigations showed that piRNA-31115 may activate epithelial-mesenchymal transition (EMT) via the PI3K/AKT signaling pathway. Hence, piR-31115 may represent an oncogene in the development of ccRCC.
Collapse
|
143
|
Sueyoshi K, Komura D, Katoh H, Yamamoto A, Onoyama T, Chijiwa T, Isagawa T, Tanaka M, Suemizu H, Nakamura M, Miyagi Y, Aburatani H, Ishikawa S. Multi-tumor analysis of cancer-stroma interactomes of patient-derived xenografts unveils the unique homeostatic process in renal cell carcinomas. iScience 2021; 24:103322. [PMID: 35079698 PMCID: PMC8767947 DOI: 10.1016/j.isci.2021.103322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 06/22/2021] [Accepted: 10/19/2021] [Indexed: 12/22/2022] Open
Abstract
The patient-derived xenograft (PDX) model is a versatile tool used to study the tumor microenvironment (TME). However, limited studies have described multi-tumor PDX screening strategies to detect hub regulators during cancer-stroma interaction. Transcriptomes of cancer (human) and stroma (mouse) components of 70 PDX samples comprising 9 distinctive tumor types were analyzed in this study. PDX models recapitulated the original tumors' features, including tumor composition and putative signaling. Particularly, kidney renal clear cell carcinoma (KIRC) stood out, with altered hypoxia-related pathways and a high proportion of endothelial cells in the TME. Furthermore, an integrated analysis conducted to predict paracrine effectors in the KIRC cancer-to-stroma communication detected well-established soluble factors responsible for the hypoxia-related reaction and the so-far unestablished soluble factor, apelin (APLN). Subsequent experiments also supported the potential role of APLN in KIRC tumor progression. Therefore, this paper hereby provides an analytical workflow to find hub regulators in cancer-stroma interactions.
Collapse
Affiliation(s)
- Kuniyo Sueyoshi
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Experimental Research Buliding, 12Floor, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
- Department of Thoracic Surgery, Tokyo Medical and Dental University, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Experimental Research Buliding, 12Floor, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Experimental Research Buliding, 12Floor, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Asami Yamamoto
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Experimental Research Buliding, 12Floor, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Takumi Onoyama
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Experimental Research Buliding, 12Floor, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Tottori 683-8504, Japan
| | - Tsuyoshi Chijiwa
- Central Institute for Experimental Animals, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210–0821, Japan
| | - Takayuki Isagawa
- Data Science Center, Jichi Medical University, Yakushiji, Shimotsuke-shi, Tochigi 329–0498, Japan
| | - Mariko Tanaka
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113–8654, Japan
| | - Hiroshi Suemizu
- Central Institute for Experimental Animals, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210–0821, Japan
| | - Masato Nakamura
- Department of Regenerative Medicine, Tokai University School of Medicine, Shimokasuya, Isehara, Kanagawa 259–1193, Japan
| | - Yohei Miyagi
- Research Institute, Kanagawa Cancer Center, Nakao, Asahi-ku, Yokohama 241–8515, Japan
| | - Hiroyuki Aburatani
- Division of Genome Sciences, RCAST, The University of Tokyo, Tokyo 113–8654, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Experimental Research Buliding, 12Floor, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| |
Collapse
|
144
|
Qi X, Li Q, Che X, Wang Q, Wu G. The Uniqueness of Clear Cell Renal Cell Carcinoma: Summary of the Process and Abnormality of Glucose Metabolism and Lipid Metabolism in ccRCC. Front Oncol 2021; 11:727778. [PMID: 34604067 PMCID: PMC8479096 DOI: 10.3389/fonc.2021.727778] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 12/27/2022] Open
Abstract
Kidney cancer is a cancer with an increasing incidence in recent years. Clear cell renal cell carcinoma (ccRCC) accounts for up to 80% of all kidney cancers. The understanding of the pathogenesis, tumor progression, and metastasis of renal carcinoma is not yet perfect. Kidney cancer has some characteristics that distinguish it from other cancers, and the metabolic aspect is the most obvious. The specificity of glucose and lipid metabolism in kidney cancer cells has also led to its being studied as a metabolic disease. As the most common type of kidney cancer, ccRCC has many characteristics that represent the specificity of kidney cancer. There are features that we are very concerned about, including the presence of lipid droplets in cells and the obesity paradox. These two points are closely related to glucose metabolism and lipid metabolism. Therefore, we hope to explore whether metabolic changes affect the occurrence and development of kidney cancer by looking for evidence of changes on expression at the genomic and protein levels in glucose metabolism and lipid metabolism in ccRCC. We begin with the representative phenomenon of abnormal cancer metabolism: the Warburg effect, through the collection of popular metabolic pathways and related genes in the last decade, as well as some research hotspots, including the role of ferroptosis and glutamine in cancer, systematically elaborated the factors affecting the incidence and metastasis of kidney cancer. This review also identifies the similarities and differences between kidney cancer and other cancers in order to lay a theoretical foundation and provide a valid hypothesis for future research.
Collapse
Affiliation(s)
- Xiaochen Qi
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Quanlin Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qifei Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
145
|
RHBDD1 promotes proliferation, migration, invasion and EMT in renal cell carcinoma via the EGFR/AKT signaling pathway. Mol Med Rep 2021; 24:826. [PMID: 34581421 PMCID: PMC8503741 DOI: 10.3892/mmr.2021.12466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/12/2021] [Indexed: 12/20/2022] Open
Abstract
Renal cell carcinoma (RCC) is a common malignant tumor of the urinary system with a poor prognosis and high mortality rate. The increasing incidence of RCC poses a serious threat to human health. It is well-documented that rhomboid domain-containing protein 1 (RHBDD1) plays a vital role in cancer progression. The present study was designed to identify the biological functions of RHBDD1 in RCC and investigate the underlying regulatory mechanism, aiming to explore the novel molecular therapeutic targets for RCC. The protein and mRNA expression levels of RHBDD1 in normal renal tubule epithelium and human RCC cell lines were analyzed using western blotting and reverse transcription-quantitative PCR. Cell proliferation was determined using Cell Counting Kit-8 assays. Wound healing and Transwell assays were performed to determine cell migration and invasion, respectively. In addition, key proteins related to migration, invasion and epithelial-mesenchymal transition (EMT), such as matrix metalloproteinase (MMP)2, MMP9, MMP13, E-cadherin, N-cadherin, vimentin and Slug, were analyzed using western blotting. In addition, the EGFR/AKT signaling pathway was further studied using western blotting to determine the potential molecular mechanism. The results of the present study revealed that RHBDD1 expression levels were significantly upregulated in RCC cell lines. The knockdown of RHBDD1 inhibited cell proliferation, migration, invasion and EMT, while the overexpression of RHBDD1 promoted cell proliferation, migration, invasion and EMT in RCC. In addition, the knockdown of RHBDD1 suppressed the activation of the EGFR/AKT signaling pathway, while the overexpression of RHBDD1 activated the EGFR/AKT signaling pathway. Moreover, these stimulatory effects of RHBDD1 overexpression on RCC progression and the EGFR/AKT signaling pathway were partly reversed by gefitinib, an EGFR inhibitor. In conclusion, the findings of the present study suggested that RHBDD1 may be a crucial regulator of RCC by modulating the EGFR/AKT signaling pathway. The present study may provide a theoretical basis and potential targets for RCC treatment.
Collapse
|
146
|
Sun H, Li J, Hu W, Yan Y, Guo Z, Zhang Z, Chen Y, Yao X, Teng L, Wang X, Li L, Chai D, Zheng J, Wang G. Co-immunizing with HMGB1 enhances anti-tumor immunity of B7H3 vaccine in renal carcinoma. Mol Immunol 2021; 139:184-192. [PMID: 34560414 DOI: 10.1016/j.molimm.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/16/2021] [Accepted: 09/02/2021] [Indexed: 11/27/2022]
Abstract
Metastatic renal carcinoma is a kind of tumor with high degree of malignancy, but there are no effective treatment methods and strategies at present. In this study, we designed a folate-grafted PEI600-CyD (H1) nanoparticle-mediated DNA vaccine containing an adjuvant of high mobility group box 1 protein (HMGB1) and a tumor-specific antigen of B7H3 (CD276) for renal carcinoma therapy. Mice bearing subcutaneous human B7H3 (hB7H3)-Renca tumor were immunized with H1-pHMGB1/pB7H3, H1-pB7H3, H1-pHMGB1, or Mock vaccine. Compared to other control groups, the growth of the tumor was significantly inhibited in H1-pHMGB1/pB7H3 vaccine group. The increased proportion and mature of CD11c+ DCs were observed in the spleen of H1-pHMGB1/pB7H3 treated mice. Likewise, HMGB1 promoted B7H3 vaccine to induce tumor-specific CD8+ T cell proliferation and CTL responses. Beyond that, H1-pHMGB1/pB7H3 vaccine strengthened the induction of functional CD8+ T cells. With the depletion of CD8+ T cells, the anti-tumor effect of H1-pHMGB1/pB7H3 also disappeared, indicating that CD8+ T cells are the key factor of the anti-tumor activity of the vaccine. So, to sum up, H1-pHMGB1/pB7H3 vaccine could achieve the desired anti-tumor effect by enhancing the response of tumor-specific functional CD8+ T cell responses. H1 nanoparticle-based vaccines may have great potential and prospect in the treatment of primary solid tumors.
Collapse
Affiliation(s)
- Huanyou Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Juan Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Wenwen Hu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Yinan Yan
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Zengli Guo
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Zichun Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Xuefan Yao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Ling Teng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Xinyuan Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Liantao Li
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, PR China.
| |
Collapse
|
147
|
Tobe A, Tanaka A, Yoshida S, Kondo T, Morimoto R, Furusawa K, Okumura T, K Bando Y, Ishii H, Murohara T. High-output Heart Failure Caused by a Tumor-related Arteriovenous Fistula: A Case Report and Literature Review. Intern Med 2021; 60:2979-2984. [PMID: 33776013 PMCID: PMC8502649 DOI: 10.2169/internalmedicine.6962-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
High-output heart failure caused by a tumor-related arteriovenous fistula in adults is a rare clinical condition. We herein report a case of high-output heart failure caused by an arteriovenous fistula associated with renal cell carcinoma and a literature review of 29 published cases to date. Renal cell carcinoma seems to be the most common underlying tumor. For the diagnosis, right heart catheterization and enhanced computed tomography (CT) are considered useful. The removal of the underlying tumor and arteriovenous fistula is the best treatment for heart failure.
Collapse
Affiliation(s)
- Akihiro Tobe
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Akihito Tanaka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Satoya Yoshida
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Toru Kondo
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Ryota Morimoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Kenji Furusawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Yasuko K Bando
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| | - Hideki Ishii
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
- Department of Cardiology, Fujita Health University Bantane Hospital, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
148
|
Circular RNA FOXP1 Induced by ZNF263 Upregulates U2AF2 Expression to Accelerate Renal Cell Carcinoma Tumorigenesis and Warburg Effect through Sponging miR-423-5p. J Immunol Res 2021; 2021:8050993. [PMID: 34514002 PMCID: PMC8433034 DOI: 10.1155/2021/8050993] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/11/2021] [Indexed: 01/17/2023] Open
Abstract
Renal cell carcinoma (RCC), as one of the most common malignant tumors in the urinary system, is featured with high morbidity and mortality. Although the improvement of clinical intervention, such as surgery technology, chemotherapy, and radiotherapy, has been made, the outcomes of RCC patients are still poor. Novel targets for RCC treatment are urgently needed. Recently, circRNA has been in-depth studied and is considered as a promising direction for gene target therapy. In this study, we explored the function of circFOXP1 in RCC progression and its underlying mechanisms. Firstly, we demonstrated the characterization and expression of circFOXP1 in RCC tissues and cells. Next, by conducting a serial experiment, we found that downregulated circFOXP1 inhibited cell proliferation, migration, invasion, and the Warburg effect. Next, our experiments found that circFOXP1 upregulated U2AF2 expression via sponging miR-423-5p in RCC cells. Moreover, we found that ZNF263 induced circFOXP1 expression in RCC cells. To sum up, our study partially demonstrated that the novel ZNF263/circFOXP1/miR-423-5p/U2AF2 axis has a role in RCC progression. Our results might provide a new direction for RCC therapeutic target exploring.
Collapse
|
149
|
Effect of Aberrant Long Noncoding RNA on the Prognosis of Clear Cell Renal Cell Carcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6533049. [PMID: 34512796 PMCID: PMC8433025 DOI: 10.1155/2021/6533049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/07/2021] [Indexed: 11/17/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a kind of lethal cancer. Although there are mature treatment methods, there is still a lack of rigorous and scientific means for cancer diagnosis. Long noncoding RNAs (lncRNAs) are a kind of noncoding RNA (ncRNA). Recent studies find that alteration of lncRNA expression is related to the occurrence of many cancers. In order to find lncRNAs which can effectively predict the prognosis of ccRCC, RNA-seq count data and clinical information were downloaded from TCGA-KIRC, and gene expression profiles from 530 patients were included. Then, K-means was used for clustering, and the number of clusters was determined to be 5. The R-package "edgeR" was used to perform differential expression analysis. Subsequently, a risk model composed of 10 lncRNA biomarkers significantly related to prognosis was identified via Cox and LASSO regression analyses. Then, patients were divided into two groups according to the model-based risk score, and then, GSEA pathway enrichment was performed. The results showed that metabolism- and mTOR-related pathways were activated while immune-related pathways were inhibited in the high-risk patients. Combined with previous studies, it is believed that these 10 lncRNAs are potential targets for the treatment of ccRCC. In addition, Cox regression analysis was used to verify the independence of the risk model, and as results revealed, the risk model can be used to independently predict the prognosis of patients. In conclusion, our study found 10 lncRNAs related to the prognosis of ccRCC and provided new ideas for clinical diagnosis and drug development.
Collapse
|
150
|
Prognostic and Therapeutic Potential of the OIP5 Network in Papillary Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13174483. [PMID: 34503297 PMCID: PMC8431695 DOI: 10.3390/cancers13174483] [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: 08/01/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Papillary renal cell carcinoma (pRCC) is an aggressive but minor type of RCC. The current understanding and management of pRCC remain poor. We report here OIP5 being a novel oncogenic factor and possessing robust prognostic values and therapeutic potential. OIP5 upregulation is observed in pRCC. The upregulation is associated with pRCC adverse features (T1P < T2P < CIMP, Stage1 + 2 < Stage 3 < Stage 4, and N0 < N1) and effectively stratifies the fatality risk. OIP5 promotes ACHN pRCC cell proliferation and xenograft formation; the latter is correlated with network alterations related to immune regulation, metabolism, and hypoxia. A set of differentially expressed genes (DEFs) was derived from ACHN OIP5 xenografts and primary pRCCs (n = 282) contingent to OIP5 upregulation; both DEG sets share 66 overlap genes. Overlap66 effectively predicts overall survival (p < 2 × 10-16) and relapse (p < 2 × 10-16) possibilities. High-risk tumors stratified by Overlap66 risk score possess an immune suppressive environment, evident by elevations in Treg cells and PD1 in CD8 T cells. Upregulation of PLK1 occurs in both xenografts and primary pRCC tumors with OIP5 elevations. PLK1 displays a synthetic lethality relationship with OIP5. PLK1 inhibitor BI2356 inhibits the growth of xenografts formed by ACHN OIP5 cells. Collectively, the OIP5 network can be explored for personalized therapies in management of pRCC patients.
Collapse
|