1
|
Yue C, Zheng X, Ye Y, Li D, Zhou Y. TNFAIP8L3 regulation of the TGF-β signaling pathway affects the proportion of macrophages during tumor antigen presentation and affects the prognosis of ovarian cancer. Front Mol Biosci 2025; 12:1566363. [PMID: 40343258 PMCID: PMC12058474 DOI: 10.3389/fmolb.2025.1566363] [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: 01/24/2025] [Accepted: 03/13/2025] [Indexed: 05/11/2025] Open
Abstract
Background Ovarian cancer is a serious disease that is a danger to a woman's life and health and is currently being extensively studied worldwide. TNFAIP8L3, a member of the tumor necrosis family, plays a significant role in tumor immune regulation; however, its role in ovarian cancer has not been fully studied and reported. Method This study used data from 381 cases of ovarian cancer (OVCA) from The Cancer Genome Analysis (TCGA)-OV to analyze the clinical phenotype and immune phenotype of TNFAIP8L3. Pearson correlation coefficients and protein-protein interaction (PPI) network analyses were used to identify potential biological functions and the genes co-expressed with TNFAIP8L3. The Gene Set Enrichment Analysis (GSEA) method was used to evaluate the relevant regulatory pathways of TNFAIP8L3. Gene Set Variation Analysis was performed to evaluate the proportions of 24 immune cell types in OVCA. Finally, a nomogram and multivariate COX regression analysis were performed to prove the independent prognostic value of TNFAIP8L3 in OVCA. Results A total of 181 genes were co-expressed with TNFAIP8L3, including 163 positively correlated and 18 negatively correlated genes. The co-expressed genes associated with TNFAIP8L3 were significantly enriched in biological processes such as cell activation involved in immune response and leukocyte activation involved in immune response. Pathway analysis of TNFAIP8L3 further revealed the association of TNFAIP8L3 with the TGF-β signaling pathway and antigen processing and presentation pathways, especially with macrophages and neutrophils in the antigen presentation process. Moreover, the expression of TNFAIP8L3 was significantly high in OVCA and other solid tumors to function as an independent risk factor for the prognosis of OVCA, with important research value for the prognosis of patients with OVCA. Conclusion Those findings indicate that TNFAIP8L3 is correlated with antigen processing and presentation pathways in macrophages and neutrophils and affects prognosis and immune infiltration in OVCA.
Collapse
Affiliation(s)
- Chaomin Yue
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiao Zheng
- Department of General Surgery, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing Ye
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, United Kingdom
| | - Danyang Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yanhong Zhou
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Yin W, Chen G, Li Y, Li R, Jia Z, Zhong C, Wang S, Mao X, Cai Z, Deng J, Zhong W, Pan B, Lu J. Identification of a 9-gene signature to enhance biochemical recurrence prediction in primary prostate cancer: A benchmarking study using ten machine learning methods and twelve patient cohorts. Cancer Lett 2024; 588:216739. [PMID: 38395379 DOI: 10.1016/j.canlet.2024.216739] [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: 08/28/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
Prostate cancer (PCa) is a prevalent malignancy among men worldwide, and biochemical recurrence (BCR) after radical prostatectomy (RP) is a critical turning point commonly used to guide the development of treatment strategies for primary PCa. However, the clinical parameters currently in use are inadequate for precise risk stratification and informing treatment choice. To address this issue, we conducted a study that collected transcriptomic data and clinical information from 1662 primary PCa patients across 12 multicenter cohorts globally. We leveraged 101 algorithm combinations that consisted of 10 machine learning methods to develop and validate a 9-gene signature, named BCR SCR, for predicting the risk of BCR after RP. Our results demonstrated that BCR SCR generally outperformed 102 published prognostic signatures. We further established the clinical significance of these nine genes in PCa progression at the protein level through immunohistochemistry on Tissue Microarray (TMA). Moreover, our data showed that patients with higher BCR SCR tended to have higher rates of BCR and distant metastasis after radical radiotherapy. Through drug target prediction analysis, we identified nine potential therapeutic agents for patients with high BCR SCR. In conclusion, the newly developed BCR SCR has significant translational potential in accurately stratifying the risk of patients who undergo RP, monitoring treatment courses, and developing new therapies for the disease.
Collapse
Affiliation(s)
- Wenjun Yin
- Department of Andrology, Guangzhou First People's Hospital, South China University of Technology, 510180, Guangzhou, Guangdong, China; Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yutong Li
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Ruidong Li
- Genetics, Genomics, and Bioinformatics Program, University of California, Riverside, CA, 92521, USA
| | - Zhenyu Jia
- Department of Botany and Plant Sciences, University of California, Riverside, CA, 92521, USA
| | - Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Shuo Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, China
| | - Zhouda Cai
- Department of Andrology, Guangzhou First People's Hospital, South China University of Technology, 510180, Guangzhou, Guangdong, China
| | - Junhong Deng
- Department of Andrology, Guangzhou First People's Hospital, South China University of Technology, 510180, Guangzhou, Guangdong, China
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, 510180, Guangzhou, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, 999078, Macau, China.
| | - Bin Pan
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China.
| | - Jianming Lu
- Department of Andrology, Guangzhou First People's Hospital, South China University of Technology, 510180, Guangzhou, Guangdong, China; Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, 510180, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Chen K, Hong C, Kong W, Li G, Liu Z, Zhu K, Lu C, Si P, Gao P, Ning G, Zhang R. ACADL-YAP axis activity in non-small cell lung cancer carcinogenicity. Cancer Cell Int 2024; 24:86. [PMID: 38402174 PMCID: PMC10894480 DOI: 10.1186/s12935-024-03276-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/17/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND The role of Acyl-CoA dehydrogenase long chain (ACADL) in different tumor types had different inhibiting or promoting effect. However, its role in non-small cell lung cancer (NSCLC) carcinogenicity is not clear. METHOD In this study, we utilized The Cancer Genome Atlas (TCGA) database to analyze ACADL expression in NSCLC and its correlation with overall survival. Furthermore, we investigated the function of ACADL on cellular proliferation, invasion, colony, apoptosis, cell cycle in vitro with NSCLC cells. Mechanistically, we evaluated the regulatory effect of ACADL expression on its downstream factor yes-associated protein (YAP) by assessing YAP phosphorylation levels and its cellular localization. Finally, we verified the tumorigenic effect of ACADL on NSCLC cells through xenograft experiments in vivo. RESULTS Compared to adjacent non-cancerous samples, ACADL significantly down-regulated in NSCLC. Overexpression of ACADL, effectively reduced the proliferative, colony, and invasive capabilities of NSCLC cells, while promoting apoptosis and inducing cell cycle arrest. Moreover, ACADL overexpression significantly enhanced YAP phosphorylation and hindered its nuclear translocation. However, the inhibitory effect of the overexpression of ACADL in NSCLC cells mentioned above can be partially counteracted by YAP activator XMU-MP-1 application both in vitro and in vivo. CONCLUSION The findings suggest that ACADL overexpression could suppress NSCLC development by modulating YAP phosphorylation and limiting its nuclear shift. This role of ACADL-YAP axis provided novel insights into NSCLC carcinogenicity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Kegong Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, 230001, China
| | - Chunqiao Hong
- Department of Critical Care Medicine, Xiamen Chang Gung Hospital Hua Qiao University, Xiamen, 361013, China
| | - Weibo Kong
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
- Departments of Thoracic Surgery, Anhui Provincial Chest Hospital, Hefei, 230001, China
| | - Guanghua Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Zhuang Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230001, China
| | - Kechao Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
| | - Chen Lu
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
| | - Panpan Si
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China
| | - Pan Gao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230001, China
| | - Guangyao Ning
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China.
| | - Renquan Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230001, China.
| |
Collapse
|
4
|
CircPIM3 regulates taxol resistance in non-small cell lung cancer via miR-338-3p/TNFAIP8 axis. Anticancer Drugs 2023; 34:115-125. [PMID: 36539365 DOI: 10.1097/cad.0000000000001347] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Numerous work has revealed the involvement of circular RNA (circRNA) in regulating chemotherapy resistance. Here, we investigate circPIM3 role in taxol (Tax) resistance in non-small cell lung cancer (NSCLC). CircPIM3, microRNA (miR)-338-3p and tumor necrosis factor-alpha-induced protein-8 (TNFAIP8) expression were detected via quantitative real-time PCR, western blot or immunohistochemistry assay. Tax resistance was evaluated using cell counting kit-8, cell proliferation was measured by colony formation assay, cell cycle and apoptosis were examined via flow cytometry. The interplay between miR-338-3p and circPIM3 or TNFAIP8 was confirmed by dual-luciferase reporter assay. Finally, the effect of circPIM3 on Tax resistance in NSCLC in vivo was investigated by xenograft models. CircPIM3 and TNFAIP8 were upregulated in Tax-resistant NSCLC tissue and cell samples. Reducing circPIM3 expression inhibited Tax resistance, proliferation and induced cycle arrest and apoptosis in Tax-resistant NSCLC cells. Mechanically, circPIM3 absence led to downregulation of TNFAIP8 via absorbing miR-338-3p. Additionally, circPIM3 depletion increased Tax sensitivity of NSCLC in vivo. Silencing of circPIM3 suppressed Tax resistance in Tax-resistant NSCLC cells through regulation of the miR-338-3p/TNFAIP8 axis.
Collapse
|
5
|
Wang Z, Sun L, Liu S, Jiang H. TIPE1 inhibits the growth of Ewing’s sarcoma cells by suppressing Wnt/β-catenin signaling. Clin Transl Oncol 2022; 25:1332-1339. [PMID: 36495466 DOI: 10.1007/s12094-022-03030-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/27/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Ewing's sarcoma is the second most common bone and soft tissue malignancy in children and adolescents. Tumor necrosis factor-α-induced protein 8-like 1 (TIPE1) functions as a tumor suppressor in several cancers. Activation of Wnt/β-catenin signaling in subpopulations of tumor cells contributes to phenotypic heterogeneity and disease progression in Ewing's sarcoma. The exact role of TIPE1 in Ewing's sarcoma remains to be elucidated. PURPOSE This study aimed to assess the expression and function of TIPE1 in Ewing's sarcoma. METHODS TIPE1 expression in Ewing's sarcoma cells was determined by qPCR and western blotting. Furthermore, the Ewing's sarcoma cell line RD-ES was transfected with a lentivirus-based TIPE1 expression system to upregulate the expression of TIPE1. The Cell Counting Kit 8 was used to assess the effect of TIPE1 on cell proliferation. The effects of TIPE1 on cell migration and invasion was detected by Transwell assay. Flow cytometry was performed to detect apoptosis. RESULTS Our results suggested lower TIPE1 expression in Ewing's sarcoma cell lines compared with normal osseous cells. TIPE1 remarkably inhibited the growth and proliferation of Ewing's sarcoma cell; TIPE1 also induced apoptosis and inhibited invasion in vitro. TIPE1 inhibited Ewing's sarcoma growth, motility, and survival through regulation of Wnt/β-catenin signaling. CONCLUSIONS Our results demonstrated the anti-tumor function of TIPE1 in Ewing's sarcoma and reveal a novel therapeutic target.
Collapse
|
6
|
Ueda Y, Miura Y, Tomishige N, Sugimoto N, Murase M, Kawamura G, Sasaki N, Ishiwata T, Ozawa T. Mechanistic insights into cancer drug resistance through optogenetic PI3K signaling hyperactivation. Cell Chem Biol 2022; 29:1576-1587.e5. [PMID: 36288730 DOI: 10.1016/j.chembiol.2022.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/26/2022] [Accepted: 09/30/2022] [Indexed: 01/31/2023]
Abstract
Hyperactivation of phosphatidylinositol 3-kinase (PI3K) signaling is a prominent feature in cancer cells. However, the mechanism underlying malignant behaviors in the state remains unknown. Here, we describe a mechanism of cancer drug resistance through the protein synthesis pathway, downstream of PI3K signaling. An optogenetic tool (named PPAP2) controlling PI3K signaling was developed. Melanoma cells stably expressing PPAP2 (A375-PPAP2) acquired resistance to a cancer drug in the hyperactivation state. Proteome analyses revealed that expression of the antiapoptotic factor tumor necrosis factor alpha-induced protein 8 (TNFAIP8) was upregulated. TNFAIP8 upregulation was mediated by protein translation from preexisting mRNA. These results suggest that cancer cells escape death via upregulation of TNFAIP8 expression from preexisting mRNA even though alkylating cancer drugs damage DNA.
Collapse
Affiliation(s)
- Yoshibumi Ueda
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan.
| | - Yuri Miura
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | | | - Naotoshi Sugimoto
- Department of Physiology, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Megumi Murase
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
| | - Genki Kawamura
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
| | - Norihiko Sasaki
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Toshiyuki Ishiwata
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
7
|
Ge X, Niture S, Lin M, Cagle P, Li PA, Kumar D. MicroRNA-205-5p inhibits skin cancer cell proliferation and increase drug sensitivity by targeting TNFAIP8. Sci Rep 2021; 11:5660. [PMID: 33707587 PMCID: PMC7952414 DOI: 10.1038/s41598-021-85097-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) is a member of the TIPE/TNFAIP8 family which regulates tumor growth and survival. Our goal is to delineate the detailed oncogenic role of TNFAIP8 in skin cancer development and progression. Here we demonstrated that higher expression of TNFAIP8 is associated with basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma development in patient tissues. Induction of TNFAIP8 expression by TNFα or by ectopic expression of TNFAIP8 in SCC or melanoma cell lines resulted in increased cell growth/proliferation. Conversely, silencing of TNFAIP8 decreased cell survival/cell migration in skin cancer cells. We also showed that miR-205-5p targets the 3'UTR of TNFAIP8 and inhibits TNFAIP8 expression. Moreover, miR-205-5p downregulates TNFAIP8 mediated cellular autophagy, increased sensitivity towards the B-RAFV600E mutant kinase inhibitor vemurafenib, and induced cell apoptosis in melanoma cells. Collectively our data indicate that miR-205-5p acts as a tumor suppressor in skin cancer by targeting TNFAIP8.
Collapse
Affiliation(s)
- Xinhong Ge
- Department of Dermatology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia Hui Autonomous Region, China.,Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, 1801 Fayetteville St., Durham, NC, 27707, USA
| | - Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, 1801 Fayetteville St., Durham, NC, 27707, USA.
| | - Minghui Lin
- Department of Respiratory Diseases, The Forth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750021, Ningxia Hui Autonomous Region, China
| | - Patrice Cagle
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, 1801 Fayetteville St., Durham, NC, 27707, USA
| | - P Andy Li
- Department of Pharmaceutical Sciences, Bio-Manufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC, 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, 1801 Fayetteville St., Durham, NC, 27707, USA.
| |
Collapse
|
8
|
Zhong M, Qiu X, Liu Y, Yang Y, Gu L, Wang C, Chen H, Liu Z, Miao J, Zhuang G. TIPE Regulates DcR3 Expression and Function by Activating the PI3K/AKT Signaling Pathway in CRC. Front Oncol 2021; 10:623048. [PMID: 33718119 PMCID: PMC7943851 DOI: 10.3389/fonc.2020.623048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/23/2020] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor-induced protein-8 (TIPE) is highly expressed in colorectal cancer (CRC). Decoy receptor 3 (DcR3) is a soluble secreted protein that can antagonize Fas ligand (FasL)-induced apoptosis and promote tumorigenesis. It remains unclear whether TIPE can regulate DcR3 expression. In this study, we examined this question by analyzing the relationship between these factors in CRC. Bioinformatics and tissue microarrays were used to determine the expression of TIPE and DcR3 and their correlation in CRC. The expression of TIPE and DcR3 in colon cancer cells was detected. Plasma samples were collected from CRC patients, and DcR3 secretion was measured. Then, dual-luciferase reporter gene analysis was performed to assess the interaction between TIPE and DcR3. We exogenously altered TIPE expression and analyzed its function and influence on DcR3 secretion. Lipopolysaccharide (LPS) was used to stimulate TIPE-overexpressing HCT116 cells, and alterations in signaling pathways were detected. Additionally, inhibitors were used to confirm molecular mechanisms. We found that TIPE and DcR3 were highly expressed in CRC patients and that their expression levels were positively correlated. DcR3 was highly expressed in the plasma of cancer patients. We confirmed that TIPE and DcR3 were highly expressed in HCT116 cells. TIPE overexpression enhanced the transcriptional activity of the DcR3 promoter. TIPE activated the PI3K/AKT signaling pathway to regulate the expression of DcR3, thereby promoting cell proliferation and migration and inhibiting apoptosis. In summary, TIPE and DcR3 are highly expressed in CRC, and both proteins are associated with poor prognosis. TIPE regulates DcR3 expression by activating the PI3K/AKT signaling pathway in CRC, thus promoting cell proliferation and migration and inhibiting apoptosis. These findings may have clinical significance and promise for applications in the treatment or prognostication of CRC.
Collapse
Affiliation(s)
- Mengya Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.,Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Xingfeng Qiu
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Yu Liu
- Department of Gastrointestinal Surgery, Zhongshan Hospital, Xiamen University, Xiamen, China.,General Surgery Center of Bazhong Central Hospital, Bazhong, China
| | - Yan Yang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Lei Gu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Chenxi Wang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Huiyu Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| | - Zhongchen Liu
- Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Jiayin Miao
- Department of Neurology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Guohong Zhuang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.,Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
9
|
Niture S, Lin M, Odera JO, Moore J, Zhe H, Chen X, Suy S, Collins SP, Kumar D. TNFAIP8 drives metabolic reprogramming to promote prostate cancer cell proliferation. Int J Biochem Cell Biol 2021; 130:105885. [PMID: 33227392 PMCID: PMC7770075 DOI: 10.1016/j.biocel.2020.105885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/04/2020] [Accepted: 11/07/2020] [Indexed: 11/20/2022]
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) is a member of TIPE/TNFAIP8 family, has been involved in the development and progression of various human cancers. We hypothesized that TNFAIP8 promotes prostate cancer (PCa) progression via regulation of oxidative phosphorylation (OXPHOS) and glycolysis. Ectopic expression of TNFAIP8 increased PCa cell proliferation/migration/spheroid formation by enhancing cell metabolic activities. Mechanistically, TNFAIP8 activated the PI3K-AKT pathway and up-regulated PCa cell survival. TNFAIP8 was also found to regulate the expression of glucose metabolizing enzymes, enhancing glucose consumption, and endogenous ATP production. Treatment with a glycolysis inhibitor, 2-deoxyglucose (2-DG), reduced TNFAIP8 mediated glucose consumption, ATP production, spheroid formation, and PCa cell migration. By maintaining mitochondrial membrane potential, TNFAIP8 increased OXPHOS and glycolysis. Moreover, TNFAIP8 modulates the production of glycolytic metabolites in PCa cells. Collectively, our data suggest that TNFAIP8 exerts its oncogenic effects by enhancing glucose metabolism and by facilitating metabolic reprogramming in PCa cells. Therefore, TNFAIP8 may be a biomarker associated with prostate cancer and indicate a potential therapeutic target.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Minghui Lin
- Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Joab O Odera
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - John Moore
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Hong Zhe
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004 China
| | - Xiaoxin Chen
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Simeng Suy
- Department of Radiation Medicine, Georgetown University Hospital, WA, DC 20057, USA
| | - Sean P Collins
- Department of Radiation Medicine, Georgetown University Hospital, WA, DC 20057, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA.
| |
Collapse
|
10
|
Zhong M, Chen Z, Yan Y, Bahet A, Cai X, Chen H, Ran H, Qu K, Han Z, Zhuang G, Zhang S, Wang Y. Expression of TIPE family members in human colorectal cancer. Oncol Lett 2020; 21:118. [PMID: 33376549 PMCID: PMC7751461 DOI: 10.3892/ol.2020.12379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 11/12/2020] [Indexed: 12/22/2022] Open
Abstract
The tumor necrosis factor α-induced protein 8 (TNFAIP8)-like (TIPE) protein family comprises four members, namely TNFAIP8, TIPE1, TIPE2 and TIPE3, which are involved in multiple processes in cancer. The current study aimed to investigate the expression patterns and potential clinical roles of the TIPE family members in human colorectal cancer (CRC). Paired tumor and adjacent tissue samples were collected from 49 patients with CRC, and the relative mRNA expression levels of the TIPE family members in these samples were evaluated by using reverse transcription-quantitative PCR, and the protein levels in five randomly selected pairs of tumor and adjacent tissue samples were detected by western blot analysis. The mRNA expression levels of the TIPE family members were significantly downregulated in CRC tumor tissues compared with those in the adjacent tissues; however, within each sample, TNFAIP8 and TIPE3 protein levels were only partially consistent with their mRNA levels. In addition, the mRNA expression levels between each pair of TIPE family members exhibited a positive linear relationship, and TIPE2 mRNA levels exhibited strong linear associations with those of TNFAIP8 and TIPE1. TNFAIP8 mRNA expression levels in tumor tissues were significantly associated with the tumor differentiation grade, and TIPE2 mRNA expression levels in tumor tissues were significantly associated with sex. TIPE1 and TIPE3 mRNA expression levels in tumor tissues exhibited no associations with patient clinicopathological characteristics. In addition, the mRNA expression patterns of the TIPE family members were analyzed using data from The Cancer Genome Atlas data set, and the results also demonstrated that TNFAIP8, TIPE2 and TIPE3 mRNA levels were downregulated in patients with colon adenocarcinoma compared with those in normal controls. These results provided evidence that the four members of the TIPE family may affect each other to mediate the carcinogenesis of CRC, and that TIPE2 may serve an important role in CRC.
Collapse
Affiliation(s)
- Mengya Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Zhijian Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Yang Yan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Argen Bahet
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Xin Cai
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Huiyu Chen
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Honggang Ran
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Kaiyong Qu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Zhaopu Han
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Guohong Zhuang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| | - Shifeng Zhang
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361004, P.R. China.,Institute of Gastrointestinal Oncology, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361004, P.R. China.,Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen, Fujian 361004, P.R. China
| | - Yinan Wang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, Fujian 361005, P.R. China
| |
Collapse
|
11
|
Gu Z, Cui X, Sun P, Wang X. Regulatory Roles of Tumor Necrosis Factor-α-Induced Protein 8 Like-Protein 2 in Inflammation, Immunity and Cancers: A Review. Cancer Manag Res 2020; 12:12735-12746. [PMID: 33364825 PMCID: PMC7751774 DOI: 10.2147/cmar.s283877] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α)-induced protein 8 (TNFAIP8/TIPE) family, including TNFAIP8 (TIPE), TNFAIP8 like-protein 1 (TNFAIP8L1/TIPE1), TNFAIP8 like-protein 2 (TNFAIP8L2/TIPE2), and TNFAIP8 like-protein 3 (TNFAIP8L3/TIPE3), plays a vital role in regulating inflammatory responses, immune homeostasis, and cancer development. Over the last decade, studies have shown that TIPE2 protein is differentially expressed in diverse cells and tissues. The dysregulation of TIPE2 protein can lead to dysregulation of inflammatory responses and immune homeostasis, and change the basic characteristics of cancers. In consideration of the immeasurable values of TIPE2 in diagnosis, treatment, and prognosis of various human diseases, this review will focus on the expression pattern, structure, and regulatory roles of TIPE2 in inflammation, immunity, and cancers.
Collapse
Affiliation(s)
- Zhengzhong Gu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Xiaohan Cui
- Nanjing Medical University, Nanjing, Jiangsu Province, People's Republic of China
| | - Pengda Sun
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| | - Xudong Wang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin Province, People's Republic of China
| |
Collapse
|
12
|
Ma HY, Li Y, Yin HZ, Yin H, Qu YY, Xu QY. TNFAIP8 Promotes Cisplatin Chemoresistance in Triple-Negative Breast Cancer by Repressing p53-Mediated miR-205-5p Expression. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:640-656. [PMID: 33230463 PMCID: PMC7581818 DOI: 10.1016/j.omtn.2020.09.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
Tumor necrosis factor alpha-induced protein 8 (TNFAIP8) is implicated in the tumor progression and prognosis of triple-negative breast cancer (TNBC), but the detailed regulatory mechanism of TNFAIP8 in cisplatin tolerance in TNBC has not yet been investigated. TNFAIP8 was evidently upregulated in TNBC tumor tissues and cell lines. Knocking down TNFAIP8 led to impaired proliferation and elevated apoptosis of TNBC cells upon cisplatin (DDP) treatment. Mechanistic studies revealed that TNFAIP8 repressed the expression of p53 and p53-promoted microRNA (miR)-205-5p; moreover, miR-205-5p targeted multiple genes required for the cell cycle and repressed Akt phosphorylation, which thus inhibited the proliferation of TNBC cells. In addition, silencing of TNFAIP8 led to the upregulation of miR-205-5p and the restraint of the TRAF2-NF-κB pathway, which thus enhanced the suppressive effects of DDP on tumor growth in nude mice. This study revealed that TNFAIP8 was essential in the DDP tolerance formation of TNBC cells by reducing p53-promoted miR-205-5p expression. Thus, targeting TNFAIP8 might become a promising strategy to suppress TNBC progression.
Collapse
Affiliation(s)
- Hong-Yu Ma
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Yang Li
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Hui-Zi Yin
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Hang Yin
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Yuan-Yuan Qu
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| | - Qing-Yong Xu
- Department of Breast Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, P.R. China
| |
Collapse
|
13
|
Bordoloi D, Banik K, Vikkurthi R, Thakur KK, Padmavathi G, Sailo BL, Girisa S, Chinnathambi A, Alahmadi TA, Alharbi SA, Buhrmann C, Shakibaei M, Kunnumakkara AB. Inflection of Akt/mTOR/STAT-3 cascade in TNF-α induced protein 8 mediated human lung carcinogenesis. Life Sci 2020; 262:118475. [PMID: 32976884 DOI: 10.1016/j.lfs.2020.118475] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 02/08/2023]
Abstract
Lung cancer is the leading cause of cancer-related death across the globe. Despite the marked advances in detection and therapeutic approaches, management of lung cancer patients remains a major challenge to oncologists which can be mainly attributed to late stage diagnosis, tumor recurrence and chemoresistance. Therefore, to overthrow these limitations, there arises a vital need to develop effective biomarkers for the successful management of this aggressive cancer type. Notably, TNF-alpha induced protein 8 (TIPE), a nuclear factor-kappa B (NF-κB)-inducible, oncogenic molecule and cytoplasmic protein which is involved in the regulation of T lymphocyte-mediated immunity and different processes in tumor cells such as proliferation, cell death and evasion of growth suppressors, might serve as one such biomarker which would facilitate effective management of lung cancer. Expression studies revealed this protein to be significantly upregulated in different lung cancer types, pathological conditions, stages and grades of lung tumor compared to normal human lung tissues. In addition, knockout of TIPE led to the reduced proliferation, survival, invasion and migration of lung cancer cells. Furthermore, TIPE was found to function through modulation of Akt/mTOR/STAT-3 signaling cascade. This is the first report which shows the involvement of TIPE in tobacco induced lung carcinogenesis. It positively regulated nicotine, NNK, NNN, and BaP induced proliferation, survival and migration of lung cancer cells possibly via Akt/STAT-3 signaling. Thus, this protein possesses important role in the pathogenesis of lung tumor and hence it can be targeted for developing newer therapeutic interventions for the clinico-management of lung cancer.
Collapse
Affiliation(s)
- Devivasha Bordoloi
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| | - Kishore Banik
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Rajesh Vikkurthi
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Ganesan Padmavathi
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Tahani Awad Alahmadi
- Department of Pediatrics, College of Medicine, King Saud University [Medical City], King Khalid University Hospital, PO Box-2925, Riyadh 11461, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Constanze Buhrmann
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mehdi Shakibaei
- Department of Anatomy, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| |
Collapse
|
14
|
Niture S, Gyamfi MA, Lin M, Chimeh U, Dong X, Zheng W, Moore J, Kumar D. TNFAIP8 regulates autophagy, cell steatosis, and promotes hepatocellular carcinoma cell proliferation. Cell Death Dis 2020; 11:178. [PMID: 32152268 PMCID: PMC7062894 DOI: 10.1038/s41419-020-2369-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-α-induced protein 8 (TNFAIP8) expression has been linked to tumor progression in various cancer types, but the detailed mechanisms of TNFAIP8 are not fully elucidated. Here we define the role of TNFAIP8 in early events associated with development of hepatocellular carcinoma (HCC). Increased TNFAIP8 levels in HCC cells enhanced cell survival by blocking apoptosis, rendering HCC cells more resistant to the anticancer drugs, sorafenib and regorafenib. TNFAIP8 also induced autophagy and steatosis in liver cancer cells. Consistent with these observations, TNFAIP8 blocked AKT/mTOR signaling and showed direct interaction with ATG3-ATG7 proteins. TNFAIP8 also exhibited binding with fatty acids and modulated expression of lipid/fatty-acid metabolizing enzymes. Chronic feeding of mice with alcohol increased hepatic levels of TNFAIP8, autophagy, and steatosis but not in high-fat-fed obese mice. Similarly, higher TNFAIP8 expression was associated with steatotic livers of human patients with a history of alcohol use but not in steatotic patients with no history of alcohol use. Our data indicate a novel role of TNFAIP8 in modulation of drug resistance, autophagy, and hepatic steatosis, all key early events in HCC progression.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Minghui Lin
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
- Ningxia Medical University, Ningxia Hui Autonomous Region, Yinchuan, 750004, China
| | - Uchechukwu Chimeh
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Xialan Dong
- Department of Pharmaceutical Sciences, Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Weifan Zheng
- Department of Pharmaceutical Sciences, Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University Durham, Durham, NC, 27707, USA
| | - John Moore
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, Durham, NC, 27707, USA.
- Department of Pharmaceutical Sciences, North Carolina Central University Durham, Durham, NC, 27707, USA.
| |
Collapse
|
15
|
Malczewska A, Kos-Kudła B, Kidd M, Drozdov I, Bodei L, Matar S, Oberg K, Modlin IM. The clinical applications of a multigene liquid biopsy (NETest) in neuroendocrine tumors. Adv Med Sci 2020; 65:18-29. [PMID: 31841822 PMCID: PMC7453408 DOI: 10.1016/j.advms.2019.10.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/19/2019] [Accepted: 10/18/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE There are few effective biomarkers for neuroendocrine tumors. Precision oncology strategies have provided liquid biopsies for real-time and tailored decision-making. This has led to the development of the first neuroendocrine tumor liquid biopsy (the NETest). The NETest represents a transcriptomic signature of neuroendocrine tumor (NETs) that captures tumor biology and disease activity. The data have direct clinical application in terms of identifying residual disease, disease progress and the efficacy of treatment. In this overview we assess the available published information on the metrics and clinical efficacy of the NETest. MATERIAL AND METHODS Published data on the NETest have been collated and analyzed to understand the clinical application of this multianalyte biomarker in NETs. RESULTS NETest assay has been validated as a standardized and reproducible clinical laboratory measurement. It is not affected by demographic characteristics, or acid suppressive medication. Clinical utility of the NETest has been documented in gastroenteropancreatic, bronchopulmonary NETs, in paragangliomas and pheochromocytomas. The test facilitates accurate diagnosis of a NET disease, and real-time monitoring of the disease status (stable/progressive disease). It predicts aggressive tumor behavior, identifies operative tumor resection, and efficacy of the medical treatment (e.g. somatostatin analogues), or peptide receptor radionuclide therapy (PRRT). NETest metrics and clinical applications out-perform standard biomarkers like chromogranin A. CONCLUSIONS The NETest exhibits clinically competent metrics as an effective biomarker for neuroendocrine tumors. Measurement of NET transcripts in blood is a significant advance in neuroendocrine tumor management and demonstrates that blood provides a viable source to identify and monitor tumor status.
Collapse
Affiliation(s)
- Anna Malczewska
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, Katowice, Poland.
| | - Beata Kos-Kudła
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, Katowice, Poland
| | - Mark Kidd
- Wren Laboratories, Branford, CT, USA
| | | | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Kjell Oberg
- Department of Endocrine Oncology, University Hospital, Uppsala, Sweden
| | - Irvin M Modlin
- Gastroenterological Surgery, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Przytycki PF, Singh M. Differential Allele-Specific Expression Uncovers Breast Cancer Genes Dysregulated by Cis Noncoding Mutations. Cell Syst 2020; 10:193-203.e4. [PMID: 32078798 PMCID: PMC7457951 DOI: 10.1016/j.cels.2020.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/04/2019] [Accepted: 01/22/2020] [Indexed: 01/23/2023]
Abstract
Identifying cancer-relevant mutations in noncoding regions is challenging due to the large numbers of such mutations, their low levels of recurrence, and difficulties in interpreting their functional impact. To uncover genes that are dysregulated due to somatic mutations in cis, we build upon the concept of differential allele-specific expression (ASE) and introduce methods to identify genes within an individual's cancer whose ASE differs from what is found in matched normal tissue. When applied to breast cancer tumor samples, our methods detect the known allele-specific effects of copy number variation and nonsense-mediated decay. Further, genes that are found to recurrently exhibit differential ASE across samples are cancer relevant. Genes with cis mutations are enriched for differential ASE, and we find 147 potentially functional noncoding mutations cis to genes that exhibit significant differential ASE. We conclude that differential ASE is a promising means for discovering gene dysregulation due to cis noncoding mutations.
Collapse
Affiliation(s)
- Pawel F Przytycki
- Department of Computer Science, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Mona Singh
- Department of Computer Science, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
17
|
Gupta SC, Awasthee N, Rai V, Chava S, Gunda V, Challagundla KB. Long non-coding RNAs and nuclear factor-κB crosstalk in cancer and other human diseases. Biochim Biophys Acta Rev Cancer 2019; 1873:188316. [PMID: 31639408 DOI: 10.1016/j.bbcan.2019.188316] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 12/29/2022]
Abstract
The regulation of the pleiotropic transcription factor, nuclear factor-κB (NF-κB) by miRNAs and proteins is extensively studied. More recently, the NF-κB signaling was also reported to be regulated by several long non-coding RNAs (lncRNAs) that constitute the major portion of the noncoding component of the human genome. The common NF-κB associated lncRNAs include NKILA, HOTAIR, MALAT1, ANRIL, Lethe, MIR31HG, and PACER. The lncRNA and NF-κB signaling crosstalk during cancer and other diseases such as cardiomyopathy, celiac disease, cerebral infarction, chronic kidney disease, diabetes mellitus, Kawasaki disease, pregnancy loss, and rheumatoid arthritis. Some NF-κB related lncRNAs can affect gene expression without modulating NF-κB signaling. Most of the lncRNAs with a potential to modulate NF-κB signaling are regulated by NF-κB itself suggesting a feedback regulation. The discovery of lncRNAs have provided a new type of regulation for the NF-κB signaling and thus could be explored for therapeutic interventions. The manner in which lncRNA and NF-κB crosstalk affects human pathophysiology is discussed in this review. The challenges associated with the therapeutic interventions of this crosstalk are also discussed.
Collapse
Affiliation(s)
- Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Nikee Awasthee
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Srinivas Chava
- Department of Biochemistry & Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Venugopal Gunda
- Pediatric Oncology Laboratory, Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kishore B Challagundla
- Department of Biochemistry & Molecular Biology, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
18
|
Zhang Y, Gao L, Ma S, Ma J, Wang Y, Li S, Hu X, Han S, Zhou M, Zhou L, Ding Z. MALAT1-KTN1-EGFR regulatory axis promotes the development of cutaneous squamous cell carcinoma. Cell Death Differ 2019; 26:2061-2073. [PMID: 30683916 PMCID: PMC6748142 DOI: 10.1038/s41418-019-0288-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 12/25/2018] [Accepted: 01/08/2019] [Indexed: 02/03/2023] Open
Abstract
Long noncoding RNAs (LncRNAs), including MALAT1, are critical regulators of tumor development. However, the roles and molecular mechanisms of LncRNAs in cutaneous squamous cell carcinoma (cSCC) remain underexplored. In this study, functional studies using in vitro cellular and in vivo xenograft models confirmed the pro-carcinogenic roles of MALAT1 in cSCC. Further, MALAT1 was identified to regulate epidermal growth factor receptor (EGFR) protein expression but did not affect EGFR mRNA expression. Transcriptomic sequencing identified kinectin 1 (KTN1) as the key mediator for MALAT1 regulation of EGFR. Mechanistic study revealed that MALAT1 interacts with c-MYC to form a complex and directly binds to the promoter region of KTN1 gene and enhances its transactivation to positively regulate EGFR protein expression. Our findings, therefore, establish a novel c-MYC-assisted MALAT1-KTN1-EGFR axis, which contributes to cSCC development and may serve as novel target for therapeutic intervention.
Collapse
Grants
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 81573076 National Natural Science Foundation of China (National Science Foundation of China)
- 81172634 National Natural Science Foundation of China (National Science Foundation of China)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- 2016A030313738 Guangdong Science and Technology Department (Science and Technology Department, Guangdong Province)
- Grants from the School of Public Health of Southern Medical University, China (Grant No.GW201612)
Collapse
Affiliation(s)
- Ying Zhang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Lin Gao
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Shudong Ma
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ji Ma
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Yinghui Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Shanshan Li
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xia Hu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Shuo Han
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Liang Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Zhenhua Ding
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
19
|
Kyuno D, Zhao K, Schnölzer M, Provaznik J, Hackert T, Zöller M. Claudin7-dependent exosome-promoted reprogramming of nonmetastasizing tumor cells. Int J Cancer 2019; 145:2182-2200. [PMID: 30945750 DOI: 10.1002/ijc.32312] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/10/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022]
Abstract
Claudin7 (cld7) is a cancer-initiating cell (CIC) marker in gastrointestinal tumors, a cld7-knockdown (kd) being accompanied by loss of tumor progression. Tumor exosomes (TEX) restoring CIC activities, we explored the contribution of cld7. This became particularly interesting, as tight junction (TJ)- and glycolipid-enriched membrane domain (GEM)-derived cld7 is recruited into distinct TEX. TEXs were derived from CIC or cld7kd cells of a rat pancreatic and a human colon cancer line. TEX derived from pancreatic cancer cld7kd cells rescued with palmitoylation site-deficient cld7 (cld7mP) allowed selectively evaluating the contribution of GEM-derived TEX, only palmitoylated cld7 being integrated into GEM. Cld7 CIC-TEX promoted tumor cell dissemination and metastatic growth without a major impact on proliferation, apoptosis resistance and epithelial-mesenchymal transition. Instead, migration, invasion and (lymph)angiogenesis were strongly supported, only migration being selectively fostered by GEM-derived cld7 TEX. CIC-TEX coculture of cld7kd cells uncovered significant changes in the cld7kd cell protein and miRNA profiles. However, changes did not correspond to the CIC-TEX profile, CIC-TEX rather initiating integrin, protease and RTK, particularly lymphangiogenic receptor activation. CIC-TEX preferentially rescuing cld7kd-associated defects in signal transduction was backed up by an RTK inhibitor neutralizing the impact of CIC-TEX on tumor progression. In conclusion, cld7 contributes to selective steps of the metastatic cascade. Defects of cld7kd and cld7mP cells in migration, invasion and (lymph)angiogenesis are effaced by CIC-TEX that act by signaling cascade activation. Accordingly, RTK inhibitors are an efficient therapeutic defeating CIC-TEX.
Collapse
Affiliation(s)
- Daisuke Kyuno
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo, Japan
| | - Kun Zhao
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martina Schnölzer
- Functional Proteome Analysis, German Cancer Research Center, Heidelberg, Germany
| | | | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Margot Zöller
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Day TF, Kallakury BVS, Ross JS, Voronel O, Vaidya S, Sheehan CE, Kasid UN. Dual Targeting of EGFR and IGF1R in the TNFAIP8 Knockdown Non-Small Cell Lung Cancer Cells. Mol Cancer Res 2019; 17:1207-1219. [PMID: 30647104 DOI: 10.1158/1541-7786.mcr-18-0731] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/10/2018] [Accepted: 01/08/2019] [Indexed: 12/16/2022]
Abstract
Aberrant regulation of EGFR is common in non-small cell lung carcinomas (NSCLC), and tumor resistance to targeted therapies has been attributed to emergence of other co-occurring oncogenic events, parallel bypass receptor tyrosine kinase pathways including IGF1R, and TNFα-driven adaptive response via NF-κB. TNFAIP8, TNFα-inducible protein 8, is an NF-κB-activated prosurvival and oncogenic molecule. TNFAIP8 expression protects NF-κB-null cells from TNFα-induced cell death by inhibiting caspase-8 activity. Here, we demonstrate that knockdown of TNFAIP8 inhibited EGF and IGF-1-stimulated migration in NSCLC cells. TNFAIP8 knockdown cells showed decreased level of EGFR and increased expression of sorting nexin 1 (SNX1), a key regulator of the EGFR trafficking through the endosomal compartments, and treatment with SNX1 siRNA partially restored EGFR expression in these cells. TNFAIP8 knockdown cells also exhibited downregulation of IGF-1-induced pIGF1R and pAKT, and increased expression of IGF-1-binding protein 3 (IGFBP3), a negative regulator of the IGF-1/IGF1R signaling. Consistently, treatment of TNFAIP8 knockdown cells with IGFBP3 siRNA restored pIGF1R and pAKT levels. TNFAIP8 knockdown cells had enhanced sensitivities to inhibitors of EGFR, PI3K, and AKT. Furthermore, IHC expression of TNFAIP8 was associated with poor prognosis in NSCLC. These findings demonstrate TNFAIP8-mediated regulation of EGFR and IGF1R via SNX1 and IGFBP3, respectively. We posit that TNFAIP8 is a viable, multipronged target downstream of the TNFα/NF-κB axis, and silencing TNFAIP8 may overcome adaptive response in NSCLC. IMPLICATIONS: TNFAIP8 and its effectors SNX1 and IGFBP3 may be exploited to improve the efficacy of molecular-targeted therapies in NSCLC and other cancers.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/5/1207/F1.large.jpg.
Collapse
Affiliation(s)
- Timothy F Day
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Bhaskar V S Kallakury
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Jeffrey S Ross
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York
| | - Olga Voronel
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York
| | - Shantashri Vaidya
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| | - Christine E Sheehan
- Department of Pathology and Laboratory Medicine, Albany Medical College, Albany, New York
| | - Usha N Kasid
- Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC.
| |
Collapse
|
21
|
Niture S, Dong X, Arthur E, Chimeh U, Niture SS, Zheng W, Kumar D. Oncogenic Role of Tumor Necrosis Factor α-Induced Protein 8 (TNFAIP8). Cells 2018; 8:cells8010009. [PMID: 30586922 PMCID: PMC6356598 DOI: 10.3390/cells8010009] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
Tumor necrosis factor (TNF)-α-induced protein 8 (TNFAIP8) is a founding member of the TIPE family, which also includes TNFAIP8-like 1 (TIPE1), TNFAIP8-like 2 (TIPE2), and TNFAIP8-like 3 (TIPE3) proteins. Expression of TNFAIP8 is strongly associated with the development of various cancers including cancer of the prostate, liver, lung, breast, colon, esophagus, ovary, cervix, pancreas, and others. In human cancers, TNFAIP8 promotes cell proliferation, invasion, metastasis, drug resistance, autophagy, and tumorigenesis by inhibition of cell apoptosis. In order to better understand the molecular aspects, biological functions, and potential roles of TNFAIP8 in carcinogenesis, in this review, we focused on the expression, regulation, structural aspects, modifications/interactions, and oncogenic role of TNFAIP8 proteins in human cancers.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | - Xialan Dong
- Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Elena Arthur
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | - Uchechukwu Chimeh
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
| | | | - Weifan Zheng
- Bio-manufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute (BBRI), North Carolina Central University, Durham, NC 27707, USA.
- Department of Pharmaceutical Sciences, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
22
|
Wang J, Gao H, Liu G, Gu L, Yang C, Zhang F, Liu T. Tumor necrosis factor α–induced protein 8 expression as a predictor of prognosis and resistance in patients with advanced ovarian cancer treated with neoadjuvant chemotherapy. Hum Pathol 2018; 82:239-248. [DOI: 10.1016/j.humpath.2018.02.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/12/2018] [Accepted: 02/16/2018] [Indexed: 10/28/2022]
|
23
|
Yu DL, Li HW, Wang Y, Li CQ, You D, Jiang L, Song YP, Li XH. Acyl-CoA dehydrogenase long chain expression is associated with esophageal squamous cell carcinoma progression and poor prognosis. Onco Targets Ther 2018; 11:7643-7653. [PMID: 30464513 PMCID: PMC6217208 DOI: 10.2147/ott.s171963] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background Acyl-CoA dehydrogenase long chain (ACADL) was revealed to have a correlation with malignant progression of cancer. However, whether ACADL plays a role in clinical therapy remains unclear. The clinicopathological role of ACADL in esophageal squamous cell carcinoma (ESCC) will be discussed in this study. Materials and methods The expression of ACADL was analyzed via real-time PCR and Western blotting to assess mRNA and protein levels in ESCC cell lines and normal esophageal epithelial cells (NEECs), in six paired ESCC tumors and relative normal tissues. Furthermore, immunohistochemical staining was performed on 135 paraffin-embedded ESCC specimens to assess ACADL expression. The clinicopathological significance of ACADL expression was further investigated via survival analysis and Cox regression analysis. Results ACADL was found to be markedly upregulated in ESCC cell lines when compared with NEECs. Moreover, various experiments such as quantitative real-time PCR, Western blot, and immunohistochemical analyses all revealed that ACADL expression was increased in all six paired ESCC tumors and matched normal tissues. Furthermore, immunohistochemical analysis revealed an increased level of ACADL protein expression in all 135 paraffin-embedded samples from ESCC patients, which increased with disease progression. Conclusion We demonstrated that ACADL is overexpressed in ESCC, both in cell lines and clinical specimens. ACADL is found to be a vital regulator in ESCC progression and can predict a worse outcome for ESCC patients, suggesting that ACADL might be a valuable molecule to be targeted for clinical therapy of ESCC treatment.
Collapse
Affiliation(s)
- Dong-Lin Yu
- Department of basic Theory of traditional Chinese Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Hong-Wei Li
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China, ;
| | - Yang Wang
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China, ;
| | - Cun-Qi Li
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China, ;
| | - Dong You
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China, ;
| | - Lei Jiang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| | - Yi-Peng Song
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China, ;
| | - Xing-Hua Li
- Department of Radiotherapy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China, ;
| |
Collapse
|
24
|
TIPE Family of Proteins and Its Implications in Different Chronic Diseases. Int J Mol Sci 2018; 19:ijms19102974. [PMID: 30274259 PMCID: PMC6213092 DOI: 10.3390/ijms19102974] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/19/2018] [Accepted: 09/22/2018] [Indexed: 12/14/2022] Open
Abstract
The tumor necrosis factor-α-induced protein 8-like (TIPE/TNFAIP8) family is a recently identified family of proteins that is strongly associated with the regulation of immunity and tumorigenesis. This family is comprised of four members, namely, tumor necrosis factor-α-induced protein 8 (TIPE/TNFAIP8), tumor necrosis factor-α-induced protein 8-like 1 (TIPE1/TNFAIP8L1), tumor necrosis factor-α-induced protein 8-like 2 (TIPE2/TNFAIP8L2), and tumor necrosis factor-α-induced protein 8-like 3 (TIPE3/TNFAIP8L3). Although the proteins of this family were initially described as regulators of tumorigenesis, inflammation, and cell death, they are also found to be involved in the regulation of autophagy and the transfer of lipid secondary messengers, besides contributing to immune function and homeostasis. Interestingly, despite the existence of a significant sequence homology among the four members of this family, they are involved in different biological activities and also exhibit remarkable variability of expression. Furthermore, this family of proteins is highly deregulated in different human cancers and various chronic diseases. This review summarizes the vivid role of the TIPE family of proteins and its association with various signaling cascades in diverse chronic diseases.
Collapse
|
25
|
Padmavathi G, Banik K, Monisha J, Bordoloi D, Shabnam B, Arfuso F, Sethi G, Fan L, Kunnumakkara AB. Novel tumor necrosis factor-α induced protein eight (TNFAIP8/TIPE) family: Functions and downstream targets involved in cancer progression. Cancer Lett 2018; 432:260-271. [DOI: 10.1016/j.canlet.2018.06.017] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 12/21/2022]
|
26
|
Modlin IM, Kidd M, Malczewska A, Drozdov I, Bodei L, Matar S, Chung KM. The NETest: The Clinical Utility of Multigene Blood Analysis in the Diagnosis and Management of Neuroendocrine Tumors. Endocrinol Metab Clin North Am 2018; 47:485-504. [PMID: 30098712 PMCID: PMC6716518 DOI: 10.1016/j.ecl.2018.05.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The neuroendocrine neoplasms test (NETest) is a multianalyte liquid biopsy that measures neuroendocrine tumor gene expression in blood. This unique signature precisely defines the biological activity of an individual tumor in real time. The assay meets the 3 critical requirements of an optimal biomarker: diagnostic accuracy, prognostic value, and predictive therapeutic assessment. NETest performance metrics are sensitivity and specificity and in head-to-head comparison are 4-fold to 10-fold more accurate than chromogranin A. NETest accurately identifies completeness of surgery and response to somatostatin analogs. Clinical registry data demonstrate significant clinical utility in watch/wait programs.
Collapse
Affiliation(s)
- Irvin M Modlin
- Gastroenterological and Endoscopic Surgery, Yale University School of Medicine, 310 Cedar Street, New Haven, CT 06520-8062, USA.
| | - Mark Kidd
- Wren Laboratories, 35 NE Industrial Road, Branford, CT 06405, USA
| | - Anna Malczewska
- Department of Endocrinology and Neuroendocrine Tumors, Medical University of Silesia, ul. Ceglana 35, Katowice 40-514, Poland
| | - Ignat Drozdov
- Wren Laboratories, 35 NE Industrial Road, Branford, CT 06405, USA
| | - Lisa Bodei
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 77, New York, NY 10065, USA
| | - Somer Matar
- Wren Laboratories, 35 NE Industrial Road, Branford, CT 06405, USA
| | - Kyung-Min Chung
- Wren Laboratories, 35 NE Industrial Road, Branford, CT 06405, USA
| |
Collapse
|
27
|
Moore KS, Yagci N, van Alphen F, Meijer AB, ‘t Hoen PAC, von Lindern M. Strap associates with Csde1 and affects expression of select Csde1-bound transcripts. PLoS One 2018; 13:e0201690. [PMID: 30138317 PMCID: PMC6107111 DOI: 10.1371/journal.pone.0201690] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 07/22/2018] [Indexed: 02/06/2023] Open
Abstract
Erythropoiesis is regulated at many levels, including control of mRNA translation. Changing environmental conditions, such as hypoxia or the availability of nutrients and growth factors, require a rapid response enacted by the enhanced or repressed translation of existing transcripts. Cold shock domain protein e1 (Csde1/Unr) is an RNA-binding protein required for erythropoiesis and strongly upregulated in erythroblasts relative to other hematopoietic progenitors. The aim of this study is to identify the Csde1-containing protein complexes and investigate their role in post-transcriptional expression control of Csde1-bound transcripts. We show that Serine/Threonine kinase receptor-associated protein (Strap/Unrip), was the protein most strongly associated with Csde1 in erythroblasts. Strap is a WD40 protein involved in signaling and RNA splicing, but its role when associated with Csde1 is unknown. Reduced expression of Strap did not alter the pool of transcripts bound by Csde1. Instead, it altered the mRNA and/or protein expression of several Csde1-bound transcripts that encode for proteins essential for translational regulation during hypoxia, such as Hmbs, eIF4g3 and Pabpc4. Also affected by Strap knockdown were Vim, a Gata-1 target crucial for erythrocyte enucleation, and Elavl1, which stabilizes Gata-1 mRNA. The major cellular processes affected by both Csde1 and Strap were ribosome function and cell cycle control.
Collapse
Affiliation(s)
- Kat S. Moore
- Sanquin Research, Department of Hematopoiesis, and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nurcan Yagci
- Sanquin Research, Department of Hematopoiesis, and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Floris van Alphen
- Sanquin Research, Department of Research Facilities, Amsterdam, The Netherlands
| | - Alexander B. Meijer
- Sanquin Research, Department of Research Facilities, Amsterdam, The Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Peter A. C. ‘t Hoen
- Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marieke von Lindern
- Sanquin Research, Department of Hematopoiesis, and Landsteiner Laboratory Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
28
|
Niture S, Ramalinga M, Kedir H, Patacsil D, Niture SS, Li J, Mani H, Suy S, Collins S, Kumar D. TNFAIP8 promotes prostate cancer cell survival by inducing autophagy. Oncotarget 2018; 9:26884-26899. [PMID: 29928491 PMCID: PMC6003558 DOI: 10.18632/oncotarget.25529] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/03/2018] [Indexed: 01/11/2023] Open
Abstract
Tumor necrosis factor-α-inducible protein 8 (TNFAIP8) is a TNF-α inducible anti-apoptotic protein with multiple roles in tumor growth and survival. Mechanisms of cell survival by TNFAIP8 remain elusive. We investigated the role of TNFAIP8 in the regulation of the cell cycle, autophagy, cell survival and neuroendocrine differentiation in prostate cancer cells. We showed that TNFAIP8 dysregulates cell-cycle-related proteins, in PC3 cells. Oncogenic cell survival, drug resistance and dysregulation of cell cycle-related proteins are often associated with autophagy. We demonstrated that TNFAIP8 induces autophagy by increasing expression of autophagy effectors such as LC3β I/II, Beclin1, 4EBP1, p62, and SIRT1. We also demonstrated that TNFAIP8 interacts with autophagy-related protein 3 (ATG3). TNFα treatment increased the expression of TNFAIP8, which was associated with increased autophagy and decreased apoptosis. We also observed an increase in expression of neuroendocrine differentiation markers, synaptophysin and chromogranin A, and drug resistance to anticancer drugs, docetaxel and doxorubicin, in cells transfected with TNFAIP8. Collectively, our findings reveal that by the creation of cellular autophagy events, TNFAIP8 promotes cell survival and drug resistance in prostate cancer cells.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 27707 NC, USA.,Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | - Malathi Ramalinga
- Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | - Habib Kedir
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 27707 NC, USA.,Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | - Dorrelyn Patacsil
- Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA
| | | | - James Li
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| | - Haresh Mani
- Department of Pathology, Inova Fairfax Hospital, Falls Church, 22042 VA, USA
| | - Simeng Suy
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| | - Sean Collins
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, 27707 NC, USA.,Cancer Research Laboratory, University of the District of Columbia, Washington, 20008 DC, USA.,Lombardi Comprehensive Cancer Center, Georgetown University, Washington, 20008 DC, USA
| |
Collapse
|
29
|
Zhang L, Liu R, Luan YY, Yao YM. Tumor Necrosis Factor-α Induced Protein 8: Pathophysiology, Clinical Significance, and Regulatory Mechanism. Int J Biol Sci 2018; 14:398-405. [PMID: 29725261 PMCID: PMC5930472 DOI: 10.7150/ijbs.23268] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor-α-induced protein-8 (TNFAIP8) is the earliest discovered component of TNFAIP8 family [tumor necrosis factor-α-induced protein-8 like (TIPE) family]. TNFAIP8 contains a putative death effector domain (DED) homologous to DED II in FLIP (Fas-associated death domain-like interleukin-1β-converting enzyme-inhibitory protein), which may affect cell survival/death process. Recently, it has been demonstrated that TNFAIP8 could inhibit apoptosis and autophagy in various types of cells. Moreover, TNFAIP8 level fluctuated evidently in patients with inflammatory, malignant, and autoimmune diseases, indicating that it might be an anti-apoptotic and oncogenetic protein. Herein we will review the discovery, gene/protein structure, pathophysiological functions, and clinical significance of TNFAIP8 together with its potential regulatory mechanism.
Collapse
Affiliation(s)
- Lei Zhang
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.,Emergency Department, The General Hospital of the Chinese PLA Rocket Force, Beijing 100088, People's Republic of China
| | - Ran Liu
- Department of Endocrinology, 307th Hospital of the Chinese PLA, Beijing 100071, People's Republic of China
| | - Ying-Yi Luan
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| |
Collapse
|
30
|
PRRT genomic signature in blood for prediction of 177Lu-octreotate efficacy. Eur J Nucl Med Mol Imaging 2018; 45:1155-1169. [PMID: 29484451 DOI: 10.1007/s00259-018-3967-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 01/31/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Peptide receptor radionuclide therapy (PRRT) utilizes somatostatin receptor (SSR) overexpression on neuroendocrine tumors (NET) to deliver targeted radiotherapy. Intensity of uptake at imaging is considered related to efficacy but has low sensitivity. A pretreatment strategy to determine individual PRRT response remains a key unmet need. NET transcript expression in blood integrated with tumor grade provides a PRRT predictive quotient (PPQ) which stratifies PRRT "responders" from "non-responders". This study clinically validates the utility of the PPQ in NETs. METHODS The development and validation of the PPQ was undertaken in three independent 177Lu-PRRT treated cohorts. Specificity was tested in two separate somatostatin analog-treated cohorts. Prognostic value of the marker was defined in a cohort of untreated patients. The developmental cohort included lung and gastroenteropancreatic [GEP] NETs (n = 72) from IRST Meldola, Italy. The majority were GEP (71%) and low grade (86% G1-G2). Prospective validation cohorts were from Zentralklinik Bad Berka, Germany (n = 44), and Erasmus Medical Center, Rotterdam, Netherlands (n = 42). Each cohort included predominantly well differentiated, low grade (86-95%) lung and GEP-NETs. The non-PRRT comparator cohorts included SSA cohort I, n = 28 (100% low grade, 100% GEP-NET); SSA cohort II, n = 51 (98% low grade; 76% GEP-NET); and an untreated cohort, n = 44 (64% low grade; 91% GEP-NET). Baseline evaluations included clinical information (disease status, grade, SSR) and biomarker (CgA). NET blood gene transcripts (n = 8: growth factor signaling and metabolism) were measured pre-therapy and integrated with tumor Ki67 using a logistic regression model. This provided a binary output: "predicted responder" (PPQ+); "predicted non-responder" (PPQ-). Treatment response was evaluated using RECIST criteria [Responder (stable, partial and complete response) vs Non-Responder)]. Sample measurement and analyses were blinded to study outcome. Statistical evaluation included Kaplan-Meier survival and standard test evaluation analyses. RESULTS In the developmental cohort, 56% responded to PRRT. The PPQ predicted 100% of responders and 84% of non-responders (accuracy: 93%). In the two validation cohorts (response: 64-79%), the PPQ was 95% accurate (Bad Berka: PPQ + =97%, PPQ- = 93%; Rotterdam: PPQ + =94%, PPQ- = 100%). Overall, the median PFS was not reached in PPQ+ vs PPQ- (10-14 months; HR: 18-77, p < 0.0001). In the comparator cohorts, the predictor (PPQ) was 47-50% accurate for SSA-treatment and 50% as a prognostic. No differences in PFS were respectively noted (PPQ+: 10-12 months vs. PPQ-: 9-15 months). CONCLUSION The PPQ derived from circulating NET specific genes and tumor grade prior to the initiation of therapy is a highly specific predictor of the efficacy of PRRT with an accuracy of 95%.
Collapse
|
31
|
Abstract
Constructing, evaluating, and interpreting gene networks generally sits within the broader field of systems biology, which continues to emerge rapidly, particular with respect to its application to understanding the complexity of signaling in the context of cancer biology. For the purposes of this volume, we take a broad definition of systems biology. Considering an organism or disease within an organism as a system, systems biology is the study of the integrated and coordinated interactions of the network(s) of genes, their variants both natural and mutated (e.g., polymorphisms, rearrangements, alternate splicing, mutations), their proteins and isoforms, and the organic and inorganic molecules with which they interact, to execute the biochemical reactions (e.g., as enzymes, substrates, products) that reflect the function of that system. Central to systems biology, and perhaps the only approach that can effectively manage the complexity of such systems, is the building of quantitative multiscale predictive models. The predictions of the models can vary substantially depending on the nature of the model and its inputoutput relationships. For example, a model may predict the outcome of a specific molecular reaction(s), a cellular phenotype (e.g., alive, dead, growth arrest, proliferation, and motility), a change in the respective prevalence of cell or subpopulations, a patient or patient subgroup outcome(s). Such models necessarily require computers. Computational modeling can be thought of as using machine learning and related tools to integrate the very high dimensional data generated from modern, high throughput omics technologies including genomics (next generation sequencing), transcriptomics (gene expression microarrays; RNAseq), metabolomics and proteomics (ultra high performance liquid chromatography, mass spectrometry), and "subomic" technologies to study the kinome, methylome, and others. Mathematical modeling can be thought of as the use of ordinary differential equations and related tools to create dynamic, semi-mechanistic models of low dimensional data including gene/protein signaling as a function of time/dose. More recently, the integration of imaging technologies into predictive multiscale modeling has begun to extend further the scales across which data can be obtained and used to gain insight into system function.There are several goals for predictive multiscale modeling including the more academic pursuit of understanding how the system or local feature thereof is regulated or functions, to the more practical or translational goals of identifying predictive (selecting which patient should receive which drug/therapy) or prognostic (disease progress and outcome in an individual patient) biomarkers and/or identifying network vulnerabilities that represent potential targets for therapeutic benefit with existing drugs (including drug repurposing) or for the development of new drugs. These various goals are not necessarily mutually exclusive or inclusive. Within this volume, readers will find examples of many of the activities noted above. Each chapter contains practical and/or methodological insights to guide readers in the design and interpretation of their own and published work.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, W405A Research Building, 3970 Reservoir NW, Washington, DC, 20057, USA.
| |
Collapse
|
32
|
Li L, Karanika S, Yang G, Wang J, Park S, Broom BM, Manyam GC, Wu W, Luo Y, Basourakos S, Song JH, Gallick GE, Karantanos T, Korentzelos D, Azad AK, Kim J, Corn PG, Aparicio AM, Logothetis CJ, Troncoso P, Heffernan T, Toniatti C, Lee HS, Lee JS, Zuo X, Chang W, Yin J, Thompson TC. Androgen receptor inhibitor-induced "BRCAness" and PARP inhibition are synthetically lethal for castration-resistant prostate cancer. Sci Signal 2017; 10:eaam7479. [PMID: 28536297 PMCID: PMC5855082 DOI: 10.1126/scisignal.aam7479] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cancers with loss-of-function mutations in BRCA1 or BRCA2 are deficient in the DNA damage repair pathway called homologous recombination (HR), rendering these cancers exquisitely vulnerable to poly(ADP-ribose) polymerase (PARP) inhibitors. This functional state and therapeutic sensitivity is referred to as "BRCAness" and is most commonly associated with some breast cancer types. Pharmaceutical induction of BRCAness could expand the use of PARP inhibitors to other tumor types. For example, BRCA mutations are present in only ~20% of prostate cancer patients. We found that castration-resistant prostate cancer (CRPC) cells showed increased expression of a set of HR-associated genes, including BRCA1, RAD54L, and RMI2 Although androgen-targeted therapy is typically not effective in CRPC patients, the androgen receptor inhibitor enzalutamide suppressed the expression of those HR genes in CRPC cells, thus creating HR deficiency and BRCAness. A "lead-in" treatment strategy, in which enzalutamide was followed by the PARP inhibitor olaparib, promoted DNA damage-induced cell death and inhibited clonal proliferation of prostate cancer cells in culture and suppressed the growth of prostate cancer xenografts in mice. Thus, antiandrogen and PARP inhibitor combination therapy may be effective for CRPC patients and suggests that pharmaceutically inducing BRCAness may expand the clinical use of PARP inhibitors.
Collapse
Affiliation(s)
- Likun Li
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Styliani Karanika
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Guang Yang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Jiangxiang Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Sanghee Park
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Bradley M Broom
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Wenhui Wu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Yong Luo
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Spyridon Basourakos
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Jian H Song
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Gary E Gallick
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Theodoros Karantanos
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Dimitrios Korentzelos
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Abul Kalam Azad
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Jeri Kim
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Paul G Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Ana M Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Timothy Heffernan
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Carlo Toniatti
- ORBIT (Oncology Research for Biologics and Immunotherapy Translation), The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Hyun-Sung Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Xuemei Zuo
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Wenjun Chang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Jianhua Yin
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA
| | - Timothy C Thompson
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, USA.
| |
Collapse
|