1
|
Wang B, Zhou X, Wu D, Gao L, Wan Z, Wu R. Development and validation of M2 macrophage-related genes in a prognostic model of lung adenocarcinoma based on bulk RNA and ScRNA datasets. Discov Oncol 2025; 16:352. [PMID: 40100580 PMCID: PMC11920479 DOI: 10.1007/s12672-025-02123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the correlation between M2 macrophages activity with the prognosis of lung adenocarcinoma (LUAD). We sought to identify key genes associated with M2 macrophage activity and examine their relationship with clinicopathological features to elucidate the underlying mechanism. METHODS Published datases were analyzed for differentially expressed genes. After quality control, batch effect removal, and annotation, the scRNA dataset identified M2 macrophage-associated differentially expressed genes in the LUAD group, which were cross-analyzed and referred to as M2 macrophage-linked genes. A risk model was generated using machine learing for these genes. Thereafter, two bulk RNA-seq datasets were used to evaluate the model. We computed risk scores for all samples and grouped them into low and high risk, aiding in the comparison of clinical characteristics, immune and stromal infiltration, and drug sensitivity. Finally, key genes were validated through immunohistochemistry in IPA samples. RESULTS We identified four key M2 macrophage-linked genes: TIMP1, CAV2, MIF, and SELENBP1. Survival durations in the high-riskscore cluster were lower across the TCGA-LUAD (P = 1.2 × 10-4), GSE14814 (P = 0.02), and GSE37745 (P = 0.01) data sets. The stromal score, fibroblast infiltration, and cytokinesis activation were increased in the high-risk subgroup. Neutrophil and endothelial cell infiltration and activation of the linolenic acid pathway occurred in the low-risk group. IHC confirmed that CAV2 and SELENBP1 expression was significantly reduced, while TIMP1 and MIF were significantly increased in LUAD, which was consistent with the bioinformatics findings. CONCLUSION The role of M2 macrophages in tumor progression could anticipate the prognosis of LUAD and develop novel immunotherapy strategies.
Collapse
Affiliation(s)
- Bolin Wang
- Graduate School of Chengde Medical College, Chengde, Hebei, China
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Xiaofeng Zhou
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Di Wu
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Lu Gao
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Zhihua Wan
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China
| | - Ruifeng Wu
- Department of Chest Surgery, Baoding First Central Hospital, Baoding, Hebei, China.
| |
Collapse
|
2
|
Pant A, Moar K, Maurya PK. Impact of estradiol in inducing endometrial cancer using RL95-2. Pathol Res Pract 2024; 263:155640. [PMID: 39383736 DOI: 10.1016/j.prp.2024.155640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/27/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Endometrial cancer is the most common gynecological malignancy that originates from the inner lining of the uterus and predominantly affects postmenopausal women. Prolonged exposure to estrogen, family history of endometrial cancer, obesity, and hormonal imbalance are some of the risk factors associated with endometrial cancer. In our study, we investigated the effect of estradiol, a potent form of estrogen at various concentrations on endometrial cell line RL95-2. METHODS Endometrial cell RL95-2 were cultured in DMEM medium with optimal conditions required to maintain the cells. MTT assay and colony formation assay were further performed after treating the cells with different concentrations of estradiol (1, 10, and 100 nM) and TAM (100 nM). Moreover, the effect of genes regulated by estradiol was also examined using microarray and validated using real-time polymerase chain reaction (qRT-PCR). RESULTS Time-dependent MTT assay shows a significant change in the ability of the cells to survive relative to concentrations. Colony formation was found to be directly proportional to the concentration of the estradiol (p < 0.05). Among genes, MMP14 (p = 0.03), SPARCL1 (p = 0.005), and CLU (p = 0.06) showed a significant up-regulation in their expression after estradiol treatment while NRN1 (p < 0.001) showed significant downregulation in expression pattern compared to control. However, the TAM treatment was found to be significantly effective after 72 h (p < 0.001) compared to control and 100 nM E2 (p = 0.0206). CONCLUSION Our study suggests that estradiol significantly contributes to regulating the viability, colony formation, and expression of genes associated with endometrial cancer.
Collapse
Affiliation(s)
- Anuja Pant
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Kareena Moar
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India.
| |
Collapse
|
3
|
Wu J, Li L, Cheng Z. System analysis based on T-cell exhaustion-related genes identifies PTPRT as a promising diagnostic and prognostic biomarker for gastric cancer. Sci Rep 2024; 14:21049. [PMID: 39251810 PMCID: PMC11384728 DOI: 10.1038/s41598-024-72135-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
Multiple investigations have demonstrated the crucial involvement of T-cell exhaustion (TEX) in anti-tumor immune response and their strong correlation with prognosis. This study aimed at creating a strong signature using TEX for gastric cancer through bioinformatics analysis and experimental validation. We utilized data from The Cancer Genome Atlas (TCGA) databases to retrieve RNA-seq data from patients with stomach adenocarcinoma (STAD). Genes related to TEX were discovered using gene set variance analysis (GSVA) and weighted gene correlation network analysis (WGCNA). Subsequently, prognostic signature based on TEX was developed using LASSO-Cox analysis. Relationship between key genes and immune cells were examined. Finally, biological function of a key TEX-related gene PTPRT in gastric cancer was verified by in vivo experiment. A total of 29 TEX-related biomarkers were screened by WGCNA and random forest. Among them, five core signatures (PTPRT, CAV2, PPIH, PRDM2, and FGF1), further identified by LASSO-Cox, were considered as strong predictors of prognosis for gastric cancer and associated with immune infiltration. PTPRT gene had the largest number of SNPs, with the most mutation types. In vivo experiments revealed that PTPRT overexpression significantly inhibited tumor malignant progression and accelerated apoptosis through stimulating the secretion of killer cytokines such as TNF-α and IFN-γ. In addition, flow cytometry revealed that PTPRT overexpression alleviated TEX by increasing the abundance of CD8+ T cells, with inhibition of cell surface PD-1 and Tim-3. The predictive prognostic value of TEX gene expression levels was evaluated in patients with gastric cancer, providing a new perspective for precision immuno-oncology studies.
Collapse
Affiliation(s)
- Jianli Wu
- Medical School, Huanghe S&T University, No. 666 Zijingshan South Road, Zhengzhou, 450015, Henan, People's Republic of China
| | - Le Li
- Medical School, Huanghe S&T University, No. 666 Zijingshan South Road, Zhengzhou, 450015, Henan, People's Republic of China
| | - Zhenyun Cheng
- Medical School, Huanghe S&T University, No. 666 Zijingshan South Road, Zhengzhou, 450015, Henan, People's Republic of China.
| |
Collapse
|
4
|
Ma Y, Li X, Zhang J, Zhao X, Lu Y, Shen G, Wang G, Liu H, Hao J. Integrating tertiary lymphoid structure-associated genes into computational models to evaluate prognostication and immune infiltration in pancreatic cancer. J Leukoc Biol 2024; 116:589-600. [PMID: 38484172 DOI: 10.1093/jleuko/qiae067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/03/2024] [Accepted: 02/27/2024] [Indexed: 09/03/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by poor response to all therapeutic modalities and dismal prognosis. The presence of tertiary lymphoid structures (TLSs) in various solid cancers is of crucial prognostic significance, highlighting the intricate interplay between the tumor microenvironment and immune cells aggregation. However, the extent to which TLSs and immune status affect PDAC prognosis remains incompletely understood. Here, we sought to unveil the unique properties of TLSs in PDAC by leveraging both single-cell and bulk transcriptomics, culminating in a risk model that predicts clinical outcomes. We used TLS scores based on a 12-gene (CCL2, CCL3, CCL4, CCL5, CCL8, CCL18, CCL19, CCL21, CXCL9, CXCL10, CXCL11, and CXCL13) and 9-gene (PTGDS, RBP5, EIF1AY, CETP, SKAP1, LAT, CCR6, CD1D, and CD79B) signature, respectively, and examined their distribution in cell clusters of single-cell data from PDAC samples. The markers involved in these clusters were selected to develop a prognostic model using The Cancer Genome Atlas Program database as the training cohort and Gene Expression Omnibus database as the validation cohort. Further, we compared the immune infiltration, drug sensitivity, and enriched and differentially expressed genes between the high- and low-risk groups in our model. Therefore, we established a risk model that has significant implications for the prognostic assessment of PADC patients with remarkable differences in immune infiltration and chemosensitivity between the low- and high-risk groups. This paradigm established by TLS-related cell marker genes provides a prognostic prediction and a panel of novel therapeutic targets for exploring potential immunotherapy.
Collapse
Affiliation(s)
- Ying Ma
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Xuesong Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Jin Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Xiangqin Zhao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Yi Lu
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Guangcong Shen
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Hong Liu
- Second Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, West Huanhu Road, Hexi District, Tianjin 300060, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, West Huanhu Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, West Huanhu Road, Hexi District, Tianjin 300060, China
| | - Jihui Hao
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, West Huanhu Road, Hexi District, Tianjin 300060, China
| |
Collapse
|
5
|
Pant A, Dakal TC, Moar K, Dhabhai B, Arora TK, Sharma NK, Ranga V, Maurya PK. Assessment of MMP14, CAV2, CLU and SPARCL1 expression profiles in endometriosis. Pathol Res Pract 2023; 251:154892. [PMID: 37898038 DOI: 10.1016/j.prp.2023.154892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/30/2023]
Abstract
Endometriotic cells exhibit a notable degree of invasiveness and some characteristics of tissue remodeling underlying lesion formation. In this regard, do matrix metalloproteinases 14 (MMP14) and other related genes such as SPARC-like protein 1 (SPARCL1), caveolin 2 (CAV2), and clusterin (CLU) exert any significant influence in the processes of endometriosis development and pathophysiology is not apparent. We aim to assess whether these genes could serve as potential diagnostic biomarkers in endometriosis. Microarray-based gene expression analysis was performed on total RNA extracted from endometriotic tissue samples treated with and without gonadotropin-releasing hormone agonist (GnRHa). The GnRHa untreated patients were considered the control group. The validation of genes was performed using quantitative real-time polymerase chain reaction (qRT-PCR). qRT-PCR analysis showed significant downregulation in the expression of MMP14 (p = 0.024), CAV2 (p = 0.017), and upregulation of CLU (p = 0.005) in endometriosis patients treated with GnRHa. SPARCL1 did not show any significant (p = 0.30) change in the expression compared to the control group. These data have the potential to contribute to the comprehension of the molecular pathways implicated in the remodeling of the extracellular matrix, which is a vital step for the physiology of the endometrium. Based on the result, it is concluded that changes in the expression of MMP14, CAV2, and CLU post-treatment imply their role in the pathophysiology of endometriosis and may serve as a potential diagnostic biomarker of endometriosis in response to GnRHa treatment in patients with ovarian endometrioma.
Collapse
Affiliation(s)
- Anuja Pant
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Kareena Moar
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Bhanupriya Dhabhai
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Taruna K Arora
- Reproductive Biology and Maternal Child Health Division, Indian Council of Medical Research, New Delhi 110029, India
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk 304022, Rajasthan, India
| | - Vipin Ranga
- Department of Biotechnology-North East Centre for Agricultural Biotechnology (DBT-NECAB), Assam Agricultural University, Jorhat 785013, Assam, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India.
| |
Collapse
|
6
|
Zhu K, Wang B, Li Y, Yu Y, Chen Z, Yue H, Meng Q, Tian D, Liu X, Shen W, Tian Y. CAVIN2/SDPR Functioned as a Tumor Suppressor in Lung Adenocarcinoma from Systematic Analysis of Caveolae-Related Genes and Experimental Validation. J Cancer 2023; 14:2001-2014. [PMID: 37497407 PMCID: PMC10367915 DOI: 10.7150/jca.84567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/20/2023] [Indexed: 07/28/2023] Open
Abstract
Background: Caveolae-Related Genes include caveolins and cavins, which are the main component of the fossa and, play important roles in a variety of physiological and pathological processes. Although increasing evidence indicated that caveolins (CAVs) and cavins (CAVINs) are involved in carcinogenesis and progression, their clinical significance and biological function in lung cancer are still limited. Methods: We investigated the expression of CAVs and CAVINs at transcriptional levels using Oncomine and Gene Expression Profiling Interactive Analysis. The protein and mRNA expression levels of CAVs and CAVINs were determined by the human protein atlas website and our surgically resected samples, respectively. The clinical value of prognostic prediction based on the expression of CAVs and CAVINs was also assessed. cBioPortal, GeneMANIA and STRING were used to analyze the molecular characteristics of CAVs and CAVINs in lung adenocarcinoma (LUAD) comprehensively. Finally, we investigated the effect of CAVIN2/SDPR (serum deprivation protein response) on LUAD cells with biological experiments in vitro. Results: The expression of CAV1/2 and CAVIN1/2/3 were significantly downregulated in LUAD and lung squamous cell carcinoma (LUSC). The patients with high expression of CAV1, CAV2, CAV3, CAVIN1 and CAVIN2/SDPR were tightly correlated with a better prognosis in LUAD, while no statistical significances in LUSC. Further, our results found that CAVIN2/SDPR can be identified as a prognostic biomarker independent of other CAVINs in patients with LUAD. Mechanically, the overexpression of CAVIN2/SDPR inhibited cell proliferation and migration owing to the cell apoptosis induction and cell cycle arrest at S phase in LUAD cells. Conclusions: CAVIN2/SDPR functioned as a tumor suppressor, and was able to serve as prognostic biomarkers in precision medicine of LUAD. Mechanically, overexpression of CAVIN2/SDPR inhibited cell proliferation by inducing cell apoptosis and S phase arrest in LUAD cells.
Collapse
Affiliation(s)
- Keyun Zhu
- Department of Thoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, P. R. China, 315040
| | - Baichuan Wang
- Anhui Medical University Clinical College of Chest, Hefei, Anhui Province, P. R. China, 230022
- Anhui Chest Hospital, Hefei, Anhui Province, P. R. China, 230022
| | - Yingxi Li
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, P. R. China, 300070
| | - Yue Yu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China, 300060
| | - Zhaohui Chen
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China, 300060
| | - Haoran Yue
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China, 300060
| | - Qingxiang Meng
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China, 300060
| | - Dongchen Tian
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China, 300060
| | - Xiaofeng Liu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China, 300060
| | - Weiyu Shen
- Department of Thoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, P. R. China, 315040
| | - Yao Tian
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, P. R. China, 300052
| |
Collapse
|
7
|
Banushi B, Joseph SR, Lum B, Lee JJ, Simpson F. Endocytosis in cancer and cancer therapy. Nat Rev Cancer 2023:10.1038/s41568-023-00574-6. [PMID: 37217781 DOI: 10.1038/s41568-023-00574-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 05/24/2023]
Abstract
Endocytosis is a complex process whereby cell surface proteins, lipids and fluid from the extracellular environment are packaged, sorted and internalized into cells. Endocytosis is also a mechanism of drug internalization into cells. There are multiple routes of endocytosis that determine the fate of molecules, from degradation in the lysosomes to recycling back to the plasma membrane. The overall rates of endocytosis and temporal regulation of molecules transiting through endocytic pathways are also intricately linked with signalling outcomes. This process relies on an array of factors, such as intrinsic amino acid motifs and post-translational modifications. Endocytosis is frequently disrupted in cancer. These disruptions lead to inappropriate retention of receptor tyrosine kinases on the tumour cell membrane, changes in the recycling of oncogenic molecules, defective signalling feedback loops and loss of cell polarity. In the past decade, endocytosis has emerged as a pivotal regulator of nutrient scavenging, response to and regulation of immune surveillance and tumour immune evasion, tumour metastasis and therapeutic drug delivery. This Review summarizes and integrates these advances into the understanding of endocytosis in cancer. The potential to regulate these pathways in the clinic to improve cancer therapy is also discussed.
Collapse
Affiliation(s)
- Blerida Banushi
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Shannon R Joseph
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Benedict Lum
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Jason J Lee
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Fiona Simpson
- Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
8
|
Zhou Y, Guo Y, Wang Y. Identification and validation of a seven-gene prognostic marker in colon cancer based on single-cell transcriptome analysis. IET Syst Biol 2022; 16:72-83. [PMID: 35352485 PMCID: PMC8965382 DOI: 10.1049/syb2.12041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/06/2021] [Accepted: 12/04/2021] [Indexed: 11/25/2022] Open
Abstract
Colon cancer (CC) is one of the most commonly diagnosed tumours worldwide. Single-cell RNA sequencing (scRNA-seq) can accurately reflect the heterogeneity within and between tumour cells and identify important genes associated with cancer development and growth. In this study, scRNA-seq was used to identify reliable prognostic biomarkers in CC. ScRNA-seq data of CC before and after 5-fluorouracil treatment were first downloaded from the Gene Expression Omnibus database. The data were pre-processed, and dimensionality reduction was performed using principal component analysis and t-distributed stochastic neighbour embedding algorithms. Additionally, the transcriptome data, somatic variant data, and clinical reports of patients with CC were obtained from The Cancer Genome Atlas database. Seven key genes were identified using Cox regression analysis and the least absolute shrinkage and selection operator method to establish signatures associated with CC prognoses. The identified signatures were validated on independent datasets, and somatic mutations and potential oncogenic pathways were further explored. Based on these features, gene signatures, and other clinical variables, a more effective predictive model nomogram for patients with CC was constructed, and a decision curve analysis was performed to assess the utility of the nomogram. A prognostic signature consisting of seven prognostic-related genes, including CAV2, EREG, NGFRAP1, WBSCR22, SPINT2, CCDC28A, and BCL10, was constructed and validated. The proficiency and credibility of the signature were verified in both internal and external datasets, and the results showed that the seven-gene signature could effectively predict the prognosis of patients with CC under various clinical conditions. A nomogram was then constructed based on features such as the RiskScore, patients' age, neoplasm stage, and tumor (T), nodes (N), and metastases (M) classification, and the nomogram had good clinical utility. Higher RiskScores were associated with a higher tumour mutational burden, which was confirmed to be a prognostic risk factor. Gene set enrichment analysis showed that high-score groups were enriched in 'cytoplasmic DNA sensing', 'Extracellular matrix receptor interactions', and 'focal adhesion', and low-score groups were enriched in 'natural killer cell-mediated cytotoxicity', and 'T-cell receptor signalling pathways', among other pathways. A robust seven-gene marker for CC was identified based on scRNA-seq data and was validated in multiple independent cohort studies. These findings provide a new potential marker to predict the prognosis of patients with CC.
Collapse
Affiliation(s)
- Yang Zhou
- Medical Oncology Department of Gastrointestinal CancerLiaoning Cancer Hospital & InstituteCancer Hospital of China Medical UniversityLiaoning ProvinceChina
| | - Yang Guo
- Shenyang Tenth People's Hospital (Shenyang Chest Hospital)ShenyangLiaoningP. R. China
| | - Yuanhe Wang
- Medical Oncology Department of Gastrointestinal CancerLiaoning Cancer Hospital & InstituteCancer Hospital of China Medical UniversityLiaoning ProvinceChina
| |
Collapse
|
9
|
Gao L, Zhao R, Liu J, Zhang W, Sun F, Yin Q, Wang X, Wang M, Feng T, Qin Y, Cai W, Li Q, Dong H, Chen X, Xiong X, Liu H, Hu J, Chen W, Han B. KIF15 Promotes Progression of Castration Resistant Prostate Cancer by Activating EGFR Signaling Pathway. Front Oncol 2021; 11:679173. [PMID: 34804913 PMCID: PMC8599584 DOI: 10.3389/fonc.2021.679173] [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: 03/11/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) continues to be a major clinical problem and its underlying mechanisms are still not fully understood. The epidermal growth factor receptor (EGFR) activation is an important event that regulates mitogenic signaling. EGFR signaling plays an important role in the transition from androgen dependence to castration-resistant state in prostate cancer (PCa). Kinesin family member 15 (KIF15) has been suggested to be overexpressed in multiple malignancies. Here, we demonstrate that KIF15 expression is elevated in CRPC. We show that KIF15 contributes to CRPC progression by enhancing the EGFR signaling pathway, which includes complex network intermediates such as mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/AKT pathways. In CRPC tumors, increased expression of KIF15 is positively correlated with EGFR protein level. KIF15 binds to EGFR, and prevents EGFR proteins from degradation in a Cdc42-dependent manner. These findings highlight the key role of KIF15 in the development of CRPC and rationalize KIF15 as a potential therapeutic target.
Collapse
Affiliation(s)
- Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ru Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Junmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenbo Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feifei Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianshuo Yin
- School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Meng Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yiming Qin
- College of Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Wenjie Cai
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianni Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanchen Dong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueqing Chen
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xueting Xiong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hui Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
10
|
Richter A, Fichtner A, Joost J, Brockmeyer P, Kauffmann P, Schliephake H, Hammerstein-Equord A, Kueffer S, Urlaub H, Oellerich T, Ströbel P, Bohnenberger H, Bremmer F. Quantitative proteomics identifies biomarkers to distinguish pulmonary from head and neck squamous cell carcinomas by immunohistochemistry. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2021; 8:33-47. [PMID: 34647699 PMCID: PMC8682946 DOI: 10.1002/cjp2.244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/23/2021] [Accepted: 09/01/2021] [Indexed: 12/16/2022]
Abstract
The differentiation between a pulmonary metastasis and a newly developed squamous cell carcinoma of the lung in patients with prior head and neck squamous cell carcinoma (HNSCC) is difficult due to a lack of biomarkers but is crucially important for the prognosis and therapy of the affected patient. By using high‐resolution mass spectrometry in combination with stable isotope labelling by amino acids in cell culture, we identified 379 proteins that are differentially expressed in squamous cell carcinomas of the lung and the head and neck. Of those, CAV1, CAV2, LGALS1, LGALS7, CK19, and UGDH were tested by immunohistochemistry on 194 tissue samples (98 lung and 96 HNSCCs). The combination of CAV1 and LGALS7 was able to distinguish the origin of the squamous cell carcinoma with high accuracy (area under the curve 0.876). This biomarker panel was tested on a cohort of 12 clinically classified lung tumours of unknown origin after HNSCC. Nine of those tumours were immunohistochemically classifiable.
Collapse
Affiliation(s)
- Annika Richter
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Alexander Fichtner
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Jasmin Joost
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Philipp Brockmeyer
- Department of Oral and Maxillofacial Surgery, University Medical Centre Göttingen, Göttingen, Germany
| | - Philipp Kauffmann
- Department of Oral and Maxillofacial Surgery, University Medical Centre Göttingen, Göttingen, Germany
| | - Henning Schliephake
- Department of Oral and Maxillofacial Surgery, University Medical Centre Göttingen, Göttingen, Germany
| | | | - Stefan Kueffer
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Bioanalytics, Institute for Clinical Chemistry, University Medical Centre Göttingen, Göttingen, Germany
| | - Thomas Oellerich
- Department of Medicine II, Haematology/Oncology, Goethe University, Frankfurt, Germany.,German Cancer Research Centre and German Cancer Consortium, Heidelberg, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | | | - Felix Bremmer
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| |
Collapse
|
11
|
Tian Y, Liu X, Hu J, Zhang H, Wang B, Li Y, Fu L, Su R, Yu Y. Integrated Bioinformatic Analysis of the Expression and Prognosis of Caveolae-Related Genes in Human Breast Cancer. Front Oncol 2021; 11:703501. [PMID: 34513683 PMCID: PMC8427033 DOI: 10.3389/fonc.2021.703501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/31/2021] [Indexed: 12/21/2022] Open
Abstract
Caveolae-related genes, including CAVs that encodes caveolins and CAVINs that encodes caveolae-associated proteins cavins, have been identified for playing significant roles in a variety of biological processes including cholesterol transport and signal transduction, but evidences related to tumorigenesis and cancer progression are not abundant to correlate with clinical characteristics and prognosis of patients with cancer. In this study, we investigated the expression of these genes at transcriptional and translational levels in patients with breast cancer using Oncomine, Gene Expression Profiling Interactive Analysis (GEPIA), cBioPortal databases, and immunohistochemistry of the patients in our hospital. Prognosis of patients with breast cancer based on the expressions of CAVs and CAVINs was summarized using Kaplan-Meier Plotter with their correlation to different subtyping. The relevant molecular pathways of these genes were further analyzed using Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database and Gene Set Enrichment Analysis (GSEA). Results elucidated that expression levels of CAV1, CAV2, CAVIN1, CAVIN2, and CAVIN3 were significantly lower in breast cancer tissues than in normal samples, while the expression level of CAVIN2 was correlated with advanced tumor stage. Furthermore, investigations on survival of patients with breast cancer indicated outstanding associations between prognosis and CAVIN2 levels, especially for the patients with estrogen receptor positive (ER+) breast cancer. In conclusion, our investigation indicated CAVIN2 is a potential therapeutic target for patients with ER+ breast cancer, which may relate to functions of cancer cell surface receptors and adhesion molecules.
Collapse
Affiliation(s)
- Yao Tian
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaofeng Liu
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jing Hu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Huan Zhang
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Baichuan Wang
- Anhui Medical University Clinical College of Chest, Hefei, China.,Department of Thoracic Surgery, Anhui Chest Hospital, Hefei, China
| | - Yingxi Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Li Fu
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ran Su
- School of Computer Software, College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Yue Yu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
12
|
Xu Y, Zuo W, Wang X, Zhang Q, Gan X, Tan N, Jia W, Liu J, Li Z, Zhou B, Zhao D, Xie Z, Tan Y, Zheng S, Liu C, Li H, Chen Z, Yang X, Huang Z. Deciphering the effects of PYCR1 on cell function and its associated mechanism in hepatocellular carcinoma. Int J Biol Sci 2021; 17:2223-2239. [PMID: 34239351 PMCID: PMC8241733 DOI: 10.7150/ijbs.58026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/16/2021] [Indexed: 12/28/2022] Open
Abstract
Overexpression of pyrroline-5-carboxylate reductase 1 (PYCR1) has been associated with the development of certain cancers; however, no studies have specifically examined the role of PYCR1 in hepatocellular carcinoma (HCC). Based on The Cancer Genome Atlas expression array and meta-analysis conducted using the Gene Expression Omnibus database, we determined that PYCR1 was upregulated in HCC compared to adjacent nontumor tissues (P < 0.05). These data were verified using quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry analysis. Additionally, patients with low PYCR1 expression showed a higher overall survival rate than patients with high PYCR1 expression. Furthermore, PYCR1 overexpression was associated with the female sex, higher levels of alpha-fetoprotein, advanced clinical stages (III and IV), and a younger age (< 45 years old). Silencing of PYCR1 inhibited cell proliferation, invasive migration, epithelial-mesenchymal transition, and metastatic properties in HCC in vitro and in vivo. Using RNA sequencing and bioinformatics tools for data-dependent network analysis, we found binary relationships among PYCR1 and its interacting proteins in defined pathway modules. These findings indicated that PYCR1 played a multifunctional role in coordinating a variety of biological pathways involved in cell communication, cell proliferation and growth, cell migration, a mitogen-activated protein kinase cascade, ion binding, etc. The structural characteristics of key pathway components and PYCR1-interacting proteins were evaluated by molecular docking, and hotspot analysis showed that better affinities between PYCR1 and its interacting molecules were associated with the presence of arginine in the binding site. Finally, a candidate regulatory microRNA, miR-2355-5p, for PYCR1 mRNA was discovered in HCC. Overall, our study suggests that PYCR1 plays a vital role in HCC pathogenesis and may potentially serve as a molecular target for HCC treatment.
Collapse
Affiliation(s)
- Yanzhen Xu
- Department of pathology, Affiliated hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Department of Pathology, Affiliated Hangzhou First People's Hospital, School of Medicine, Zhejiang University, 310000, Hangzhou, China
| | - Wenpu Zuo
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Medical Scientific Research Center, Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Xiao Wang
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Qinle Zhang
- Genetic and metabolic central laboratory, the maternal and children's health hospital of Guangxi, Nanning, 530000, Guangxi, China
| | - Xiang Gan
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Ning Tan
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Wenxian Jia
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Jiayi Liu
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhouquan Li
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Bo Zhou
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Dong Zhao
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhibin Xie
- Department of Urology, the Five Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Yanjun Tan
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Shengfeng Zheng
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Chengwu Liu
- Department of Pathophysiology, Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Hongtao Li
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhijian Chen
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| | - Xiaoli Yang
- Scientific Research Center, Guilin Medical University, Guilin, 541001, Guangxi, China
- Guangxi Health Commission Key Laboratory of Disease Proteomics Research, Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Zhaoquan Huang
- Department of pathology, Affiliated hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
- Department of Pathology, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, Guangxi, China
| |
Collapse
|
13
|
Senturk A, Sahin AT, Armutlu A, Kiremit MC, Acar O, Erdem S, Bagbudar S, Esen T, Tuncbag N, Ozlu N. Quantitative Proteomics Identifies Secreted Diagnostic Biomarkers as well as Tumor-Dependent Prognostic Targets for Clear Cell Renal Cell Carcinoma. Mol Cancer Res 2021; 19:1322-1337. [PMID: 33975903 DOI: 10.1158/1541-7786.mcr-21-0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/12/2021] [Accepted: 04/30/2021] [Indexed: 11/16/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third most common and most malignant urological cancer, with a 5-year survival rate of 10% for patients with advanced tumors. Here, we identified 10,160 unique proteins by in-depth quantitative proteomics, of which 955 proteins were significantly regulated between tumor and normal adjacent tissues. We verified four putatively secreted biomarker candidates, namely, PLOD2, FERMT3, SPARC, and SIRPα, as highly expressed proteins that are not affected by intratumor and intertumor heterogeneity. Moreover, SPARC displayed a significant increase in urine samples of patients with ccRCC, making it a promising marker for the detection of the disease in body fluids. Furthermore, based on molecular expression profiles, we propose a biomarker panel for the robust classification of ccRCC tumors into two main clusters, which significantly differed in patient outcome with an almost three times higher risk of death for cluster 1 tumors compared with cluster 2 tumors. Moreover, among the most significant clustering proteins, 13 were targets of repurposed inhibitory FDA-approved drugs. Our rigorous proteomics approach identified promising diagnostic and tumor-discriminative biomarker candidates which can serve as therapeutic targets for the treatment of ccRCC. IMPLICATIONS: Our in-depth quantitative proteomics analysis of ccRCC tissues identifies the putatively secreted protein SPARC as a promising urine biomarker and reveals two molecular tumor phenotypes.
Collapse
Affiliation(s)
- Aydanur Senturk
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Ayse T Sahin
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Ayse Armutlu
- Department of Pathology, Koc University School of Medicine, Istanbul, Turkey
| | - Murat C Kiremit
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Omer Acar
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Selcuk Erdem
- Department of Urology, Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Sidar Bagbudar
- Department of Pathology, Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Tarik Esen
- Department of Urology, Koc University School of Medicine, Istanbul, Turkey
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey.,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey. .,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| |
Collapse
|
14
|
Tu Z, He X, Zeng L, Meng D, Zhuang R, Zhao J, Dai W. Exploration of Prognostic Biomarkers for Lung Adenocarcinoma Through Bioinformatics Analysis. Front Genet 2021; 12:647521. [PMID: 33968130 PMCID: PMC8100590 DOI: 10.3389/fgene.2021.647521] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/30/2021] [Indexed: 12/30/2022] Open
Abstract
With the development of computer technology, screening cancer biomarkers based on public databases has become a common research method. Here, an eight-gene prognostic model, which could be used to judge the prognosis of patients with lung adenocarcinoma (LUAD), was developed through bioinformatics methods. This study firstly used several gene datasets from GEO database to mine differentially expressed genes (DEGs) in LUAD tissue and healthy tissue via joint analysis. Later, enrichment analysis for the DEGs was performed, and it was found that the DEGs were mainly activated in pathways involved in extracellular matrix, cell adhesion, and leukocyte migration. Afterward, a TCGA cohort was used to perform univariate Cox, least absolute shrinkage and selection operator method, and multivariate Cox regression analyses for the DEGs, and a prognostic model consisting of eight genes (GPX3, TCN1, ASPM, PCP4, CAV2, S100P, COL1A1, and SPOK2) was established. Receiver operation characteristic (ROC) curve was then used to substantiate the diagnostic efficacy of the prognostic model. The survival significance of signature genes was verified through the GEPIA database, and the results exhibited that the risk coefficients of the eight genes were basically congruous with the effects of these genes on the prognosis in the GEPIA database, which suggested that the results were accurate. Finally, combined with clinical characteristics of patients, the diagnostic independence of the prognostic model was further validated through univariate and multivariate regression, and the results indicated that the model had independent prognostic value. The overall finding of the study manifested that the eight-gene prognostic model is closely related to the prognosis of LUAD patients, and can be used as an independent prognostic indicator. Additionally, the prognostic model in this study can help doctors make a better diagnosis in treatment and ultimately benefit LUAD patients.
Collapse
Affiliation(s)
- Zhengliang Tu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangfeng He
- Department of Thoracic Surgery, Zhuji People's Hospital, Zhuji, China
| | - Liping Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di Meng
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runzhou Zhuang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangang Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wanrong Dai
- Department of Pharmacy, The First Affiliated Hospital, College of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Xu L, Zhang B, Li W. Downregulated expression levels of USP46 promote the resistance of ovarian cancer to cisplatin and are regulated by PUM2. Mol Med Rep 2021; 23:263. [PMID: 33576437 PMCID: PMC7893694 DOI: 10.3892/mmr.2021.11902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OC) is a major contributor to cancer‑related mortality in women. Despite numerous drugs being available for the treatment and improving the prognosis of OC, resistance to clinical chemotherapy remains a major obstacle for the treatment of advanced OC. Therefore, determining how to reverse the chemoresistance of OC has become a research hotspot in recent years. The present study aimed to reveal the potential mechanism of OC chemoresistance. Reverse transcription‑quantitative PCR and western blot analysis were performed to detect the expression levels of Ubiquitin‑specific peptidase 46 (USP46) and Pumilio 2 (PUM2) in OC. Cell viability and apoptosis were evaluated by Cell Counting Kit‑8 assay and flow cytometry, respectively. The association between USP46 and PUM2 was assessed by RNA immunoprecipitation. The results of the present study revealed that the expression levels of USP46 which is associated with tumor progression, was downregulated, while PUM2 expression levels were upregulated in cisplatin (DDP)‑resistant OC cells and patient tissues. The downregulation of USP46 expression levels in SKOV3 cells significantly inhibited cell apoptosis and increased cell viability. In SKOV3/DDP cells, the upregulation of USP46 expression levels notably suppressed cell viability and increased cell apoptosis. The results of the RNA immunoprecipitation chip assay demonstrated that PUM2 bound to USP46 and regulated its expression. Furthermore, following the knockdown of USP46 expression, the mRNA and protein expression levels of the cell apoptosis‑related protein, Bcl‑2, were upregulated, whereas the expression levels of caspase‑3, caspase‑9 and Bax were significantly downregulated. In addition, phosphorylated AKT expression levels were notably upregulated. Following the overexpression of USP46 in SKOV3/DDP cells, the opposite trends were observed. In SKOV3 cells, the knockdown of PUM2 could reverse the DDP resistance induced by small interfering RNA‑USP46 as the expression levels of Bcl‑2 were downregulated whereas those of caspase‑3, caspase‑9 and Bax were upregulated compared with the small interfering‑USP46 group. Similarly, in SKOV3/DDP cells, the overexpression of PUM2 could reverse DDP sensitivity induced by the overexpression of USP46. In conclusion, the findings of the present study suggested that the downregulation of USP46 expression levels may promote DDP resistance in OC, which may be regulated by PUM2. Therefore, targeting PUM2/USP46 may be an effective way to reverse DDP resistance in OC.
Collapse
Affiliation(s)
- Lei Xu
- Department of Gynecology, People's Hospital of Qingdao West Coast New Area, Qingdao, Shandong 266400, P.R. China
| | - Bin Zhang
- Department of Surgery, People's Hospital of Qingdao West Coast New Area, Qingdao, Shandong 266400, P.R. China
| | - Wenlan Li
- Department of Outpatient Department, People's Hospital of Qingdao West Coast New Area, Qingdao, Shandong 266400, P.R. China
| |
Collapse
|
16
|
RNA sequencing and bioinformatics analysis of human lens epithelial cells in age-related cataract. BMC Ophthalmol 2021; 21:152. [PMID: 33771123 PMCID: PMC7995707 DOI: 10.1186/s12886-021-01915-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/16/2021] [Indexed: 01/19/2023] Open
Abstract
Background Age-related cataract (ARC) is the main cause of blindness in older individuals but its specific pathogenic mechanism is unclear. This study aimed to identify differentially expressed genes (DEGs) associated with ARC and to improve our understanding of the disease mechanism. Methods Anterior capsule samples of the human lens were collected from ARC patients and healthy controls and used for RNA sequencing to detect DEGs. Identified DEGs underwent bioinformatics analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Subsequently, reverse transcription quantitative RT-qPCR was used to validate the different expression levels of selected genes. Results A total of 698 up-regulated DEGs and 414 down-regulated DEGs were identified in ARC patients compared with controls by transcriptome analysis. Through GO and KEGG bioinformatics analysis, the functions of significantly DEGs and their possible molecular mechanisms were determined. Sequencing results were verified by RT-qPCR as being accurate and reliable. Conclusions This study identified several genes associated with ARC, which improves our knowledge of the disease mechanism.
Collapse
|
17
|
Zhang X, Yan L, Yuan X, Bai T, Zhang L, Han S. Rapid exacerbation featuring acute leukemoid reaction after retrolaparoscopic nephrectomy: a rare case report of renal cell carcinoma with postoperative comprehensive genomic profiling. World J Surg Oncol 2020; 18:155. [PMID: 32631368 PMCID: PMC7339471 DOI: 10.1186/s12957-020-01926-4] [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: 11/06/2019] [Accepted: 06/18/2020] [Indexed: 11/24/2022] Open
Abstract
Background Rapid lethal exacerbation and recurrence featuring acute leukemoid reaction (ALR) after retrolaparoscopic radical nephrectomy (RN) is a relatively rare clinical incident. Performing the reoperation for the patient and analyzing the tissue-based genetic mutation information postoperatively are a skill-demanding and meaningful task, which have been even more rarely reported. Case presentation We present a case with a large right renal mass (13.0 × 10.0 × 8.0 cm). This 71-year-old male patient underwent the retrolaparoscopic RN in our department. The operation was technically precise and successful with final pathological diagnosis of hybrid (clear cell and papillary type) renal cell carcinoma (RCC). However, 10 days after the patient was discharged, he was readmitted with the chief complaint of high fever with severe right flank pain. CT scanning revealed that right retroperitoneal hematoma and the blood routine showed the dramatic elevation of white blood cell count (WBC). Even though the immediate broad-spectrum antibiotics were administered without delay and subsequent percutaneous puncturing and drainage was performed, the patient’s condition still exacerbated rapidly. In spite of the reoperation of hematoma evacuation, the patient died of multiple organ failure 10 days after the reoperation. The pathological result of reoperation showed the necrotic and hematoma tissue blended with RCC tumor cells (nuclear grading III), and both of the postoperative tissue-originated comprehensive genomic profiling by using the specimens from the RN and reoperation respectively indicated significant mutations of some oncogenes which might have potential relevance with ALR. Besides, both of the immunohistochemical (IHC) staining results from primary surgical renal mass and reoperative resected tissue revealed the positive expressions of granulocyte colony-stimulating factor (G-CSF). Conclusions ALR may be a predictor of poor prognosis in patients with RCC, and comprehensive genomic profiling as well as the alterative expression of G-CSF can help to provide potential valuable genetic etiological information and evidence for guiding the potential effective molecular-targeting therapy.
Collapse
Affiliation(s)
- Xuhui Zhang
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China.
| | - Lijuan Yan
- Shanxi Cancer Institute, Shanxi Cancer Hospital, Taiyuan, 030000, Shanxi, China
| | - Xiaobin Yuan
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China
| | - Tao Bai
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Lei Zhang
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China
| | - Shuaihong Han
- Department of Urology, First Hospital of Shanxi Medical University, No. 85, Jiefangnan Road, Taiyuan, 030001, Shanxi, China
| |
Collapse
|
18
|
Liu C, Liu L, Wang K, Li XF, Ge LY, Ma RZ, Fan YD, Li LC, Liu ZF, Qiu M, Hao YC, Shi ZF, Xia CY, Strååt K, Huang Y, Ma LL, Xu D. VHL-HIF-2α axis-induced SMYD3 upregulation drives renal cell carcinoma progression via direct trans-activation of EGFR. Oncogene 2020; 39:4286-4298. [PMID: 32291411 DOI: 10.1038/s41388-020-1291-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 03/30/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022]
Abstract
It has been well established that the von Hippel-Lindau/hypoxia-inducible factor α (VHL-HIFα) axis and epidermal growth factor receptor (EGFR) signaling pathway play a critical role in the pathogenesis and progression of renal cell carcinoma (RCC). However, few studies have addressed the relationship between the two oncogenic drivers in RCC. SET and MYND domain-containing protein 3 (SMYD3) is a histone methyltransferase involved in gene transcription and oncogenesis, but its expression and function in RCC remain unclear. In the present study, we found that SMYD3 expression was significantly elevated in RCC tumors and correlated with advanced tumor stage, histological and nuclear grade, and shorter survival. Depletion of SMYD3 inhibited RCC cell proliferation, colony numbers, and xenograft tumor formation, while promoted apoptosis. Mechanistically, SMYD3 cooperates with SP1 to transcriptionally promote EGFR expression, amplifying its downstream signaling activity. TCGA data analyses revealed a significantly increased SMYD3 expression in primary RCC tumors carrying the loss-of-function VHL mutations. We further showed that HIF-2α can directly bind to the SMYD3 promoter and subsequently induced SMYD3 transcription and expression. Taken together, we identify the VHL/HIF-2α/SMYD3 signaling cascade-mediated EGFR hyperactivity through which SMYD3 promotes RCC progression. Our study suggests that SMYD3 is a potential therapeutic target and prognostic factor in RCC.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Urology, Peking University Third Hospital, Beijing, China.
| | - Li Liu
- School of Nursing, Beijing University of Chinese Medicine, Beijing, China
| | - Kun Wang
- Key Lab for Cancer Prevention and Therapy, National Clinical Research Centre for Cancer, Department of Urology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiao-Feng Li
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Li-Yuan Ge
- Department of Urology, Peking University Third Hospital, Beijing, China.,Department of Urology, The People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang, China
| | - Run-Zhuo Ma
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yi-Dong Fan
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Lu-Chao Li
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Zheng-Fang Liu
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Min Qiu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yi-Chang Hao
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Zhen-Feng Shi
- Department of Urology, The People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang, China
| | - Chuan-You Xia
- Department of Medicine, Division of Hematology, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Klas Strååt
- Department of Medicine, Division of Hematology, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | - Yi Huang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Lu-Lin Ma
- Department of Urology, Peking University Third Hospital, Beijing, China.
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden.,Karolinska Institute-Shandong University Collaborative Laboratory for Cancer and Stem Cell Research, Jinan, China
| |
Collapse
|
19
|
Zhu C, Na N, Sheng H, Feng B, Wang H, Zhu P, Zhang W, Zhang M, Deng Z. Ginkgolic acid inhibits the growth of renal cell carcinoma cells via inactivation of the EGFR signaling pathway. Exp Ther Med 2020; 19:2949-2956. [PMID: 32256780 PMCID: PMC7086188 DOI: 10.3892/etm.2020.8570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022] Open
Abstract
Renal cell carcinoma (RCC) is one of the most common urological malignancies occurring in adult human kidneys worldwide. Recent research on antitumor drugs has focused on plant extracts, a class of compounds that play critical roles in cancer treatment. The present study aimed to investigate the potential antitumor effect of ginkgolic acid (GA) in RCC. Transwell invasion assay, cell counting kit-8 assay and flow cytometry were used to measure cell migration, cell viability and apoptosis, respectively. A network pharmacology approach was applied to identify pathway information, combining molecular docking techniques to screen for key target information. In the present study, GA inhibited the viability and proliferation of RCC cells (786-O and A498), both in vitro and in vivo, via G1 arrest. GA also reduced RCC cell invasion and migration. In addition, the epidermal growth factor receptor (EGFR) was identified as a critical target protein of GA, which significantly inactivated EGFR signaling in RCC (P<0.05). Collectively, the present study provided evidence that GA exerts its anticancer function by directly targeting the EGFR signaling pathway, revealing the potential of GA therapy for RCC.
Collapse
Affiliation(s)
- Chao Zhu
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Na Na
- Department of Outpatients, 900th Hospital of The Joint Logistics Support Force (People's Liberation Army), Fuzhou, Fujian 350000, P.R. China
| | - Haibo Sheng
- Department of Urology, Airforce Medical Center (People's Liberation Army), Beijing 100142, P.R. China
| | - Bing Feng
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Hao Wang
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Ping Zhu
- Department of Pathology, 971st Navy Hospital of PLA, Qingdao, Shandong 266071, P.R. China
| | - Wei Zhang
- Department of Pathology, 971st Navy Hospital of PLA, Qingdao, Shandong 266071, P.R. China
| | - Meina Zhang
- Department of Pathology, 971st Navy Hospital of PLA, Qingdao, Shandong 266071, P.R. China
| | - Zhen Deng
- Department of Urology, 900th Hospital of The Joint Logistics Support Force (People's Liberation Army), Fuzhou, Fujian 350000, P.R. China
| |
Collapse
|
20
|
Wang G, Huang Y, Yang F, Tian X, Wang K, Liu L, Fan Y, Li X, Li L, Shi B, Hao Y, Xia C, Nie Q, Xin Y, Shi Z, Ma L, Xu D, Liu C. High expression of SMYD3 indicates poor survival outcome and promotes tumour progression through an IGF-1R/AKT/E2F-1 positive feedback loop in bladder cancer. Aging (Albany NY) 2020; 12:2030-2048. [PMID: 32007952 PMCID: PMC7041758 DOI: 10.18632/aging.102718] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
Abstract
The AKT/mTOR pathway is critical for bladder cancer (BC) pathogenesis and is hyper-activated during BC progression. In the present study, we identified a novel positive feedback loop involving oncogenic factors histone methyltransferase SMYD3, insulin-like growth factor-1 receptor (IGF-1R), AKT, and E2F-1. SMYD3 expression was significantly up-regulated in BC tumors and positively associated with histological grade, lymph node metastasis, and shorter patient survival. Depletion of SMYD3 inhibited BC cell proliferation, colony formation, migration, invasion, and xenograft tumor growth. Mechanistically, SMYD3 inhibition led to the diminished AKT/mTOR signaling activity, thereby triggering deleterious effects on BC cells. Furthermore, SMYD3 directly activates the expression of IGF-1R, a critical activator of AKT in BC, by inducing hyper-methylation of histone H3-K4 and subsequent chromatin remodeling in the IGF-1R promoter region. On the other hand, E2F-1, a downstream factor of the AKT pathway, binds to the E2F-1 binding motifs at the SMYD3 promoter and consequently induces SMYD3 transcription and expression. Thus, SMYD3/IGF-1R/AKT/E2F-1 forms a positive feedback loop leading to the hyper-activated AKT signaling. Our findings provide not only profound insights into SMYD3-mediated oncogenic activity but also present a unique avenue for treating BC by directly disrupting this signaling circuit.
Collapse
Affiliation(s)
- Guoliang Wang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yi Huang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Feilong Yang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Xiaojun Tian
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Kun Wang
- Department of Urology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Centre for Cancer, Key Lab for Cancer Prevention and Therapy, Tianjin, China
| | - Li Liu
- School of Nursing, Beijing University of Chinese Medicine, Beijing, China
| | - Yidong Fan
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Xiaofeng Li
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Luchao Li
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Benkang Shi
- Department of Urology, Shandong University Qilu Hospital, Jinan, China
| | - Yichang Hao
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Chuanyou Xia
- Department of Medicine, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna and Karolinska Institutet, Stockholm, Sweden
| | - Qingsheng Nie
- Department of Urology, The Central Hospital of Zibo, Zibo, China
| | - Yue Xin
- Department of Urology, Chifeng University Second Hospital, Chifeng, China
| | - Zhenfeng Shi
- Department of Urology, The People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang, China
| | - Lulin Ma
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Dawei Xu
- Department of Medicine, Bioclinicum and Centre for Molecular Medicine, Karolinska University Hospital Solna and Karolinska Institutet, Stockholm, Sweden.,Karolinska Institute-Shandong University Collaborative Laboratory for Cancer and Stem Cell Research, Jinan, China
| | - Cheng Liu
- Department of Urology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
21
|
Zhu J, Wu K, Lin Z, Bai S, Wu J, Li P, Xue H, Du J, Shen B, Wang H, Liu Y. Identification of susceptibility gene mutations associated with the pathogenesis of familial nonmedullary thyroid cancer. Mol Genet Genomic Med 2019; 7:e1015. [PMID: 31642198 PMCID: PMC6900395 DOI: 10.1002/mgg3.1015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/24/2019] [Accepted: 10/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background Familial nonmedullary thyroid cancer (FNMTC) accounts for approximately 3%–9% of all thyroid cancers; however, the mechanisms underlying FNMTC remain unclear. Environmental and genetic (especially genetic mutation) factors may play important roles in FNMTC etiology, development, and pathogenesis. Methods Three affected members, including two first‐degree relatives, and three healthy members of a family with FNMTC were studied. We performed whole‐exome and targeted gene sequencing to identify gene mutations that may be associated with FNMTC pathogenesis. The results were analyzed using Exome Aggregation Consortium data and the Genome Aggregation Database and further validated using Sanger sequencing. Results Of 28 pivotal genes with rare nonsynonymous mutations found, 7 were identified as novel candidate FNMTC pathogenic genes (ANO7, CAV2, KANK1, PIK3CB, PKD1L1, PTPRF, and RHBDD2). Among them, three genes (PIK3CB, CAV2, and KANK1) are reportedly involved in tumorigenesis through the PI3K/Akt signaling pathway. Conclusion We identified seven pathogenic genes in affected members of a family with FNMTC. The PI3K/Akt signaling pathway is thought to be closely related to the development of FNMTC, and three of the susceptibility genes identified herein are associated with this pathway. These findings expand our understanding of FNMTC pathogenesis and underscore PI3K/Akt pathology as a potential therapy target.
Collapse
Affiliation(s)
- Junwei Zhu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Kaile Wu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhangying Lin
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Suwen Bai
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Jing Wu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Peikun Li
- Department of General surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Haowei Xue
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juan Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Huiyin Wang
- Department of Pathology, Microbiology & Immunology, Vanderbilt Children's Hospital, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yehai Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
22
|
Peng J, Chen XL, Cheng HZ, Xu ZY, Wang H, Shi ZZ, Liu J, Ning XG, Peng H. Silencing of KCNK15-AS1 inhibits lung cancer cell proliferation via upregulation of miR-202 and miR-370. Oncol Lett 2019; 18:5968-5976. [PMID: 31788071 PMCID: PMC6865154 DOI: 10.3892/ol.2019.10944] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the most common cause of cancer-associated mortality globally. Long non-coding RNAs (lncRNAs) are transcripts with a length of >200 nucleotides, which are not translated into proteins. Growing evidence has indicated that certain lncRNAs are associated with various biological processes in cancer. However, the functions of KCNK15 and WISP2 antisense RNA 1 (KCNK15-AS1) in lung cancer carcinogenesis and progression have remained elusive. The present study indicated that KCNK15-AS1 was overexpressed in lung adenocarcinoma tissues compared with paracancerous normal tissues, and the high expression of KCNK15-AS1 was significantly associated with poor prognosis compared with the patients with low expression (P<0.001). Furthermore, the knockdown of KCNK15-AS1 was performed in A549 and H460 lung cancer cells with small interfering RNA, resulting in a significant inhibition of the proliferation, a decrease in the mRNA and protein expression of cyclin D1 (CCND1) and epidermal growth factor receptor (EGFR), in addition to the phosphorylation of protein kinase B, with a concomitant increase in the expression of microRNA (miR)-202 and miR-370 compared with negative control group. Rescue experiments demonstrated that the inhibition of miR-202 or miR-370 partially recovered the EGFR and CCND1 expression and the proliferation rates, which were reduced by KCNK15-AS1 silencing. In conclusion, these results suggested that KCNK15-AS1 functions as an oncogene via regulating the miR-202/miR-370/EGFR axis in lung cancer and may provide a potential target for lung cancer treatment.
Collapse
Affiliation(s)
- Jun Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Xin-Long Chen
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Hong-Zhong Cheng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Zhe-Yuan Xu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Han Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Zhi-Zhou Shi
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Jun Liu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Xian-Gu Ning
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Hao Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
23
|
Gui D, Peng W, Jiang W, Huang G, Liu G, Ye Z, Wang Y, Xu Z, Fu J, Luo S, Zhao Y. Ubiquitin-specific peptidase 46 (USP46) suppresses renal cell carcinoma tumorigenesis through AKT pathway inactivation. Biochem Biophys Res Commun 2019; 519:689-696. [PMID: 31542232 DOI: 10.1016/j.bbrc.2019.09.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022]
Abstract
USP46, a member of the ubiquitin-specific protease family, plays essential roles in cancer cell proliferation and metastasis and is used as a candidate target for cancer therapeutics. However, the effects of USP46 on renal cell carcinoma (RCC) and its underlying molecular mechanism remain unknown. In this study, the predictive and prognostic relevance of USP46 in RCC, patient-derived primary tissues, and normal liver tissues obtained from the TCGA dataset were analyzed for the USP46 mRNA levels or prognostic relevance. Gain-of-function or loss-of-function assays were used to evaluate the vital roles of USP46 in tumor cell proliferation and cell migration. As a result, the USP46 expression level in RCC is highly decreased compared to normal tissues, and the Kaplan-Meier curve showed that USP46 high expression patients had good prognoses. Functionally, the forced expression of USP46 significantly restrained tumor cell proliferation, colony formation, and cell migration. The shRNA mediated USP46 knockdown cells exhibited the opposite results. We further showed that ectopically expressed USP46 obviously inhibited the AKT signaling pathway in cancer cells, while USP46 depletion caused a dramatic increase in AKT activity reflected by phosphorylation in the serine and threonine residues of AKT or downstream p70S6K1. Importantly, MK2206, a specific AKT inhibitor, completely counteracted the effects on cell proliferation, cell migration, and AKT activity in the USP46 depletion cells. We thus revealed a novel mechanism of USP46 regulation in RCC, and our data indicate that USP46 is a tumor suppressor in RCC via AKT signaling pathway inactivation.
Collapse
Affiliation(s)
- Dingwen Gui
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China.
| | - Wei Peng
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Weidong Jiang
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Geng Huang
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Gang Liu
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Zhihua Ye
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Yang Wang
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Zuwei Xu
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Jinlun Fu
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Shuai Luo
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| | - Yunfei Zhao
- Department of Urology, Huangshi Central Hospital (Affiliated Hospital of Hubei Polytechnic University), Edong Healthcare Group, Huangshi, Hubei, 435000, PR China; Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, Huangshi, Hubei, 435000, PR China
| |
Collapse
|
24
|
Wang L, Chen S, Zhang J, Mao S, Mao W, Zhang W, Guo Y, Wu Y, Wang R, Yan Y, Yao X. Suppressed OGT expression inhibits cell proliferation and modulates EGFR expression in renal cell carcinoma. Cancer Manag Res 2019; 11:2215-2223. [PMID: 30962710 PMCID: PMC6433112 DOI: 10.2147/cmar.s190642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose O-linked N-acetylglucosamine (O-GlcNAc or O-GlcNAcylation) is a post-translational modification, which plays a vital role in the progression of various cancers. The purpose of the present study was to assess O-GlcNAcylation in human renal cell carcinoma (RCC). Methods O-GlcNAcylation levels and O-GlcNAc-transferase (OGT) expression in human RCC cell lines and 10 paired clinical tissues were detected by Western blot and Immunohistochemis-try. Then, the effects of O-GlcNAcylation on RCC cell proliferation in vitro were investigated by Cell Counting Kit-8 assay. A xenograft assay was performed to assess the in vivo effects of OGT knockdown in RCC cells. Cell apoptosis and cell cycle assays were performed by flow cytometry. Co-immunoprecipitation assays were used to assess epidermal growth factor receptor (EGFR) O-GlcNAcylation and the interaction between OGT and EGFR. Results O-GlcNAcylation levels and OGT expression were increased in RCC, and the high amounts correlated with poor patient prognosis. OGT knockdown significantly suppressed RCC cell proliferation in vitro and in vivo. Notably, EGFR was modulated by O-GlcNAcylation and directly interacted with OGT. Conclusion These findings provide novel insights into the oncogenic roles of O-GlcNAcylation and OGT in the development of RCC, indicating that OGT might be used as a target for RCC therapy in the future.
Collapse
Affiliation(s)
- Longsheng Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Shaojun Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200080, China
| | - Junfeng Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Shiyu Mao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Weipu Mao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Wentao Zhang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Yadong Guo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Yuan Wu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Ruiliang Wang
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Yang Yan
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| | - Xudong Yao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai 200072, China,
| |
Collapse
|