1
|
Haibo Z, Tianyun L, Xiaoman C, Xiaoyan H. Cell Senescence-Related Genes as Biomarkers for Prognosis and Immunotherapeutic Response in Colon Cancer. Biochem Genet 2025; 63:124-143. [PMID: 38411939 DOI: 10.1007/s10528-024-10690-z] [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: 11/01/2023] [Accepted: 01/05/2024] [Indexed: 02/28/2024]
Abstract
Colon adenocarcinoma (COAD) stands out as the most prevalent malignancy diagnosed within the gastrointestinal tract, bearing substantial incidence and mortality rates. The processes of ageing and senescence intricately intertwine with tumorigenesis and immune regulation, concurrently exerting influence on the remodelling of the tumor microenvironment (TME). This phenomenon, in turn, significantly impacts the efficacy of immunotherapeutic interventions. Despite this awareness, the comprehensive understanding of the intricate interplay between cellular senescence and TME in the context of COAD remains elusive. Further inquiry is imperative to comprehensively gauge the relevance of cellular senescence-related genes (CSGs) in the realms of immune infiltration and the prognostication of COAD. Differentially expressed cell senescence-related genes (DE-CSGs) within COAD tumors and normal specimens were discerned through analysis of the TCGA-COAD dataset. Leveraging univariate, LASSO, and multivariate Cox regression analyses, we formulated a prognostic risk signature. Subsequent validation utilised two independent GEO datasets. Furthermore, a nomogram was devised to gauge the prognostic significance of this signature. Additionally, the immune landscape of the Cell Senescence-related Signature (CSS) was characterised using CIBERSORT and TIMER algorithms. The expression levels of CSGs were quantified through RT-PCR in COAD specimens. Drawing upon mRNA expression profiles of 191 DE-CSGs, we successfully established a 9-gene CSS, demonstrating its autonomy as a prognostic determinant for COAD patients. Those assigned high-risk scores exhibited an immunosuppressive phenotype, marked by elevated proportions of resting CD4+memory T cells and macrophages M0, correlating with diminished overall survival. Subsequent analyses uncovered that the amalgamation of CSS with the expression profiles of immune checkpoint key genes effectively predicted patient prognosis. Furthermore, patients with low-risk scores demonstrated a potential association with more favourable therapeutic outcomes in the context of immunotherapy. This study has culminated in the development of a prognostic risk signature grounded in cell senescence-related genes for COAD. We posit that the CSS plays a regulatory role in immune infiltration, emerging as a robust biomarker for prognosis and a predictive indicator for immunotherapeutic responsiveness within the COAD landscape.
Collapse
Affiliation(s)
- Zhang Haibo
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Lan Tianyun
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Chen Xiaoman
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Han Xiaoyan
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
2
|
Lin Q, Li J, Sun Y, Abudousalamu Z, Xue M, Yao L, Chen M. Proteome-Wide Mendelian Randomization Analysis to Identify Potential Plasma Biomarkers and Therapeutic Targets for Epithelial Ovarian Cancer Subtypes. Int J Womens Health 2024; 16:2263-2279. [PMID: 39726690 PMCID: PMC11669594 DOI: 10.2147/ijwh.s491414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Background Epithelial ovarian cancer (EOC) remains an unmet medical challenge due to its insidious onset, atypical symptoms, and increasing resistance to conventional chemotherapeutic agents. It is imperative to explore novel biomarkers and generate innovative target drugs. Methods To identify potential proteins with causal association to EOC subtypes, we conducted a Mendelian Randomization (MR) analysis using 15,419 protein quantitative trait loci (pQTLs) associated with 2015 proteins. Bayesian colocalization analysis, Summary-data-based MR, and Heterogeneity in Dependent Instruments tests were employed for validation. Enrichment and druggability analyses were performed to assess the biological significance and therapeutic potential of identified proteins. Results Our analysis identified 455 unique proteins associated with at least one EOC subtype, with 14 protein-cancer associations confirmed by further validation. Ten proteins were prioritized as potential therapeutic targets, including α1B-glycoprotein (A1BG) and ephrin-A1 (EFNA1), which interact with the known drug targets human epidermal growth factor receptor 2 (HER2) and vascular endothelial growth factor receptor (VEGFR). Conclusion This study elucidated the plasma proteins causally associated with EOC subtypes, potentially offering easily detectable biomarkers and promising therapeutic targets. A1BG and EFNA1 were identified as druggable targets and confirmed to correspond with current pharmacological targets. Targeting these proteins in drug development potentially offers an avenue for innovative treatment strategies.
Collapse
Affiliation(s)
- Qianhan Lin
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - Jiajia Li
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - Yating Sun
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - Zulimire Abudousalamu
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - Mengyang Xue
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| | - Liangqing Yao
- Department of Gynecologic Oncology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510005, People’s Republic of China
| | - Mo Chen
- Department of Gynecologic Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, People’s Republic of China
| |
Collapse
|
3
|
Poongkodi K, Periyasamy S, Gurunathan RA, Krishnasamy V, Jayakumar D, Subburaman R, Jayaraman S, Prabhudas SK. The genomic landscape of papillary thyroid carcinoma on next-generation sequencing in patients undergoing total thyroidectomy. World J Surg 2024; 48:2880-2891. [PMID: 39501038 DOI: 10.1002/wjs.12389] [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: 04/26/2024] [Accepted: 10/16/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Thyroidectomy is increasingly performed for suspected malignancy. This cohort study aimed to identify genetic markers associated with malignancy and determine the molecular landscape of papillary thyroid carcinoma (PTC) through next-generation sequencing (NGS) in patients undergoing total thyroidectomy. PATIENTS AND METHODS Among 116 surgical candidates, 103 patients (age = 42.9 ± 13.7 years; Male: Female = 14:89) with benign or malignant thyroid nodules were eligible. Live thyroid tissue samples harvested intraoperatively with adequate DNA and RNA yield were subjected to NGS on the Illumina NovaSeq 6000 platform for genomic and transcriptomic analysis, respectively. RESULTS Histopathology comprised 20 malignant (19.4%) and 83 benign (80.6%) cases, including 16 PTC (15.5%) cases. On NGS, single nucleotide polymorphisms (SNPs) in NTRK1 at NC_000001.11:156879016 on chromosome 1 and ALK at NC_000002.12:29717663 on chromosome 2 were frequent in malignant lesions (p < 0.05). A SNP in ALK at NC_000002.12:29193706 was consistently a homozygous alternate allele across the cohort. DNA-sequencing of PTC lesions identified recurrent somatic mutations in BRAF (100%), ALK (56.3%), RET (18.8%), PIK3CA (12.5%), NTRK1 (12.5%), NTRK2 (87.5%), NTRK3 (12.5%), NRAS (6.3%), and PTCH1 (31.3%) genes. RNA sequencing revealed novel fusion genes, including MKRN1-BRAF, RN7SL1-CDH1, IRF2BPL-MED12, MED12-IRF2BPL, CPM-MDM2, and AC005895.3-PDGFRB. In receiver operative characteristics analysis, the AUCs of ALK mutation predicting recurrence and metastases were 0.818 and 0.783. CONCLUSION This Indian study identified novel somatic mutations and fusion genes in PTC, revealing a distinct genomic landscape with implications in precision diagnostics and personalized therapies. NGS with intraoperative live sampling shows promise in prognostication and therapeutic optimization of advanced/metastatic PTC cases. TRIAL REGISTRATION NO CTRI/2020/09/027607 dt 04/09/2020; REF/2020/08/036119.
Collapse
Affiliation(s)
- Karunakaran Poongkodi
- Chief Civil Surgeon and Assistant Professor of Endocrine Surgery, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
- Principal Investigator, ICMR Extramural Scheme, Multidisciplinary Research Unit, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
- University Online Journal, The Tamilnadu Dr MGR Medical University, Chennai, Tamil Nadu, India
| | - Sumathi Periyasamy
- Department of General Surgery, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
| | - Raj Ashok Gurunathan
- Department of General Surgery, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
| | - Vijayakumar Krishnasamy
- Department of General Surgery, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
| | - Deepak Jayakumar
- Former Research Scientist, Multidisciplinary Research Unit, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
| | - Ramesh Subburaman
- Department of Pathology, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
| | - Sujatha Jayaraman
- Department of Pathology, Government Mohan Kumaramangalam Medical College, Salem, Tamil Nadu, India
| | - Sudheesh K Prabhudas
- Research Associate, Centre for Bioinformatics, ICAR-Central Institute of Brackish Water Aquaculture, Chennai, Tamil Nadu, India
| |
Collapse
|
4
|
Kuriki Y, Sogawa M, Komatsu T, Kawatani M, Fujioka H, Fujita K, Ueno T, Hanaoka K, Kojima R, Hino R, Ueo H, Ueo H, Kamiya M, Urano Y. Modular Design Platform for Activatable Fluorescence Probes Targeting Carboxypeptidases Based on ProTide Chemistry. J Am Chem Soc 2024; 146:521-531. [PMID: 38110248 DOI: 10.1021/jacs.3c10086] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Carboxypeptidases (CPs) are a family of hydrolases that cleave one or more amino acids from the C-terminal of peptides or proteins and play indispensable roles in various physiological and pathological processes. However, only a few highly activatable fluorescence probes for CPs have been reported, and there is a need for a flexibly tunable molecular design platform to afford a range of fluorescence probes for CPs for biological and medical research. Here, we focused on the unique activation mechanism of ProTide-based prodrugs and established a modular design platform for CP-targeting florescence probes based on ProTide chemistry. In this design, probe properties such as fluorescence emission wavelength, reactivity/stability, and target CP can be readily tuned and optimized by changing the four probe modules: the fluorophore, the substituent on the phosphorus atom, the linker amino acid at the P1 position, and the substrate amino acid at the P1' position. In particular, switching the linker amino acid at position P1 enabled us to precisely optimize the reactivity for target CPs. As a proof-of-concept, we constructed probes for carboxypeptidase M (CPM) and prostate-specific membrane antigen (also known as glutamate carboxypeptidase II). The developed probes were applicable for the imaging of CP activities in live cells and in clinical specimens from patients. This design strategy should be useful in studying CP-related biological and pathological phenomena.
Collapse
Affiliation(s)
- Yugo Kuriki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Mari Sogawa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Toru Komatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Minoru Kawatani
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259, Nagatsuda-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Hiroyoshi Fujioka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259, Nagatsuda-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Kyohhei Fujita
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tasuku Ueno
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kenjiro Hanaoka
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ryosuke Kojima
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Rumi Hino
- Department of Sports and Health Science, Daito Bunka University, 560 Iwadono, Higashimatsuyama, Saitama 355-8501, Japan
| | - Hiroki Ueo
- Ueo Breast Cancer Hospital, 1-3-5 Futamatacho, Oita, Oita 870-0887, Japan
| | - Hiroaki Ueo
- Ueo Breast Cancer Hospital, 1-3-5 Futamatacho, Oita, Oita 870-0887, Japan
| | - Mako Kamiya
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259, Nagatsuda-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
- Living Systems Materialogy (LiSM) Research Group, International Research Frontiers Initiative (IRFI), Tokyo Institute of Technology, 4259, Nagatsuda-cho, Midori-ku, Yokohama, Kanagawa 226-8501, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
5
|
Stuardo-Parada A, López-Muñoz R, Villarroel-Espindola F, Figueroa CD, Ehrenfeld P. Minireview: functional roles of tissue kallikrein, kinins, and kallikrein-related peptidases in lung cancer. Med Oncol 2023; 40:224. [PMID: 37405520 DOI: 10.1007/s12032-023-02090-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
Despite campaigns and improvements in detection and treatment, lung cancer continues to increase worldwide and represents a major public health problem. One approach to treating patients suffering from lung cancer is to target surface receptors overexpressed on tumor cells, such as GPCR-family kinin receptors, and proteases that control tumor progression, such as kallikrein-related peptidases (KLKs). These proteases have been visualized in recent years due to their contribution to the progression of cancers, such as prostate and ovarian cancer, facilitating the invasive and metastatic capacity of tumor cells in these tissues. In fact, KLK3 is the specific prostate antigen, the only tissue-specific biomarker used to diagnose this malignancy. In lung cancer to date, evidence indicates that KLK5, KLK6, KLK8, KLK11, and KLK14 are the major peptidases regulated and involved in its progression. The expression levels of KLKs in this neoplasm are modulated by the secretome of the different cell types present in the tumor microenvironment, the cancer subtype and the tumor stage, among others. Considering the multiple functions of kinin receptors and KLKs, this review highlights their roles, even considering the SARS-CoV-2 effects. Since lung cancer is often diagnosed in advanced stages, our efforts should focus on early diagnosis, validating for example specific KLKs, especially in high-risk populations such as smokers and people exposed to carcinogenic fumes, oil fields, and contaminated workplaces, unexplored fields to investigate. Furthermore, their modulation could be considered as a promising approach in lung cancer therapeutics.
Collapse
Affiliation(s)
- Adriana Stuardo-Parada
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo López-Muñoz
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | | | - Carlos D Figueroa
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.
- Center for Interdisciplinary Studies on Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
6
|
Cheng X, Wei Y, Fu Y, Li J, Han L. A novel enterocyte-related 4-gene signature for predicting prognosis in colon adenocarcinoma. Front Immunol 2022; 13:1052182. [PMID: 36532007 PMCID: PMC9755665 DOI: 10.3389/fimmu.2022.1052182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background Colon adenocarcinoma (COAD) is a fatal disease, and its cases are constantly increasing worldwide. Further, the therapeutic and management strategies for patients with COAD are still unsatisfactory due to the lack of accurate patient classification and prognostic models. Therefore, our study aims to identify prognostic markers in patients with COAD and construct a cell subtype-specific prognostic model with high accuracy and robustness. Methods Single-cell transcriptomic data of six samples were retrieved from the Gene expression omnibus (GEO) database. The cluster annotation and cell-cell communication analysis identified enterocytes as a key player mediating signal communication networks. A four-gene signature prognostic model was constructed based on the enterocyte-related differentially expressed genes (ERDEGs) in patients with COAD of the Cancer Genome Atlas cohort. The prognostic model was validated on three external validation cohorts from the GEO database. The correlation between immune cell infiltration, immunotherapy response, drug sensitivity, and the four-gene signature prognostic model was investigated. Finally, immunohistochemistry (IHC) was performed to determine the expression of the four genes. Results We found that the proportion of epithelial cells was obviously large in COAD samples. Further analysis of epithelial cells showed that the activity of the enterocytes was highest in the cell-cell communication network. Based on enterocyte data, 30 ERDEGs were identified and a 4-gene prognostic model including CPM, CLCA4, ELOVL6, and ATP2A3 was developed and validated. The risk score derived from this model was considered as an independent variable factor to predict overall survival. The patients were divided into high- and low-risk groups based on the median riskscore value. The correlation between immune cell infiltration, immunotherapy response, immune status, clinical characteristics, drug sensitivity, and risk score was analyzed. IHC confirmed the expression of signature genes in tissues from normal individuals, patients with polyps, and COAD. Conclusion In this study, we constructed and validated a novel four-gene signature prognostic model, which could effectively predict the response to immunotherapy and overall survival in patients with COAD. More importantly, this model provides useful insight into the management of colon cancer patients and aids in designing personalized therapy.
Collapse
Affiliation(s)
- Xuehua Cheng
- Department of Traditional Chinese Medicine (TCM) Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yong Wei
- Translational Medicine Department, GeneScience Pharmaceuticals Co. Ltd., Changchun, China
| | - Yugang Fu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Li Han, ; Jiacheng Li,
| | - Li Han
- Department of Traditional Chinese Medicine (TCM) Geriatrics, Huadong Hospital Affiliated to Fudan University, Shanghai, China,*Correspondence: Li Han, ; Jiacheng Li,
| |
Collapse
|
7
|
Liu S, Wang H, Jiang X, Ji Y, Wang Z, Zhang Y, Wang P, Xiao H. Integrated N-glycoproteomics Analysis of Human Saliva for Lung Cancer. J Proteome Res 2022; 21:1589-1602. [PMID: 35715216 DOI: 10.1021/acs.jproteome.1c00701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Aberrant protein N-glycosylation is a cancer hallmark, which has great potential for cancer detection. However, large-scale and in-depth analysis of N-glycosylation remains challenging because of its high heterogeneity, complexity, and low abundance. Human saliva is an attractive diagnostic body fluid, while few efforts explored its N-glycoproteome for lung cancer. Here, we utilized a zwitterionic-hydrophilic interaction chromatography-based strategy to specifically enrich salivary glycopeptides. Through quantitative proteomics analysis, 1492 and 1234 intact N-glycopeptides were confidently identified from pooled saliva samples of 10 subjects in the nonsmall-cell lung cancer group and 10 subjects in the normal control group. Accordingly, 575 and 404 N-glycosites were revealed for the lung cancer group and normal control group. In particular, 154 N-glycosites and 259 site-specific glycoforms were significantly dysregulated in the lung cancer group. Several N-glycosites located at the same glycoprotein and glycans attached to the same N-glycosites were observed with differential expressions, including haptoglobin, Mucin-5B, lactotransferrin, and α-1-acid glycoprotein 1. These N-glycoproteins were mainly related to inflammatory responses, infectious diseases, and cancers. Our study achieved comprehensive characterization of salivary N-glycoproteome, and dysregulated site-specific glycoforms hold promise for noninvasive detection of lung cancer.
Collapse
Affiliation(s)
- Sha Liu
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huiyu Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoteng Jiang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yin Ji
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China
| | - Zeyuan Wang
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Peng Wang
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China
| | - Hua Xiao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
8
|
Mangiola S, McCoy P, Modrak M, Souza-Fonseca-Guimaraes F, Blashki D, Stuchbery R, Keam SP, Kerger M, Chow K, Nasa C, Le Page M, Lister N, Monard S, Peters J, Dundee P, Williams SG, Costello AJ, Neeson PJ, Pal B, Huntington ND, Corcoran NM, Papenfuss AT, Hovens CM. Transcriptome sequencing and multi-plex imaging of prostate cancer microenvironment reveals a dominant role for monocytic cells in progression. BMC Cancer 2021; 21:846. [PMID: 34294073 PMCID: PMC8296706 DOI: 10.1186/s12885-021-08529-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/23/2021] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Prostate cancer is caused by genomic aberrations in normal epithelial cells, however clinical translation of findings from analyses of cancer cells alone has been very limited. A deeper understanding of the tumour microenvironment is needed to identify the key drivers of disease progression and reveal novel therapeutic opportunities. RESULTS In this study, the experimental enrichment of selected cell-types, the development of a Bayesian inference model for continuous differential transcript abundance, and multiplex immunohistochemistry permitted us to define the transcriptional landscape of the prostate cancer microenvironment along the disease progression axis. An important role of monocytes and macrophages in prostate cancer progression and disease recurrence was uncovered, supported by both transcriptional landscape findings and by differential tissue composition analyses. These findings were corroborated and validated by spatial analyses at the single-cell level using multiplex immunohistochemistry. CONCLUSIONS This study advances our knowledge concerning the role of monocyte-derived recruitment in primary prostate cancer, and supports their key role in disease progression, patient survival and prostate microenvironment immune modulation.
Collapse
Affiliation(s)
- Stefano Mangiola
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick McCoy
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Martin Modrak
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Fernando Souza-Fonseca-Guimaraes
- University of Queensland Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Daniel Blashki
- The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Ryan Stuchbery
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Simon P Keam
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Michael Kerger
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ken Chow
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Chayanica Nasa
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Melanie Le Page
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Natalie Lister
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Simon Monard
- Flow Cytometry Facility, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Justin Peters
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Phil Dundee
- Epworth Center of Cancer Research, Clayton, Victoria, Australia
| | - Scott G Williams
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Anthony J Costello
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Paul J Neeson
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Bhupinder Pal
- The Olivia Newton-John Cancer Research Institute, Heidelberg, Melbourne, Australia
| | - Nicholas D Huntington
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Niall M Corcoran
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Urology, Frankston Hospital, Frankston, Victoria, Australia
| | - Anthony T Papenfuss
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- School of Mathematics and Statistics, University of Melbourne, Melbourne, VIC, 3010, Australia.
| | - Christopher M Hovens
- Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
9
|
Xanthoulea S, Konings GFJ, Saarinen N, Delvoux B, Kooreman LFS, Koskimies P, Häkkinen MR, Auriola S, D'Avanzo E, Walid Y, Verhaegen F, Lieuwes NG, Caiment F, Kruitwagen R, Romano A. Pharmacological inhibition of 17β-hydroxysteroid dehydrogenase impairs human endometrial cancer growth in an orthotopic xenograft mouse model. Cancer Lett 2021; 508:18-29. [PMID: 33762202 DOI: 10.1016/j.canlet.2021.03.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 01/12/2023]
Abstract
Endometrial cancer (EC) is the most common gynaecological tumor in developed countries and its incidence is increasing. Approximately 80% of newly diagnosed EC cases are estrogen-dependent. Type 1 17β-hydroxysteroid dehydrogenase (17β-HSD-1) is the enzyme that catalyzes the final step in estrogen biosynthesis by reducing the weak estrogen estrone (E1) to the potent estrogen 17β-estradiol (E2), and previous studies showed that this enzyme is implicated in the intratumoral E2 generation in EC. In the present study we employed a recently developed orthotopic and estrogen-dependent xenograft mouse model of EC to show that pharmacological inhibition of the 17β-HSD-1 enzyme inhibits disease development. Tumors were induced in one uterine horn of athymic nude mice by intrauterine injection of the well-differentiated human endometrial adenocarcinoma Ishikawa cell line, modified to express human 17β-HSD-1 in levels comparable to EC, and the luciferase and green fluorescent protein reporter genes. Controlled estrogen exposure in ovariectomized mice was achieved using subcutaneous MedRod implants that released either the low active estrone (E1) precursor or vehicle. A subgroup of E1 supplemented mice received daily oral gavage of FP4643, a well-characterized 17β-HSD-1 inhibitor. Bioluminescence imaging (BLI) was used to measure tumor growth non-invasively. At sacrifice, mice receiving E1 and treated with the FP4643 inhibitor showed a significant reduction in tumor growth by approximately 65% compared to mice receiving E1. Tumors exhibited metastatic spread to the peritoneum, to the lymphovascular space (LVI), and to the thoracic cavity. Metastatic spread and LVI invasion were both significantly reduced in the inhibitor-treated group. Transcriptional profiling of tumors indicated that FP4643 treatment reduced the oncogenic potential at the mRNA level. In conclusion, we show that 17β-HSD-1 inhibition represents a promising novel endocrine treatment for EC.
Collapse
Affiliation(s)
- Sofia Xanthoulea
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands.
| | - Gonda F J Konings
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands
| | - Niina Saarinen
- Forendo Pharma Ltd., Turku, Finland; Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling (TCDM), University of Turku, Finland
| | - Bert Delvoux
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands
| | - Loes F S Kooreman
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Pathology, Maastricht University Medical Centre, the Netherlands
| | | | - Merja R Häkkinen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Elisabetta D'Avanzo
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands
| | - Youssef Walid
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands
| | - Frank Verhaegen
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands
| | - Natasja G Lieuwes
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; MAASTRO Lab, Maastricht University Medical Centre, the Netherlands
| | - Florian Caiment
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Toxicogenomics, Maastricht University Medical Centre, the Netherlands
| | - Roy Kruitwagen
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands
| | - Andrea Romano
- GROW - School for Oncology & Developmental Biology, Maastricht University, the Netherlands; Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, the Netherlands
| |
Collapse
|
10
|
Fang Z, Xu S, Xie Y, Yan W. Identification of a prognostic gene signature of colon cancer using integrated bioinformatics analysis. World J Surg Oncol 2021; 19:13. [PMID: 33441161 PMCID: PMC7807455 DOI: 10.1186/s12957-020-02116-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/25/2020] [Indexed: 12/17/2022] Open
Abstract
Background Colon cancer is a worldwide leading cause of cancer-related mortality, and the prognosis of colon cancer is still needed to be improved. This study aimed to construct a prognostic model for predicting the prognosis of colon cancer. Methods The gene expression profile data of colon cancer were obtained from the TCGA, GSE44861, and GSE44076 datasets. The WGCNA module genes and common differentially expressed genes (DEGs) were used to screen out the prognosis-associated DEGs, which were used to construct a prognostic model. The performance of the prognostic model was assessed and validated in the TCGA training and microarray validation sets (GSE38832 and GSE17538). At last, the model and prognosis-associated clinical factors were used for the construction of the nomogram. Results Five colon cancer-related WGCNA modules (including 1160 genes) and 1153 DEGs between tumor and normal tissues were identified, inclusive of 556 overlapping DEGs. Stepwise Cox regression analyses identified there were 14 prognosis-associated DEGs, of which 12 DEGs were included in the optimized prognostic gene signature. This prognostic model presented a high forecast ability for the prognosis of colon cancer both in the TCGA training dataset and the validation datasets (GSE38832 and GSE17538; AUC > 0.8). In addition, patients’ age, T classification, recurrence status, and prognostic risk score were associated with the prognosis of TCGA patients with colon cancer. The nomogram was constructed using the above factors, and the predictive 3- and 5-year survival probabilities had high compliance with the actual survival proportions. Conclusions The 12-gene signature prognostic model had a high predictive ability for the prognosis of colon cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-020-02116-y.
Collapse
Affiliation(s)
- Zhengyu Fang
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang Province, China
| | - Sumei Xu
- Department of General Practice, The First Affiliated Hospital of Zhejiang Chinese Medical University, #54 Youdian Road, Shangcheng District, Hangzhou, 310006, Zhejiang Province, China.
| | - Yiwen Xie
- Department of General Practice, The First Affiliated Hospital of Zhejiang Chinese Medical University, #54 Youdian Road, Shangcheng District, Hangzhou, 310006, Zhejiang Province, China
| | - Wenxi Yan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, Zhejiang Province, China
| |
Collapse
|
11
|
|
12
|
G. Keller S, Kamiya M, Urano Y. Recent Progress in Small Spirocyclic, Xanthene-Based Fluorescent Probes. Molecules 2020; 25:E5964. [PMID: 33339370 PMCID: PMC7766215 DOI: 10.3390/molecules25245964] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
The use of fluorescent probes in a multitude of applications is still an expanding field. This review covers the recent progress made in small molecular, spirocyclic xanthene-based probes containing different heteroatoms (e.g., oxygen, silicon, carbon) in position 10'. After a short introduction, we will focus on applications like the interaction of probes with enzymes and targeted labeling of organelles and proteins, detection of small molecules, as well as their use in therapeutics or diagnostics and super-resolution microscopy. Furthermore, the last part will summarize recent advances in the synthesis and understanding of their structure-behavior relationship including novel computational approaches.
Collapse
Affiliation(s)
- Sascha G. Keller
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; (S.G.K.); (M.K.)
| | - Mako Kamiya
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; (S.G.K.); (M.K.)
| | - Yasuteru Urano
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan; (S.G.K.); (M.K.)
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
13
|
Ye Y, An Y, Wang M, Liu H, Guan L, Wang Z, Li W. Expression of Carboxypeptidase X M14 Family Member 2 Accelerates the Progression of Hepatocellular Carcinoma via Regulation of the gp130/JAK2/Stat1 Pathway. Cancer Manag Res 2020; 12:2353-2364. [PMID: 32280274 PMCID: PMC7127851 DOI: 10.2147/cmar.s228984] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background Carboxypeptidase X, M14 family member 2 (CPXM2) has been reported to be involved with several human malignancies. However, the impact of CPXM2 on human hepatocellular carcinoma (HCC) tumorigenesis has not been studied. Materials and Methods Using immunohistochemistry, the detailed CPXM2 expression patterns were examined in HCC cell lines and tissues. Additionally, a hepatic stellate cell line overexpressing CPXM2 and an HCC CPXM2-knockdown cell line were established by lipofection of an expression plasmid or short hairpin RNA, respectively. The transfection efficiencies were confirmed by reverse transcription-quantitative PCR, Western blotting and immunofluorescence. Moreover, Western blotting was conducted to determine the phosphorylation levels of the tyrosine kinase 2 (JAK2)/signal transducer and activator of transcription 3 (Stat1) pathway. Furthermore, gp130-specific hairpin RNA was used to knockdown gp130 expression in hepatic stellate cells overexpressing CPXM2. The malignant phenotype of cultured HCC cells was assessed by a Cell Counting Kit-8 (CCK8) assay, plate cloning assay, Matrigel invasion assay and wound-healing assay in vitro. Results It was demonstrated that CPXM2 was upregulated in HCC, and its upregulation predicted a poor prognosis. Besides, the upregulation of CPXM2 markedly enhanced the metastatic potential of HCC via the gp130/JAK2/Stat1 signaling pathway in vitro. Conclusion In summary, this evidence suggests a positive role for CPXM2 in HCC progression via modulation of the gp130/JAK2/Stat1 signaling pathway in HCC.
Collapse
Affiliation(s)
- Yanshuo Ye
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Yuan An
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Min Wang
- Department of Pathology, Jilin Provincial Cancer Hospital, Changchun 130012, People's Republic of China
| | - Hongyu Liu
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Lianyue Guan
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhanpeng Wang
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Wei Li
- Department of Hepatobiliary-Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
14
|
Zhao X, Li R, Wang Q, Wu M, Wang Y. Overexpression of carboxypeptidase X M14 family member 2 predicts an unfavorable prognosis and promotes proliferation and migration of osteosarcoma. Diagn Pathol 2019; 14:118. [PMID: 31651348 PMCID: PMC6813969 DOI: 10.1186/s13000-019-0887-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/09/2019] [Indexed: 11/23/2022] Open
Abstract
Background Carboxypeptidase X, M14 family member 2 (CPXM2), has been associated with several human developmental disorders. However, whether CPXM2 is involved in oncogenesis or tumor progression remains unclear. Currently, the clinical relevance and function of CPXM2 in human osteosarcoma were investigated. Materials and methods The expression of CPXM2 in osteosarcoma cell lines and tissues were explored by immunohistochemistry and western blotting assays. A eukaryotic expression plasmid was transfected into fetal osteoblast cells to overexpress CPXM2 and the endogenous CPXM2 in osteosarcoma cells was silenced through an RNA interference (RNAi) method transfection. These transfections were validated via western blotting, and the expression levels of several key molecules involved in the epithelial mesenchymal transition was also determined via western blotting. The expression levels of CPXM2 in a fetal osteoblast cell line with CPXM2 overexpressing and an osteosarcoma CPXM2-knockout cell line was confirmed via reverse transcription-quantitative polymerase chain reaction (RT-qPCR), western blotting and immunofluorescence. The malignant phenotype of osteosarcoma cells was indicated by the cholecystokinin octapeptide, colony formation assay, scratch wound healing assay, and Transwell® migration assay. Results We found that CPXM2 was overexpressed in osteosarcoma and that the overexpression was associated with an unfavorable prognosis and tumor node metastasis staging. The knockdown of CPXM2 in cultured osteosarcoma cells significantly impeded cell proliferation and migration. In addition, the upregulation of CPXM2 in fetal osteoblast cells significantly promoted cell proliferation and migration. Besides, western blotting results revealed that several key molecules involved in the epithelial mesenchymal transition (EMT) were regulated by CPXM2. Conclusion Taken together, these results imply an active role for CPXM2 in promoting tumor aggressiveness via epithelial to mesenchymal transition (EMT) modulation in osteosarcoma.
Collapse
Affiliation(s)
- Xin Zhao
- Orthopedic Department, The Second Hospital of Jilin University, No. 128 Ziqiang Road, Changchun, 130041, China
| | - Ronghang Li
- Department of Joint Surgery and Sports Medicine, The Second Hospital of Jilin University, No. 128 Ziqiang Road, Changchun, 130041, China
| | - Qian Wang
- Otolaryngology Head and Neck Surgery, First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Minfei Wu
- Orthopedic Department, The Second Hospital of Jilin University, No. 128 Ziqiang Road, Changchun, 130041, China.
| | - Yanbing Wang
- Orthopedic Department, The Second Hospital of Jilin University, No. 128 Ziqiang Road, Changchun, 130041, China.
| |
Collapse
|
15
|
Niu G, Yang Y, Ren J, Song T, Hu Z, Chen L, Hong R, Xia J, Ke C, Wang X. Overexpression of CPXM2 predicts an unfavorable prognosis and promotes the proliferation and migration of gastric cancer. Oncol Rep 2019; 42:1283-1294. [PMID: 31364750 PMCID: PMC6718098 DOI: 10.3892/or.2019.7254] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/24/2019] [Indexed: 12/24/2022] Open
Abstract
Carboxypeptidase X, M14 family member 2 (CPXM2), has been associated with several human disorders such as developmental diseases. However, whether CPXM2 is involved in oncogenesis or tumor progression remains unclear. In the present study, we used clinical samples from gastric cancer (GC) patients to investigate potential roles of CPXM2 in GC. We also analyzed datasets from the Oncomine database, The Cancer Genome Atlas (TCGA), and the Kaplan‑Meier Plotter to validate these results. We found that CPXM2 was overexpressed in GC and that the overexpression was associated with an unfavorable prognosis, regardless of the Lauren classification and tumor node metastasis staging. In addition, knockdown of CPXM2 in cultured GC cells significantly impeded cell proliferation and migration, as indicated by the cholecystokinin octapeptide, colony formation assay, scratch wound healing assay, and Transwell® migration assay. Furthermore, gene set enrichment analysis using RNA‑seq data from TCGA indicated that high CPXM2 expression in GC patients was positively correlated with the HALLMARK_APICAL_JUNCTION and HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION gene sets. Finally, western blotting results revealed that several key molecules involved in the epithelial mesenchymal transition were regulated by CPXM2. Taken together, these results imply an active role for CPXM2 in promoting tumor aggressiveness via epithelial to mesenchymal transition (EMT) modulation in GCs.
Collapse
Affiliation(s)
- Gengming Niu
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Yazhe Yang
- Queen Mary College of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Ren
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Tao Song
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Zhiqing Hu
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Liang Chen
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Runqi Hong
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Jie Xia
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Chongwei Ke
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Xin Wang
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
16
|
Interactions between carboxypeptidase M and kinin B1 receptor in endothelial cells. Inflamm Res 2019; 68:845-855. [DOI: 10.1007/s00011-019-01264-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/03/2019] [Accepted: 06/13/2019] [Indexed: 11/25/2022] Open
|
17
|
Ahn J, Wu H, Lee K. Integrative Analysis Revealing Human Adipose-Specific Genes and Consolidating Obesity Loci. Sci Rep 2019; 9:3087. [PMID: 30816281 PMCID: PMC6395763 DOI: 10.1038/s41598-019-39582-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Identification of adipose-specific genes has contributed to an understanding of mechanisms underlying adipocyte development and obesity. Herein, our analyses of the recent Genotype-Tissue Expression (GTEx) database revealed 38 adipose-specific/enhanced protein coding genes, among which 3 genes were novel adipose-specific, and 414 highly differentially expressed genes (DEGs) between subcutaneous and omental adipose depots. By integrative analyses of genome-wide association studies (GWASs), 14 adipose-specific/enhanced genes and 60 DEGs were found to be associated with obesity-related traits and diseases, consolidating evidence for contribution of these genes to the regional fat distribution and obesity phenotypes. In addition, expression of HOXC cluster was up-regulated in subcutaneous adipose tissue, and the majority of the HOXB cluster was expressed highly in omental adipose tissue, indicating differential expression patterns of HOX clusters in adipose depots. Our findings on the distinct gene expression profiles in adipose tissue and their relation to obesity provide an important foundation for future functional biological studies and therapeutic targets in obesity and associated diseases.
Collapse
Affiliation(s)
- Jinsoo Ahn
- Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Huiguang Wu
- Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210, USA.,College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
18
|
Kuriki Y, Kamiya M, Kubo H, Komatsu T, Ueno T, Tachibana R, Hayashi K, Hanaoka K, Yamashita S, Ishizawa T, Kokudo N, Urano Y. Establishment of Molecular Design Strategy To Obtain Activatable Fluorescent Probes for Carboxypeptidases. J Am Chem Soc 2018; 140:1767-1773. [DOI: 10.1021/jacs.7b11014] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Mako Kamiya
- PRESTO (Japan)
Science
and Technology Agency (JST), 4-1-8
Honcho Kawaguchi-shi, Saitama 332-0012, Japan
| | - Hidemasa Kubo
- Division
of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | | | | | | | | | | | | | - Takeaki Ishizawa
- Department
of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | - Norihiro Kokudo
- Department
of Surgery, National Center for Global Health and Medicine, 1-21-1
Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Yasuteru Urano
- CREST (Japan)
Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
19
|
Testero SA, Granados C, Fernández D, Gallego P, Covaleda G, Reverter D, Vendrell J, Avilés FX, Pallarès I, Mobashery S. Discovery of Mechanism-Based Inactivators for Human Pancreatic Carboxypeptidase A from a Focused Synthetic Library. ACS Med Chem Lett 2017; 8:1122-1127. [PMID: 29057062 DOI: 10.1021/acsmedchemlett.7b00346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/22/2017] [Indexed: 12/22/2022] Open
Abstract
Metallocarboxypeptidases (MCPs) are involved in many biological processes such as fibrinolysis or inflammation, development, Alzheimer's disease, and various types of cancer. We describe the synthesis and kinetic characterization of a focused library of 22 thiirane- and oxirane-based potential mechanism-based inhibitors, which led to discovery of an inhibitor for the human pro-carboxypeptidase A1. Our structural analyses show that the thiirane-based small-molecule inhibitor penetrates the barrier of the pro-domain to bind within the active site. This binding leads to a chemical reaction that covalently modifies the catalytic Glu270. These results highlight the importance of combined structural, biophysical, and biochemical evaluation of inhibitors in design strategies for the development of spectroscopically nonsilent probes as effective beacons for in vitro, in cellulo, and/or in vivo localization in clinical and industrial applications.
Collapse
Affiliation(s)
- Sebastián A. Testero
- Department
of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Carla Granados
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Daniel Fernández
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Pablo Gallego
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Giovanni Covaleda
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - David Reverter
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Josep Vendrell
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Francesc X. Avilés
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Irantzu Pallarès
- Departament
de Bioquímica i Biologia Molecular, Facultat de Biociències,
and Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain
| | - Shahriar Mobashery
- Department
of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
20
|
Pan Y, Zhang H, Zhang M, Zhu J, Yu J, Wang B, Qiu J, Zhang J. A five-gene based risk score with high prognostic value in colorectal cancer. Oncol Lett 2017; 14:6724-6734. [PMID: 29344121 PMCID: PMC5754913 DOI: 10.3892/ol.2017.7097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring malignancies worldwide. The outcomes of patients with similar clinical symptoms or at similar pathological stages remain unpredictable. This inherent clinical diversity is most likely due to the genetic heterogeneity. The present study aimed to create a predicting tool to evaluate patient survival based on genetic profile. Firstly, three Gene Expression Omnibus (GEO) datasets (GSE9348, GSE44076 and GSE44861) were utilized to identify and validate differentially expressed genes (DEGs) in CRC. The GSE14333 dataset containing survival information was then introduced in order to screen and verify prognosis-associated genes. Of the 66 DEGs, the present study screened out 46 biomarkers closely associated to patient overall survival. By Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis, it was demonstrated that these genes participated in multiple biological processes which were highly associated with cancer proliferation, drug-resistance and metastasis, thus further affecting patient survival. The five most important genes, MET proto-oncogene, receptor tyrosine kinase, carboxypeptidase M, serine hydroxymethyltransferase 2, guanylate cyclase activator 2B and sodium voltage-gated channel a subunit 9 were selected by a random survival forests algorithm, and were further made up to a linear risk score formula by multivariable cox regression. Finally, the present study tested and verified this risk score within three independent GEO datasets (GSE14333, GSE17536 and GSE29621), and observed that patients with a high risk score had a lower overall survival (P<0.05). Furthermore, this risk score was the most significant compared with other predicting factors including age and American Joint Committee on Cancer stage, in the model, and was able to predict patient survival independently and directly. The findings suggest that this survival associated DEGs-based risk score is a powerful and accurate prognostic tool and is promisingly implemented in a clinical setting.
Collapse
Affiliation(s)
- Yida Pan
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Hongyang Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Mingming Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing 210008, P.R. China
| | - Jie Zhu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jianghong Yu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Bangting Wang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jigang Qiu
- Department of General Surgery, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
21
|
Wang Y, Liu H, Liang D, Huang Y, Zeng Y, Xing X, Xia J, Lin M, Han X, Liao N, Liu X, Liu J. Reveal the molecular signatures of hepatocellular carcinoma with different sizes by iTRAQ based quantitative proteomics. J Proteomics 2016; 150:230-241. [PMID: 27693406 DOI: 10.1016/j.jprot.2016.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 08/03/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
Tumor size of hepatocellular carcinoma (HCC) is a key parameter for predicting prognosis of HCC patients. The biological behaviors of HCC, such as tumor growth, recurrence and metastasis are significantly associated with tumor size. However, the underlying molecular mechanisms remain unclear. Here, we applied iTRAQ-based proteomic strategy to analyze the proteome differences among small, media, large and huge primary HCC tissues. In brief,88 proteins in small HCC, 69 proteins in media HCC, 118 proteins in large HCC and 215 proteins in huge HCC, were identified by comparing the proteome of cancerous tissues with its corresponding non-cancerous tissues. Further analysis of dysregulated proteins involved in signaling revealed that alteration of ERK1/2 and AKT signaling played important roles in the tumorigenesis or tumor growth in all subtypes. Interestingly, alteration of specific signaling was discovered in small and huge HCC, which might reflect specific molecular mechanisms of tumor growth. Furthermore, the dysregulation degree of a group of proteins has been confirmed to be significantly correlated with the tumor size; these proteins might be potential targets for studying tumor growth of HCC. Overall, we have revealed the molecular signatures of HCC with different tumor sizes, and provided fundamental information for further in-depth study. BIOLOGICAL SIGNIFICANCE In this study, we compared the protein expression profiles among different HCC subtypes, including small HCC, media HCC, large HCC and huge HCC for the first time. The results clearly proved that different molecular alterations and specific signaling pathways were indeed involved in different HCC subtypes, which might explain the different malignancy biological behaviors. In addition, the dysregulation degree of a group of proteins has been confirmed to be significantly correlated with the tumor size. We believe that these findings would help us better understand the underlying molecular mechanisms of the tumorigenesis and development of HCC.
Collapse
Affiliation(s)
- Yingchao Wang
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Hongzhi Liu
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China; Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350007, People's Republic of China
| | - Dong Liang
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China; Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350007, People's Republic of China
| | - Yao Huang
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China; Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350007, People's Republic of China
| | - Yongyi Zeng
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China; Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350007, People's Republic of China
| | - Xiaohua Xing
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Jiangbao Xia
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Minjie Lin
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Xiao Han
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, People's Republic of China
| | - Naishun Liao
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China
| | - Xiaolong Liu
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China.
| | - Jingfeng Liu
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, People's Republic of China; The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, People's Republic of China; Liver Disease Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350007, People's Republic of China.
| |
Collapse
|
22
|
Zinc and zinc-containing biomolecules in childhood brain tumors. J Mol Med (Berl) 2016; 94:1199-1215. [PMID: 27638340 DOI: 10.1007/s00109-016-1454-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 07/27/2016] [Indexed: 12/21/2022]
Abstract
Zinc ions are essential cofactors of a wide range of enzymes, transcription factors, and other regulatory proteins. Moreover, zinc is also involved in cellular signaling and enzymes inhibition. Zinc dysregulation, deficiency, over-supply, and imbalance in zinc ion transporters regulation are connected with various diseases including cancer. A zinc ion pool is maintained by two types of proteins: (i) zinc-binding proteins, which act as a buffer and intracellular donors of zinc and (ii) zinc transporters responsible for zinc fluxes into/from cells and organelles. The decreased serum zinc ion levels have been identified in patients suffering from various cancer diseases, including head and neck tumors and breast, prostate, liver, and lung cancer. On the contrary, increased zinc ion levels have been found in breast cancer and other malignant tissues. Zinc metalloproteomes of a majority of tumors including brain ones are still not yet fully understood. Current knowledge show that zinc ion levels and detection of certain zinc-containing proteins may be utilized for diagnostic and prognostic purposes. In addition, these proteins can also be promising therapeutic targets. The aim of the present work is an overview of the importance of zinc ions, zinc transporters, and zinc-containing proteins in brain tumors, which are, after leukemia, the second most common type of childhood cancer and the second leading cause of death in children after accidents.
Collapse
|
23
|
Kallikrein in the Interstitial Space. Protein Sci 2016. [DOI: 10.1201/9781315374307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
24
|
Taccioli C, Garofalo M, Chen H, Jiang Y, Tagliazucchi GM, Di Leva G, Alder H, Fadda P, Middleton J, Smalley KJ, Selmi T, Naidu S, Farber JL, Croce CM, Fong LY. Repression of Esophageal Neoplasia and Inflammatory Signaling by Anti-miR-31 Delivery In Vivo. J Natl Cancer Inst 2015; 107:djv220. [PMID: 26286729 PMCID: PMC4675101 DOI: 10.1093/jnci/djv220] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 01/31/2015] [Accepted: 07/20/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Overexpression of microRNA-31 (miR-31) is implicated in the pathogenesis of esophageal squamous cell carcinoma (ESCC), a deadly disease associated with dietary zinc deficiency. Using a rat model that recapitulates features of human ESCC, the mechanism whereby Zn regulates miR-31 expression to promote ESCC is examined. METHODS To inhibit in vivo esophageal miR-31 overexpression in Zn-deficient rats (n = 12-20 per group), locked nucleic acid-modified anti-miR-31 oligonucleotides were administered over five weeks. miR-31 expression was determined by northern blotting, quantitative polymerase chain reaction, and in situ hybridization. Physiological miR-31 targets were identified by microarray analysis and verified by luciferase reporter assay. Cellular proliferation, apoptosis, and expression of inflammation genes were determined by immunoblotting, caspase assays, and immunohistochemistry. The miR-31 promoter in Zn-deficient esophagus was identified by ChIP-seq using an antibody for histone mark H3K4me3. Data were analyzed with t test and analysis of variance. All statistical tests were two-sided. RESULTS In vivo, anti-miR-31 reduced miR-31 overexpression (P = .002) and suppressed the esophageal preneoplasia in Zn-deficient rats. At the same time, the miR-31 target Stk40 was derepressed, thereby inhibiting the STK40-NF-κΒ-controlled inflammatory pathway, with resultant decreased cellular proliferation and activated apoptosis (caspase 3/7 activities, fold change = 10.7, P = .005). This same connection between miR-31 overexpression and STK40/NF-κΒ expression was also documented in human ESCC cell lines. In Zn-deficient esophagus, the miR-31 promoter region and NF-κΒ binding site were activated. Zn replenishment restored the regulation of this genomic region and a normal esophageal phenotype. CONCLUSIONS The data define the in vivo signaling pathway underlying interaction of Zn deficiency and miR-31 overexpression in esophageal neoplasia and provide a mechanistic rationale for miR-31 as a therapeutic target for ESCC.
Collapse
Affiliation(s)
- Cristian Taccioli
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Michela Garofalo
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Hongping Chen
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Yubao Jiang
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Guidantonio Malagoli Tagliazucchi
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Gianpiero Di Leva
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Hansjuerg Alder
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Paolo Fadda
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Justin Middleton
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Karl J Smalley
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Tommaso Selmi
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Srivatsava Naidu
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - John L Farber
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN)
| | - Louise Y Fong
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH (CT, MG, GDL, HA, PF, JM, CMC); Kimmel Cancer Center (HC, YJ, KJS, LYF) and Department of Pathology, Anatomy, and Cell Biology (YJ, JLF, LYF), Thomas Jefferson University, Philadelphia, PA; Center for Genome Research (CT, GMT), Department of Life Sciences (TS), University of Modena and Reggio Emilia, Modena, Italy (CT, GMT); Transcriptional Networks in Lung Cancer Group, Manchester Institute, University of Manchester, UK (MG, SN).
| |
Collapse
|