1
|
Li H, Su B, Jiang Y, Zhang B, Du R, Song C, Hou B, Xu K, Wu L, Gu Y. Circular RNA circDCUN1D4 suppresses hepatocellular carcinoma development via targeting the miR-590-5p/ TIMP3 axis. Mol Cancer 2025; 24:95. [PMID: 40128740 PMCID: PMC11934760 DOI: 10.1186/s12943-025-02300-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global health concern, necessitating innovative therapeutic strategies. In this study, we investigated the functional role of circular RNA circDCUN1D4 in HCC progression and its potential therapeutic implications. It was found that HCC patients exhibiting higher levels of circDCUN1D4 demonstrated a more favorable survival rate. Furthermore, we revealed that circDCUN1D4 suppressed HCC cell proliferation, migration, and invasion. Mechanistically, circDCUN1D4 was identified as a sponge for miR-590-5p, leading to the downregulation of its downstream target, Tissue Inhibitor of Metalloproteinase 3 (TIMP3). Importantly, circDCUN1D4 administration through In vivo jet-PEI exhibited a robust inhibitory effect on tumor progression without causing notable toxicity in mice. Overall, our findings highlight circDCUN1D4 as a promising therapeutic candidate for HCC, unraveling its intricate regulatory role through the miR-590-5p/TIMP3 axis. This study contributes valuable insights into the potential clinical applications of circRNA-based therapies for HCC.
Collapse
Affiliation(s)
- Hongyu Li
- College of Chemistry and Lie Science, Beijing University of Technology, Beijing, 100124, China.
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China.
| | - Bing Su
- College of Chemistry and Lie Science, Beijing University of Technology, Beijing, 100124, China
| | - Yan Jiang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Boyang Zhang
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China
| | - Rulong Du
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China
| | - Can Song
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China
| | - Bin Hou
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China
| | - Kun Xu
- College of Chemistry and Lie Science, Beijing University of Technology, Beijing, 100124, China.
| | - Lida Wu
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China.
| | - Yuchun Gu
- Allife Medical Science and Technology Co., Ltd. Economic and Technological Development Zone, Beijing, 100176, China.
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, 100871, China.
| |
Collapse
|
2
|
Kazemi F, Sadeghian F, Pirsadeghi A, Asadi F, Javdani H, Yousefi-Ahmadipour A. Adipose mesenchymal stem cell conditioned medium and extract: A promising therapeutic option for regenerative breast cancer therapy. SAGE Open Med 2024; 12:20503121241306606. [PMID: 39691866 PMCID: PMC11650577 DOI: 10.1177/20503121241306606] [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: 04/09/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Breast cancer is the second most common cancer and a leading cause of cancer death in U.S. women. The tumor microenvironment, especially nearby adipocytes, plays a crucial role in its progression. Therefore, this study aimed to investigate the effects of human adipose mesenchymal stem cells-derived conditioned medium (SUP) and extract (CE) from on breast cancer cells. Methods Human adipose-derived mesenchymal stem cells were isolated and characterized by flow cytometry using Cluster of Differentiation (CD) markers (CD34, CD45, CD90, and CD105). The differentiation potential was confirmed via adipogenic and osteogenic induction. MCF-7 and MDA-MB-231 cells were treated with SUP and CE, and cell viability was assessed using the 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay at 24, 48, and 72 h. Doubling time, colony formation, wound healing, and gene expression for key cancer-related genes (TIMP1, TIMP2, MMP2, PDL1, IDO, Bax, caspase 3, and caspase 9) were also evaluated. Results Both SUP and CE significantly inhibited the viability of MCF-7 and MDA-MB-231 cells, reduced their doubling time, and suppressed colony formation. In wound healing assays, cell migration was notably impaired in MDA-MB-231 cells but less so in MCF-7 cells. Real-time polymerase chain reaction revealed downregulation of TIMP1, MMP2, PDL1, and IDO in MDA-MB-231 cells after treatment, while CE increased certain gene expressions in MCF-7 cells. Bax, caspase 3, and caspase 9 expressions were significantly upregulated in MDA-MB-231 cells but not in MCF-7 cells after treatment. Conclusion Human adipose-derived mesenchymal stem cells-derived SUP and CE exhibit antitumor effects on breast cancer cells, suggesting a potential therapeutic strategy to suppress tumor progression. Mesenchymal stem cells-SUP and CE could be a safe and novel regenerative approach for breast reconstruction postmastectomy without tumor recurrence risk.
Collapse
Affiliation(s)
- Faezeh Kazemi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Faculty of Paramedicine, Department of Laboratory Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Sadeghian
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Faculty of Paramedicine, Department of Laboratory Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Pirsadeghi
- Faculty of Paramedicine, Department of Laboratory Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Asadi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Cancer and Stem Cell Research Laboratory, Faculty of Paramedicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Javdani
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Aliakbar Yousefi-Ahmadipour
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Faculty of Paramedicine, Department of Laboratory Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Cancer and Stem Cell Research Laboratory, Faculty of Paramedicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
3
|
Chen C, Wang W, Ning C, Lu Z, Zhang M, Zhu Y, Tian J, Li H, Ge Y, Yang B, Miao X. Integrated systematic functional screen and fine-mapping decipher the role and genetic regulation of RPS19 in colorectal cancer development. Arch Toxicol 2024; 98:3453-3465. [PMID: 39012505 DOI: 10.1007/s00204-024-03822-2] [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: 05/09/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Despite genome-wide association studies (GWAS) have identified more than 200 risk loci associated with colorectal cancer (CRC), the causal genes or risk variants within these loci and their biological functions remain not fully revealed. Recently, the genomic locus 19q13.2, with the lead SNP rs1800469 was identified as a crucial CRC risk locus in Asian populations. However, the functional mechanism of this region has not been fully elucidated. Here we employed an RNA interfering-based on-chip approach to screen for the genes essential for cell proliferation in the CRC risk locus 19q13.2. Notably, we found that RPS19 exhibited the most significant effect among the identified genes and acted as a critical oncogene facilitating CRC cell proliferation. Subsequently, combining integrative fine-mapping analysis and a large-scale population study consisting of 6027 cases and 6099 controls, we prioritized rs1025497 as a potential causal candidate for CRC risk, demonstrating that rs1025497[A] allele significantly reduced the risk of CRC (OR 0.70, 95% confidence interval = 0.56-0.83, P = 1.12 × 10-6), which was further validated in UK Biobank cohort comprising 5,313 cases and 21,252 controls. Mechanistically, we experimentally elucidated that variant rs1025497 might acted as an allele-specific silencer, inhibiting the expression level of oncogene RPS19 mediated by the transcription suppressive factor HBP1. Taken together, our sturdy unveils the significant role of RPS19 during CRC pathogenesis and delineates its distal regulatory mechanism mediated by rs1025497, advancing our understanding of the etiology of CRC and provided new insights into the personalized medicine of human cancer.
Collapse
Affiliation(s)
- Can Chen
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenzhuo Wang
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Caibo Ning
- Department of Epidemiology and Biostatistics, School of Public Health, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Zequn Lu
- Department of Epidemiology and Biostatistics, School of Public Health, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Haijie Li
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Ge
- Department of Epidemiology and Biostatistics, School of Public Health, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Beifang Yang
- Hubei Institute for Infectious Disease Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, 430079, China.
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, Key Laboratory for Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Liu X, Ping G, Ji D, Wen Z, Chen Y. Reclassify High-Grade Serous Ovarian Cancer Patients Into Different Molecular Subtypes With Discrepancy Prognoses and Therapeutic Responses Based on Cancer-Associated Fibroblast-Enriched Prognostic Genes. Biomed Eng Comput Biol 2024; 15:11795972241274024. [PMID: 39221174 PMCID: PMC11365035 DOI: 10.1177/11795972241274024] [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: 12/14/2023] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play critical roles in the metastasis and therapeutic response of high-grade serous ovarian cancer (HGSC). Our study intended to select HGSC patients with unfavorable prognoses and therapeutic responses based on CAF-enriched prognostic genes. The bulk RNA and single-cell RNA sequencing (scRNA-seq) data of tumor tissues were collected from the TCGA and GEO databases. The infiltrated levels of immune and stromal cells were estimated by multiple immune deconvolution algorithms and verified through immunohistochemical analysis. The univariate Cox regression analyses were used to identify prognostic genes. Gene Set Enrichment Analysis (GSEA) was conducted to annotate enriched gene sets. The Genomics of Drug Sensitivity in Cancer (GDSC) database was used to explore potential alternative drugs. We found the infiltered levels of CAFs were remarkedly elevated in advanced and metastatic HGSC tissues and identified hundreds of genes specifically enriched in CAFs. Then we selected 6 CAF-enriched prognostic genes based on which HGSC patients were reclassified into 2 subclusters with discrepancy prognoses. Further analysis revealed that the HGSC patients in cluster-2 tended to undergo poor responses to traditional chemotherapy and immunotherapy. Subsequently, we selected 24 novel potential therapeutic drugs for cluster-2 HGSC patients. Moreover, we discovered a positive correlation of infiltrated levels between CAFs and monocytes/macrophages in HGSC tissues. Collectively, our study successfully reclassified HGSC patients into 2 different subgroups that have discrepancy prognoses and responses to current therapeutic methods.
Collapse
Affiliation(s)
- Xiangxiang Liu
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| | - Guoqiang Ping
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongze Ji
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhifa Wen
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| | - Yajun Chen
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Deng Y, Huang J, Chen C, Wen Y, Qiu D. Fe 3O 4 coated stent prevent artery neointimal hyperplasia by inhibiting vascular smooth muscle cell proliferation. Mater Today Bio 2024; 27:101133. [PMID: 39027680 PMCID: PMC11254736 DOI: 10.1016/j.mtbio.2024.101133] [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: 03/17/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
In-stent restenosis (ISR), caused by aggressive vascular smooth muscle cell (VSMC) proliferation, is a serious complication of stenting. Therefore, developing therapeutic approaches that target VSMC inhibition is imperative. Our previous study showed that VSMC hyperplasia was attenuated after iron stent degradation, and VSMC proliferation around the stented section was arrested. The corrosion products of the iron stents were primarily Fe3O4 particles. Therefore, we hypothesized that Fe3O4 particles generated by iron stents would prevent neointimal hyperplasia by inhibiting VSMC proliferation. To test this hypothesis, culture assays and flow cytometry were performed to investigate the proliferation of VSMC. Global gene sequencing and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to investigate the underlying mechanisms. Fe3O4-coated stents were implanted into rabbit carotid arteries to evaluate the inhibitory effects of Fe3O4 on neointimal hyperplasia. The major findings of the study were as follows: 1) Fe3O4 attenuated neointimal hyperplasia by preventing VSMC proliferation after stenting; 2) Fe3O4 exerted inhibitory effects on VSMCs by downregulating proliferative genes such as SOX9, EGR4, and TGFB1, but upregulated inhibitory genes such as DNMT1, TIMP3, and PCNA; 3) Fe3O4 inhibited VSMCs by preventing phenotypic transformation from the contractile to the synthetic phase; and 4) Fe3O4-coated stents achieved satisfactory hemocompatibility in a rabbit model. Our study highlights the additional benefits of Fe3O4 particles in inhibiting VSMC proliferation, indicating that Fe3O4 coated stent potentially served as an attractive therapeutic approach for ISR prevention.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Ultrasonic Imaging, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Cancer Proteomics & Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Jiabing Huang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, PR China
| | - Changqing Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Yanbing Wen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Dongxu Qiu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| |
Collapse
|
6
|
Wen CY, Chen PH, Hsieh FR, Chang R, Wu MY, Yong SB, Li CJ. CSF-1R: A promising therapeutic target for various diseases. Pharmacol Res 2024; 204:107196. [PMID: 38677533 DOI: 10.1016/j.phrs.2024.107196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Affiliation(s)
- Chen-Yueh Wen
- Division of Urology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Po-Hung Chen
- Department of Emergency Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung 802, Taiwan
| | - Feng-Renn Hsieh
- Department of Emergency Medicine, Kaohsiung Municipal United Hospital, Kaohsiung 804, Taiwan
| | - Renin Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan
| | - Su-Boon Yong
- Department of Allergy and Immunology, China Medical University Children's Hospital, Taichung 404, Taiwan; Research Center for Allergy, Immunology, and Microbiome (A.I.M.), China Medical University Hospital, Taichung 404, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynaecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
7
|
Varriano S, Yu A, Xu YQ, Natelson DM, Ramadei A, Kleiman FE. Estrogen receptor alpha (ERα) regulates PARN-mediated nuclear deadenylation and gene expression in breast cancer cells. RNA Biol 2024; 21:14-23. [PMID: 39392174 PMCID: PMC11487348 DOI: 10.1080/15476286.2024.2413821] [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] [Revised: 09/10/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
The estrogen signalling pathway is highly dynamic and primarily mediated by estrogen receptors (ERs) that transcriptionally regulate the expression of target genes. While transcriptional functions of ERs have been widely studied, their roles in RNA biology have not been extensively explored. Here, we reveal a novel biological role of ER alpha (ERα) in mRNA 3' end processing in breast cancer cells, providing an alternative mechanism in regulating gene expression at the post-transcriptional level. We show that ERα activates poly(A) specific ribonuclease (PARN) deadenylase using in vitro assays, and that this activation is further increased by tumour suppressor p53, a factor involved in mRNA processing. Consistent with this, we confirm ERα-mediated activation of nuclear deadenylation by PARN in samples from MCF7 and T47D breast cancer cells that vary in expression of ERα and p53. We further show that ERα can form complex(es) with PARN and p53. Lastly, we identify and validate expression of common mRNA targets of ERα and PARN known to be involved in cell invasion, metastasis and angiogenesis, supporting the functional overlap of these factors in regulating gene expression in a transactivation-independent manner. Together, these results show a new regulatory mechanism by which ERα regulates mRNA processing and gene expression post-transcriptionally, highlighting its contribution to unique transcriptomic profiles and breast cancer progression.
Collapse
Affiliation(s)
- Sophia Varriano
- Chemistry Department, Hunter College, The City University of New York, New York, NY, USA
- Biology Program, The Graduate Center, The City University of New York, New York, NY, USA
| | - Amy Yu
- Chemistry Department, Hunter College, The City University of New York, New York, NY, USA
| | - Yu Qing Xu
- Chemistry Department, Hunter College, The City University of New York, New York, NY, USA
| | - Devorah M. Natelson
- Chemistry Department, Hunter College, The City University of New York, New York, NY, USA
- Biology Program, The Graduate Center, The City University of New York, New York, NY, USA
| | - Anthony Ramadei
- Chemistry Department, Hunter College, The City University of New York, New York, NY, USA
- Biology Program, The Graduate Center, The City University of New York, New York, NY, USA
| | - Frida E. Kleiman
- Chemistry Department, Hunter College, The City University of New York, New York, NY, USA
- Biology Program, The Graduate Center, The City University of New York, New York, NY, USA
| |
Collapse
|
8
|
Jia X, Zhang T, Lv X, Du H, Sun Y, Guan Y. Identification of the six-hormone secretion-related gene signature as a prognostic biomarker for colon adenocarcinoma. Cancer Biomark 2023; 38:523-535. [PMID: 38143338 DOI: 10.3233/cbm-230126] [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] [Indexed: 12/26/2023]
Abstract
BACKGROUND Colon adenocarcinoma (COAD) is a globally prevalent cancer, with hormone secretion playing a crucial role in its progression. Despite this, there is limited understanding of the impact of hormone secretion on COAD prognosis. This study aimed to establish a prognostic signature based on hormone secretion-related genes and to elucidate the potential functional mechanisms of these genes in COAD. METHODS Using data from The Cancer Genome Atlas COAD cohort (TCGA-COAD), six hormone secretion-related genes were identified (CYP19A1, FOXD1, GRP, INHBB, SPP1, and UCN). These genes were used to develop a Hormone secretion score (HSS), which was then evaluated using the Kaplan-Meier curve and multivariable Cox analysis. The HSS model was further validated with external GEO cohorts (GSE41258, GSE39582, and GSE87211). Functional enrichment analyses were performed, and the CIBERSORT and TIDE algorithms were used to assess tumor infiltration. RESULTS The study developed a prognostic signature, dividing patients into HSS-high and HSS-low groups. The HSS-high group showed a notably worse prognosis within the TCGA-COAD dataset and in three independent datasets: GSE41258, GSE39582, and GSE87211. Moreover, the HSS-high group predicted a shorter overall survival rate in patients maintaining microsatellite stability (MSS). The functional analysis associated HSS-high with the hypoxic, epithelial-mesenchymal transition (EMT), and TGF-β signaling pathways and correlated with distant and lymph node metastases. The tumor immune microenvironment analysis revealed an elevated CIBERSORT score in the HSS-high group, suggesting an association with tumor metastasis. Further, the HSS-high group showed a higher TIDE score, indicating that patients with high HSS scores are less likely to benefit from Immune Checkpoint Inhibitor (ICI) therapy. CONCLUSIONS This study demonstrated the prognostic significance of a HSS signature based on six hormone secretion-related genes in COAD. The findings suggest that this gene signature may serve as a reliable biomarker for predicting survival outcomes in COAD patients.
Collapse
Affiliation(s)
- Xiongjie Jia
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Heibei, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Heibei, China
| | - Tao Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Heibei, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Hebei University, Baoding, Heibei, China
| | - Xinze Lv
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Haiwei Du
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Yongkun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Guan
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|