1
|
Zhao Z, Feng X, Chen B, Wu Y, Wang X, Tang Z, Huang M, Guo X. CDCA genes as prognostic and therapeutic targets in Colon adenocarcinoma. Hereditas 2025; 162:19. [PMID: 39924497 PMCID: PMC11809055 DOI: 10.1186/s41065-025-00368-w] [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: 11/15/2024] [Accepted: 01/13/2025] [Indexed: 02/11/2025] Open
Abstract
OBJECTIVES The study investigates the role of Cell Division Cycle Associated (CDCA) genes in colorectal cancer (COAD) by analyzing their differential expression, epigenetic alterations, prognostic significance, and functional associations. METHODOLOGY This study employed a detailed in silico and in vitro experiments-based methodology. RESULTS RT-qPCR assays reveal significantly elevated mRNA levels of CDCA2, CDCA3, CDCA4, CDCA5, CDCA7, and CDCA8 genes in COAD cell lines compared to controls. Bisulfite sequencing indicates reduced promoter methylation of CDCA gene promoters in COAD cell lines, suggesting an epigenetic regulatory mechanism. Analysis of large TCGA datasets confirms increased CDCA gene expression in COAD tissues. Survival analysis using cSurvival database demonstrates negative correlations between CDCA gene expression and patient overall survival. Additionally, Lasso regression-based models of CDCA genes predict survival outcomes in COAD patients. Investigating immune modulation, CDCA gene expression inversely correlates with immune cell infiltration and immune modulators. miRNA-mRNA network analysis identifies regulatory miRNAs targeting CDCA genes, validated by RT-qPCR showing up-regulation of has-mir-10a-5p and has-mir-20a-5p in COAD cell lines and tissues. Drug sensitivity analysis suggests resistance to specific drugs in COAD patients with elevated CDCA gene expression. Furthermore, CDCA gene expression correlates with crucial functional states in COAD, including "angiogenesis, apoptosis, differentiation, hypoxia, inflammation, and metastasis." Additional in vitro experiments revealed that CDCA2 and CDCA3 knockdown in SW480 and SW629 cells significantly reduced cell proliferation and colony formation while enhancing cell migration. CONCLUSION Overall, the study elucidates the multifaceted role of CDCA genes in COAD progression, providing insights into potential diagnostic, prognostic, and therapeutic implications.
Collapse
Affiliation(s)
- Zongquan Zhao
- Department of General Practice, Pingjiang New Town Community Health Service Center Sujin Street Gusu District, Suzho, 215000, Jiangsu, China
| | - Xinwei Feng
- Department of Digestive Internal Medicine, Shanghai Changzheng Hospital, Shanghai, 200003, China
| | - Bo Chen
- Department of Oncology, Chengdu First People's Hospital, Chengdu Sichuan, 610041, China
| | - Yihong Wu
- Department of General Practice, Runda Community Health Service Center, Wumenqiao Street, Gusu District, Suzhou, 215000, Jiangsu, China
| | - Xiaohong Wang
- Department of General Practice, Pingjiang New Town Community Health Service Center Sujin Street Gusu District, Suzho, 215000, Jiangsu, China
| | - Zhenyuan Tang
- Department of General Practice, Community Health Management Center of Suzhou Municipal Hospital, Suzhou, 215000, Jiangsu, China
| | - Min Huang
- Department of General Practice, Suzhou Municipal Hospital, Suzhou, 215000, Jiangsu, China
| | - Xiaohua Guo
- Department of Digestive Surgery, Xi'an Jiaotong University School of Medicine Affiliated Honghui Hospital, Xi'an, Shaanxi, 700054, China.
| |
Collapse
|
2
|
Grimes JM, Ghosh S, Manzoor S, Li LX, Moran MM, Clements JC, Alexander SD, Markert JM, Leavenworth JW. Oncolytic reprogramming of tumor microenvironment shapes CD4 T-cell memory via the IL6ra-Bcl6 axis for targeted control of glioblastoma. Nat Commun 2025; 16:1095. [PMID: 39885128 PMCID: PMC11782536 DOI: 10.1038/s41467-024-55455-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/13/2024] [Indexed: 02/01/2025] Open
Abstract
Oncolytic viruses (OVs) emerge as a promising cancer immunotherapy. However, the temporal impact on tumor cells and the tumor microenvironment, and the nature of anti-tumor immunity post-therapy remain largely unclear. Here we report that CD4+ T cells are required for durable tumor control in syngeneic murine models of glioblastoma multiforme after treatment with an oncolytic herpes simplex virus (oHSV) engineered to express IL-12. The upregulated MHCII on residual tumor cells facilitates programmed polyfunctional CD4+ T cells for tumor control and for recall responses. Mechanistically, the proper ratio of Bcl-6 to T-bet in CD4+ T cells navigates their enhanced anti-tumor capacity, and a reciprocal IL6ra-Bcl-6 regulatory axis in a memory CD4+ T-cell subset, which requires MHCII signals from reprogrammed tumor cells, tumor-infiltrating and resident myeloid cells, is necessary for the prolonged response. These findings uncover an OV-induced tumor/myeloid-CD4+ T-cell partnership, leading to long-term anti-tumor immune memory, and improved OV therapeutic efficacy.
Collapse
Affiliation(s)
- Jeffrey M Grimes
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadashib Ghosh
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shamza Manzoor
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Li X Li
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Monica M Moran
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- Graduate Biomedical Sciences Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jennifer C Clements
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sherrie D Alexander
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- The O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianmei W Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
- The O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Parry TL, Gilmore LA, Khamoui AV. Pan-cancer secreted proteome and skeletal muscle regulation: insight from a proteogenomic data-driven knowledge base. Funct Integr Genomics 2025; 25:14. [PMID: 39812750 DOI: 10.1007/s10142-024-01524-7] [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: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025]
Abstract
Large-scale, pan-cancer analysis is enabled by data driven knowledge bases that link tumor molecular profiles with phenotypes. A debilitating cancer-related phenotype is skeletal muscle loss, or cachexia, which occurs partly from tumor products secreted into circulation. Using the LinkedOmicsKB knowledge base assembled from the Clinical Proteomics Tumor Analysis Consortium proteogenomic analysis, along with catalogs of human secretome proteins, ligand-receptor pairs and molecular signatures, we sought to identify candidate pan-cancer proteins secreted to blood that could regulate skeletal muscle phenotypes in multiple solid cancers. Tumor proteins having significant pan-cancer associations with muscle were referenced against secretome proteins secreted to blood from the Human Protein Atlas, then verified as increased in paired tumor vs. normal tissues in pan-cancer manner. This workflow revealed seven secreted proteins from cancers afflicting kidneys, head and neck, lungs and pancreas that classified as protein-binding activity modulator, extracellular matrix protein or intercellular signaling molecule. Concordance of these biomarkers with validated molecular signatures of cachexia and senescence supported relevance to muscle and cachexia disease biology, and high tumor expression of the biomarker set associated with lower overall survival. In this article, we discuss avenues by which skeletal muscle and cachexia may be regulated by these candidate pan-cancer proteins secreted to blood, and conceptualize a strategy that considers them collectively as a biomarker signature with potential for refinement by data analytics and radiogenomics for predictive testing of future risk in a non-invasive, blood-based panel amenable to broad uptake and early management.
Collapse
Affiliation(s)
- Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA.
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA.
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
4
|
Shibu S, Vasa S, Samantaray S, Joshi N, Zala D, G Chaudhari R, Chauhan K, Patel H, Parekh B, Modi A. A bioinformatics analysis of gene expression in endometrial cancer, endometriosis and obesity. Women Health 2025; 65:60-70. [PMID: 39653677 DOI: 10.1080/03630242.2024.2437493] [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: 06/22/2024] [Revised: 10/29/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Endometrial cancer (EC), endometriosis (ENDO), and obesity (OBY) are interconnected conditions in women that may share underlying genetic pathways. This study aimed to identify shared genetic pathways and differential gene expressions across these conditions to uncover potential therapeutic targets. A bioinformatics pipeline was applied using gene expression datasets from the GEO database, incorporating differential expression analysis, functional and pathway enrichment, PPI network construction, survival analysis, and mutational profiling across 198 samples. The analysis revealed 26 shared differentially expressed genes (DEGs), with IGF-1, CREBBP, EP300, and PIAS1 identified as key hub genes. Elevated IGF-1 expression was significantly linked to poorer survival outcomes in EC patients (p < .05). Frequent mutations were observed in these hub genes, suggesting their critical role in disease mechanisms. This study highlights genetic links among EC, ENDO, and OBY, emphasizing high IGF-1 expression as a potential prognostic marker in EC and recurrent alterations in hub genes as promising therapeutic targets. These findings provide insights into the shared genetic underpinnings of these conditions and present new avenues for targeted therapies.
Collapse
Affiliation(s)
- Shan Shibu
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Shrinal Vasa
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Swayamprabha Samantaray
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Nidhi Joshi
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Dolatsinh Zala
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Rajeshkumar G Chaudhari
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Kartik Chauhan
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Harsh Patel
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| | - Bhavin Parekh
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
- Department of Validation of Indic Knowledge Through Advanced Research, Gujarat University, Ahmedabad, India
| | - Anupama Modi
- School Of Applied Sciences and Technology, Gujarat Technological University, Ahmedabad, India
| |
Collapse
|
5
|
Cai K, Fu W, Liu H, Yang X, Wang Z, Zhao X. Leveraging Bioinformatics and Machine Learning for Identifying Prognostic Biomarkers and Predicting Clinical Outcomes in Lung Adenocarcinoma. Genes (Basel) 2024; 15:1497. [PMID: 39766765 PMCID: PMC11675206 DOI: 10.3390/genes15121497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: There exist significant challenges for lung adenocarcinoma (LUAD) due to its poor prognosis and limited treatment options, particularly in the advanced stages. It is crucial to identify genetic biomarkers for improving outcome predictions and guiding personalized therapies. Methods: In this study, we utilize a multi-step approach that combines principled sure independence screening, penalized regression methods and information gain to identify the key genetic features of the ultra-high dimensional RNA-sequencing data from LUAD patients. We then evaluate three methods of survival analysis: the Cox model, survival tree, and random survival forests (RSFs), to compare their predictive performance. Additionally, a protein-protein interaction network is used to explore the biological significance of identified genes. Results:DKK1 and TNS4 are consistently selected as significant predictors across all feature selection methods. The Kaplan-Meier method shows that high expression levels of these genes are strongly correlated with poorer survival outcomes, suggesting their potential as prognostic biomarkers. RSF outperforms Cox and survival tree methods, showing higher AUC and C-index values. The protein-protein interaction network highlights key nodes such as VEGFC and LAMA3, which play central roles in LUAD progression. Conclusions: Our findings provide valuable insights into the genetic mechanisms of LUAD. These results contribute to the development of more accurate prognostic tools and personalized treatment strategies for LUAD.
Collapse
Affiliation(s)
- Kaida Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China
- Department of Statistics and Actuarial Science, School of Mathematics, Southeast University, Nanjing 211189, China; (W.F.); (H.L.); (X.Y.); (Z.W.); (X.Z.)
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Wenzhi Fu
- Department of Statistics and Actuarial Science, School of Mathematics, Southeast University, Nanjing 211189, China; (W.F.); (H.L.); (X.Y.); (Z.W.); (X.Z.)
| | - Hanwen Liu
- Department of Statistics and Actuarial Science, School of Mathematics, Southeast University, Nanjing 211189, China; (W.F.); (H.L.); (X.Y.); (Z.W.); (X.Z.)
| | - Xiaofang Yang
- Department of Statistics and Actuarial Science, School of Mathematics, Southeast University, Nanjing 211189, China; (W.F.); (H.L.); (X.Y.); (Z.W.); (X.Z.)
| | - Zhengyan Wang
- Department of Statistics and Actuarial Science, School of Mathematics, Southeast University, Nanjing 211189, China; (W.F.); (H.L.); (X.Y.); (Z.W.); (X.Z.)
| | - Xin Zhao
- Department of Statistics and Actuarial Science, School of Mathematics, Southeast University, Nanjing 211189, China; (W.F.); (H.L.); (X.Y.); (Z.W.); (X.Z.)
- Key Laboratory of Measurement and Control of Complex Systems of Engineering, Ministry of Education, Southeast University, Nanjing 210096, China
| |
Collapse
|
6
|
Majd E, Xing L, Zhang X. Segmentation of patients with small cell lung cancer into responders and non-responders using the optimal cross-validation technique. BMC Med Res Methodol 2024; 24:83. [PMID: 38589775 PMCID: PMC11000309 DOI: 10.1186/s12874-024-02185-7] [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: 12/04/2022] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND The timing of treating cancer patients is an essential factor in the efficacy of treatment. So, patients who will not respond to current therapy should receive a different treatment as early as possible. Machine learning models can be built to classify responders and nonresponders. Such classification models predict the probability of a patient being a responder. Most methods use a probability threshold of 0.5 to convert the probabilities into binary group membership. However, the cutoff of 0.5 is not always the optimal choice. METHODS In this study, we propose a novel data-driven approach to select a better cutoff value based on the optimal cross-validation technique. To illustrate our novel method, we applied it to three clinical trial datasets of small-cell lung cancer patients. We used two different datasets to build a scoring system to segment patients. Then the models were applied to segment patients into the test data. RESULTS We found that, in test data, the predicted responders and non-responders had significantly different long-term survival outcomes. Our proposed novel method segments patients better than the standard approach using a cutoff of 0.5. Comparing clinical outcomes of responders versus non-responders, our novel method had a p-value of 0.009 with a hazard ratio of 0.668 for grouping patients using the Cox proportion hazard model and a p-value of 0.011 using the accelerated failure time model which approved a significant difference between responders and non-responders. In contrast, the standard approach had a p-value of 0.194 with a hazard ratio of 0.823 using the Cox proportion hazard model and a p-value of 0.240 using the accelerated failure time model indicating the responders and non-responders do not differ significantly in survival. CONCLUSION In summary, our novel prediction method can successfully segment new patients into responders and non-responders. Clinicians can use our prediction to decide if a patient should receive a different treatment or stay with the current treatment.
Collapse
Affiliation(s)
- Elham Majd
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada
| | - Li Xing
- Department of Mathematics and Statistics, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xuekui Zhang
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
7
|
Saghapour E, Yue Z, Sharma R, Kumar S, Sembay Z, Willey CD, Chen JY. Explorative Discovery of Gene Signatures and Clinotypes in Glioblastoma Cancer Through GeneTerrain Knowledge Map Representation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587278. [PMID: 38617348 PMCID: PMC11014492 DOI: 10.1101/2024.04.01.587278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
This study introduces the GeneTerrain Knowledge Map Representation (GTKM), a novel method for visualizing gene expression data in cancer research. GTKM leverages protein-protein interactions to graphically display differentially expressed genes (DEGs) on a 2-dimensional contour plot, offering a more nuanced understanding of gene interactions and expression patterns compared to traditional heatmap methods. The research demonstrates GTKM's utility through four case studies on glioblastoma (GBM) datasets, focusing on survival analysis, subtype identification, IDH1 mutation analysis, and drug sensitivities of different tumor cell lines. Additionally, a prototype website has been developed to showcase these findings, indicating the method's adaptability for various cancer types. The study reveals that GTKM effectively identifies gene patterns associated with different clinical outcomes in GBM, and its profiles enable the identification of sub-gene signature patterns crucial for predicting survival. The methodology promises significant advancements in precision medicine, providing a powerful tool for understanding complex gene interactions and identifying potential therapeutic targets in cancer treatment.
Collapse
Affiliation(s)
- Ehsan Saghapour
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, US
| | - Zongliang Yue
- Health Outcome Research and Policy Department, Harrison College of Pharmacy, Auburn University, AL, US
| | - Rahul Sharma
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, US
| | - Sidharth Kumar
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, US
| | - Zhandos Sembay
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, US
| | - Christopher D Willey
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, US
| | - Jake Y Chen
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, US
- Systems Pharmacology AI Research Center, University of Alabama at Birmingham, AL, US
| |
Collapse
|
8
|
Chen KC, Dhar T, Chen CR, Chen ECY, Peng CC. Nicotinamide phosphoribosyltransferase modulates PD-L1 in bladder cancer and enhances immunotherapeutic sensitivity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167106. [PMID: 38428685 DOI: 10.1016/j.bbadis.2024.167106] [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: 10/31/2023] [Revised: 02/11/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Bladder cancer (BLCA) is one of the most prevalent malignancies worldwide with a high mortality rate and poor response to immunotherapy in patients expressing lower programmed death ligand 1 (PD-L1) levels. Nicotinamide phosphoribosyltransferase (NAMPT), a rate-limiting enzyme responsible for the biosynthesis of nicotinamide adenine dinucleotide (NAD+) from nicotinamide was reported to be overexpressed in various cancers; however, the role of NAMPT in BLCA is obscure. Immunohistochemistry of tissue microarrays, a real-time polymerase chain reaction, Western blotting, proliferation assay, NAD+ quantification, transwell-migration assay, and colony-formation assay were performed to measure NAMPT and PD-L1 expression levels in patients and the effect of NAMPT inhibition on T24 cells. Our study revealed that NAMPT expression was upregulated in BLCA patients with different grades and associated with poor T-cell infiltration. Notably, FK866-mediated NAMPT inhibition decreased cell viability by depleting NAD+, and reducing the migration ability and colony-formation ability of T24 cells. Interestingly, NAMPT negatively regulated PD-L1 under an interferon (IFN)-γ-mediated microenvironment. However, exogenous NAMPT activator has no effect on PD-L1. NAD+ supplementation also only increased PD-L1 in the absence of IFN-γ. Conclusively, NAMPT is crucial for BLCA tumorigenic properties, and it regulates expression of the PD-L1 immune checkpoint protein. NAMPT could be considered a target for modulating sensitivity to immunotherapy.
Collapse
Affiliation(s)
- Kuan-Chou Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Urology, Taipei Medical University Shuang-Ho Hospital, Zhong-He District, New Taipei City 23561, Taiwan; Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; TMU-Research Center of Urology and Kidney, Taipei Medical University, Taipei, 11031, Taiwan
| | - Trayee Dhar
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chang-Rong Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Eugene Chang-Yu Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chiung-Chi Peng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
9
|
Pugh S, Fosdick BK, Nehring M, Gallichotte EN, VandeWoude S, Wilson A. Estimating cutoff values for diagnostic tests to achieve target specificity using extreme value theory. BMC Med Res Methodol 2024; 24:30. [PMID: 38331732 PMCID: PMC10851584 DOI: 10.1186/s12874-023-02139-5] [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: 09/11/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Rapidly developing tests for emerging diseases is critical for early disease monitoring. In the early stages of an epidemic, when low prevalences are expected, high specificity tests are desired to avoid numerous false positives. Selecting a cutoff to classify positive and negative test results that has the desired operating characteristics, such as specificity, is challenging for new tests because of limited validation data with known disease status. While there is ample statistical literature on estimating quantiles of a distribution, there is limited evidence on estimating extreme quantiles from limited validation data and the resulting test characteristics in the disease testing context. METHODS We propose using extreme value theory to select a cutoff with predetermined specificity by fitting a Pareto distribution to the upper tail of the negative controls. We compared this method to five previously proposed cutoff selection methods in a data analysis and simulation study. We analyzed COVID-19 enzyme linked immunosorbent assay antibody test results from long-term care facilities and skilled nursing staff in Colorado between May and December of 2020. RESULTS We found the extreme value approach had minimal bias when targeting a specificity of 0.995. Using the empirical quantile of the negative controls performed well when targeting a specificity of 0.95. The higher target specificity is preferred for overall test accuracy when prevalence is low, whereas the lower target specificity is preferred when prevalence is higher and resulted in less variable prevalence estimation. DISCUSSION While commonly used, the normal based methods showed considerable bias compared to the empirical and extreme value theory-based methods. CONCLUSIONS When determining disease testing cutoffs from small training data samples, we recommend using the extreme value based-methods when targeting a high specificity and the empirical quantile when targeting a lower specificity.
Collapse
Affiliation(s)
- Sierra Pugh
- Department of Statistics, Colorado State University, 102 Statistics Building, Fort Collins, 80523, Colorado, USA
| | - Bailey K Fosdick
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Mary Nehring
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Emily N Gallichotte
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Sue VandeWoude
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Ander Wilson
- Department of Statistics, Colorado State University, 102 Statistics Building, Fort Collins, 80523, Colorado, USA.
| |
Collapse
|
10
|
Zhang Y, Zhou Y, Zhou Y, Yu X, Shen X, Hong Y, Zhang Y, Wang S, Mou M, Zhang J, Tao L, Gao J, Qiu Y, Chen Y, Zhu F. TheMarker: a comprehensive database of therapeutic biomarkers. Nucleic Acids Res 2024; 52:D1450-D1464. [PMID: 37850638 PMCID: PMC10767989 DOI: 10.1093/nar/gkad862] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023] Open
Abstract
Distinct from the traditional diagnostic/prognostic biomarker (adopted as the indicator of disease state/process), the therapeutic biomarker (ThMAR) has emerged to be very crucial in the clinical development and clinical practice of all therapies. There are five types of ThMAR that have been found to play indispensable roles in various stages of drug discovery, such as: Pharmacodynamic Biomarker essential for guaranteeing the pharmacological effects of a therapy, Safety Biomarker critical for assessing the extent or likelihood of therapy-induced toxicity, Monitoring Biomarker indispensable for guiding clinical management by serially measuring patients' status, Predictive Biomarker crucial for maximizing the clinical outcome of a therapy for specific individuals, and Surrogate Endpoint fundamental for accelerating the approval of a therapy. However, these data of ThMARs has not been comprehensively described by any of the existing databases. Herein, a database, named 'TheMarker', was therefore constructed to (a) systematically offer all five types of ThMAR used at different stages of drug development, (b) comprehensively describe ThMAR information for the largest number of drugs among available databases, (c) extensively cover the widest disease classes by not just focusing on anticancer therapies. These data in TheMarker are expected to have great implication and significant impact on drug discovery and clinical practice, and it is freely accessible without any login requirement at: https://idrblab.org/themarker.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Diagnosis and Treatment of Severe Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yuan Zhou
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyuan Yu
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yanfeng Hong
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuxin Zhang
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shanshan Wang
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jinsong Zhang
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunqing Qiu
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Diagnosis and Treatment of Severe Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yuzong Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
11
|
Dubois‐Chevalier J, Gheeraert C, Berthier A, Boulet C, Dubois V, Guille L, Fourcot M, Marot G, Gauthier K, Dubuquoy L, Staels B, Lefebvre P, Eeckhoute J. An extended transcription factor regulatory network controls hepatocyte identity. EMBO Rep 2023; 24:e57020. [PMID: 37424431 PMCID: PMC10481658 DOI: 10.15252/embr.202357020] [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: 02/16/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/11/2023] Open
Abstract
Cell identity is specified by a core transcriptional regulatory circuitry (CoRC), typically limited to a small set of interconnected cell-specific transcription factors (TFs). By mining global hepatic TF regulons, we reveal a more complex organization of the transcriptional regulatory network controlling hepatocyte identity. We show that tight functional interconnections controlling hepatocyte identity extend to non-cell-specific TFs beyond the CoRC, which we call hepatocyte identity (Hep-ID)CONNECT TFs. Besides controlling identity effector genes, Hep-IDCONNECT TFs also engage in reciprocal transcriptional regulation with TFs of the CoRC. In homeostatic basal conditions, this translates into Hep-IDCONNECT TFs being involved in fine tuning CoRC TF expression including their rhythmic expression patterns. Moreover, a role for Hep-IDCONNECT TFs in the control of hepatocyte identity is revealed in dedifferentiated hepatocytes where Hep-IDCONNECT TFs are able to reset CoRC TF expression. This is observed upon activation of NR1H3 or THRB in hepatocarcinoma or in hepatocytes subjected to inflammation-induced loss of identity. Our study establishes that hepatocyte identity is controlled by an extended array of TFs beyond the CoRC.
Collapse
Affiliation(s)
| | - Céline Gheeraert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| | - Alexandre Berthier
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| | - Clémence Boulet
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| | - Vanessa Dubois
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
- Basic and Translational Endocrinology (BaTE), Department of Basic and Applied Medical SciencesGhent UniversityGhentBelgium
| | - Loïc Guille
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| | - Marie Fourcot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 – UAR 2014 – PLBSLilleFrance
| | - Guillemette Marot
- Univ. Lille, Inria, CHU Lille, ULR 2694 – METRICS: Évaluation des technologies de santé et des pratiques médicalesLilleFrance
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon (IGFL), CNRS UMR 5242, INRAE USC 1370, École Normale Supérieure de LyonLyonFrance
| | - Laurent Dubuquoy
- Univ. Lille, Inserm, CHU Lille, U1286 – INFINITE – Institute for Translational Research in InflammationLilleFrance
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| | - Philippe Lefebvre
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| | - Jérôme Eeckhoute
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011‐EGIDLilleFrance
| |
Collapse
|
12
|
Liu HT, Chen SY, Peng LL, Zhong L, Zhou L, Liao SQ, Chen ZJ, Wang QL, He S, Zhou ZH. Spatially resolved transcriptomics revealed local invasion-related genes in colorectal cancer. Front Oncol 2023; 13:1089090. [PMID: 36816947 PMCID: PMC9928961 DOI: 10.3389/fonc.2023.1089090] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Objective Local invasion is the first step of metastasis, the main cause of colorectal cancer (CRC)-related death. Recent studies have revealed extensive intertumoral and intratumoral heterogeneity. Here, we focused on revealing local invasion-related genes in CRC. Methods We used spatial transcriptomic techniques to study the process of local invasion in four CRC tissues. First, we compared the pre-cancerous, cancer center, and invasive margin in one section (S115) and used pseudo-time analysis to reveal the differentiation trajectories from cancer center to invasive margin. Next, we performed immunohistochemical staining for RPL5, STC1, AKR1B1, CD47, and HLA-A on CRC samples. Moreover, we knocked down AKR1B1 in CRC cell lines and performed CCK-8, wound healing, and transwell assays to assess cell proliferation, migration, and invasion. Results We demonstrated that 13 genes were overexpressed in invasive clusters, among which the expression of CSTB and TM4SF1 was correlated with poor PFS in CRC patients. The ribosome pathway was increased, while the antigen processing and presentation pathway was decreased along CRC progression. RPL5 was upregulated, while HLA-A was downregulated along cancer invasion in CRC samples. Pseudo-time analysis revealed that STC1, AKR1B1, SIRPA, C4orf3, EDNRA, CES1, PRRX1, EMP1, PPIB, PLTP, SULF2, and EGFL6 were unpregulated along the trajectories. Immunohistochemic3al staining showed the expression of STC1, AKR1B1, and CD47 was increased along cancer invasion in CRC samples. Knockdown of AKR1B1 inhibited CRC cells' proliferation, migration, and invasion. Conclusions We revealed the spatial heterogeneity within CRC tissues and uncovered some novel genes that were associated with CRC invasion.
Collapse
Affiliation(s)
- Hong-Tao Liu
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si-Yuan Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Ling-Long Peng
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhong
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Si-Qi Liao
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Ji Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing-Liang Wang
- Department of Pathology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song He
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Zhi-Hang Zhou, ; Song He,
| | - Zhi-Hang Zhou
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Zhi-Hang Zhou, ; Song He,
| |
Collapse
|