1
|
Comertpay B, Gov E. Multiomics Analysis and Machine Learning-based Identification of Molecular Signatures for Diagnostic Classification in Liver Disease Types Along the Microbiota-gut-liver Axis. J Clin Exp Hepatol 2025; 15:102552. [PMID: 40292334 PMCID: PMC12019836 DOI: 10.1016/j.jceh.2025.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Background Liver disease, responsible for around two million deaths annually, remains a pressing global health challenge. Microbial interactions within the microbiota-gut-liver axis play a substantial role in the pathogenesis of various liver conditions, including early chronic liver disease (eCLD), chronic liver disease (CLD), acute liver failure (ALF), acute-on-chronic liver failure (ACLF), non-alcoholic fatty liver disease (NAFLD), steatohepatitis, and cirrhosis. This study aimed to identify key molecular signatures involved in liver disease progression by analyzing transcriptomic and gut microbiome data, and to evaluate their diagnostic utility using machine learning models. Methods Transcriptomic analysis identified differentially expressed genes (DEGs) that, when integrated with regulatory elements microRNAs, transcription factors, receptors, and the gut microbiome highlight disease-specific molecular interactions. To assess the diagnostic potential of these molecular signatures, a two-step analysis involving principal component analysis (PCA) and Random Forest classification was conducted, achieving accuracies of 75% for ALF and 89% for NAFLD. Additionally, machine learning algorithms, including K-neighbors, multi-layer perceptron (MLP), decision tree, Random Forest, logistic regression, gradient boosting, CatBoost, Extreme Gradient Boosting (XGB), and Light Gradient Boosting Machine (LGBM), were applied to gene expression data for ALF and NAFLD. Results Key genes including CLDN14, EGFR, GSK3B, MYC, and TJP2, alongside regulatory miRNAs let-7a-5p, miR-124-3p, and miR-195-5p and transcription factors NFKB1 and SP1 may be suggested as critical to liver disease progression. Additionally, gut microbiota members, Dictyostelium discoideum and Eikenella might be novel candidates associated with liver disease, highlighting the importance of the gut-liver axis. The Random Forest model reached 75% accuracy and 83% area under the curve for ALF, while NAFLD classification achieved 100% accuracy, precision, recall, and area under the curve underscoring robust diagnostic potential. Conclusion This study establishes a solid foundation for further research and therapeutic advancement by identifying key biomolecules and pathways critical to liver disease. Additional experimental validation is needed to confirm clinical applicability.
Collapse
Affiliation(s)
- Betul Comertpay
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| | - Esra Gov
- Department of Bioengineering, Faculty of Engineering, Adana Alparslan Türkeş Science and Technology University, Adana, Turkey
| |
Collapse
|
2
|
Yang R, Yin N, Faiola F. Integrative transcriptomics analysis reveals convergent toxicological effects of perfluorooctanoic acid and perfluorooctane sulfonate on human liver: Evidence from multiple models. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138112. [PMID: 40203759 DOI: 10.1016/j.jhazmat.2025.138112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/12/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025]
Abstract
Perfluorooctanoic acid and perfluorooctane sulfonate are well-known eight-carbon per- and polyfluoroalkyl substances (8C-PFAS) potentially toxic for the human liver. However, direct experimental evidence demonstrating their toxicity on the human liver remains limited. Consequently, this study aimed to extrapolate the 8C-PFAS liver toxicity mechanisms by leveraging omics data to integrate mouse and human findings. Through integration analyses of nine datasets (one human, six murine, and two rat), we identified 199 genes with known biological functions that are commonly affected by 8C-PFAS across species. We delineated a comprehensive regulatory network of 8C-PFAS toxicity, demonstrating that 8C-PFAS may trigger fatty liver disease by up-regulating CD36 and PPARα pathway; dysregulate xenobiotic metabolism by disrupting CAR and CYP family genes; and induce cancer by dysregulating WNT, TGFβ, FGF21, and P53 pathways. We also identified ATF3, EGR1, ESR1, NFATC4, SNAI2, TP53, and EZH2 as transcriptionally regulated by 8C-PFAS, along with PPARα, RXRα, FGFR1, TCF3, and SMAD3 as potentially functionally impacted. Collectively, these factors account for over 90 % of 8C-PFAS-affected key genes. This study not only developed a novel method for extrapolating human toxicity risks by integrating scattered toxicity evidence based on transcriptomics data, but also proposes new mechanisms by which 8C-PFAS contributes to fatty liver disease and cancer.
Collapse
Affiliation(s)
- Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Silkina M, Razumovskaya A, Kulagin T, Fatkulin A, Klycheva K, Olkhovik D, Averinskaya D, Kolodeeva O, Kolodeeva O, Tonevitsky A, Nikulin S. Divergent ferroptotic pathways in breast cancer cells: IGFBP6-regulated mitochondrial lipid peroxidation under erastin and omega-3 DHA treatment. Biochimie 2025; 234:48-61. [PMID: 40180025 DOI: 10.1016/j.biochi.2025.03.010] [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: 02/19/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Breast cancer remains a major challenge and new therapeutic approaches are needed for its treatment. Ferroptosis is considered a promising alternative cell death mechanism to eliminate resistant cancer cells. In previous works, we identified that lower IGFBP6 gene expression in tumor tissue corresponds to a worse prognosis for breast cancer patients and, at the same, time makes them more sensitive to ferroptosis. In this study, we further investigated the mechanism of ferroptosis induction in IGFBP6 knockdown and control MDA-MB-231 breast cancer cells by the canonical ferroptosis inducer erastin and omega-3 docosahexaenoic acid (DHA). Our results indicate that there is a significant overlap between the mechanisms of action of both of these molecules, as they regulate the same subset of genes, and their action can be inhibited by canonical ferroptosis inhibitors. On the other hand, we also observed significant differences between the effects of erastin and DHA. The most notable of these are the additional activation of apoptosis-related genes by DHA and its minor peroxidation of mitochondrial lipid membranes. Interestingly, our kinetic analysis of ferroptosis induction showed that IGFBP6 knockdown cells began to die earlier and could hardly be rescued from erastin-induced ferroptosis by mitochondrial antioxidant SkQ1, in contrast to control cells. Overall, our data suggest that the action of DHA is less dependent on mitochondrial membrane peroxidation during ferroptosis induction, and this molecule can be a promising candidate for the treatment of breast cancer, especially in the case of reduced IGFBP6 gene expression in cancer cells.
Collapse
Affiliation(s)
- Mariia Silkina
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Alexandra Razumovskaya
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia; P. A. Hertsen Moscow Oncology Research Center, Branch of the National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Timur Kulagin
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Artem Fatkulin
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Karina Klycheva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Darya Olkhovik
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Darya Averinskaya
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Oksana Kolodeeva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Olga Kolodeeva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Art Photonics GmbH, Berlin, Germany
| | - Sergey Nikulin
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia.
| |
Collapse
|
4
|
Huang P, Chen Y, Shi Y, Zhong C, Lin H, Yu X, Chen K, Huang Z, Zhang L, Fang S, Lu J, Chen J. Phosphoribosyl transferase domain containing 1: A prognostic biomarker in testicular germ cell tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200958. [PMID: 40241724 PMCID: PMC12001118 DOI: 10.1016/j.omton.2025.200958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
Due to the heterogeneity and complex classification of testicular germ cell tumors (TGCTs), prognostic evaluation and therapeutic targets remain unclear. Therefore, identifying a novel biomarker to comprehensively assess TGCT prognosis and immunotherapy response is crucial. We collected data from 457 TGCT patient samples and 12 normal testicular samples across six cohorts. Differential expression analysis combined with univariate Cox regression identified prognostic markers for TGCT. Multivariate Cox regression and survival analysis further evaluated the prognostic value of phosphoribosyl transferase domain containing 1 (PRTFDC1). Immunohistochemistry on tissue microarrays validated PRTFDC1's predictive value in clinical samples. We then investigated the relationship between PRTFDC1 and somatic mutations, copy number variations, immune cell infiltration, and immunotherapy response. Through these analyses, we identified PRTFDC1 as an independent risk factor indicating poor prognosis in TGCT. Immunohistochemistry demonstrated high PRTFDC1 expression in TGCT tissues. Gene set enrichment analysis revealed that PRTFDC1 suppresses immune-related pathways. Immune infiltration showed that high PRTFDC1 expression is associated with low CD8+ T cell infiltration. Immunotherapy response analysis indicated that low PRTFDC1 expression predicts better immunotherapy response and favorable prognosis. In conclusion, this study elucidates the biological and clinical significance of PRTFDC1, suggesting it as an effective and reliable biomarker for predicting TGCT prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Peisheng Huang
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Yihao Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Yongcheng Shi
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Huawei Lin
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Xiaoxue Yu
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Kai Chen
- Department of Pathology, Guangzhou Medical University Affiliated Women and Children′s Medical Center, Guangzhou 510623, China
| | - Zhuoya Huang
- Department of Pathology, Huizhou Central People’s Hospital, No. 41, Eling North Road, Huizhou, Guangdong 516001, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California, Riverside, CA 92507, USA
| | - Shumin Fang
- Science Research Center, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Jianming Lu
- Department of Andrology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Jiahong Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| |
Collapse
|
5
|
Wu W, Song X, Li B. Identification of VDAC1 as a mitochondria-related target of Duchenne muscular dystrophy based on bioinformatics analysis and in vitro experiments. Int Immunopharmacol 2025; 158:114836. [PMID: 40359883 DOI: 10.1016/j.intimp.2025.114836] [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: 02/10/2025] [Revised: 04/10/2025] [Accepted: 05/07/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Mitochondrial dysfunction is a well-recognized pathological feature of Duchenne Muscular Dystrophy (DMD). The potential regulatory role of mitochondria-related genes (MRGs) in DMD remains to be further explored. METHODS GEO datasets and MRGs were used to analysis mitochondrial scores and evaluate patients' immunological characteristics. Weighted gene co-expression network analysis, differentially expressed genes (DEGs) and MRGs were used to identify hub genes. A specific hub gene was selected, and the effects of this gene overexpression on a horse serum (HS) treated C2C12 cell in vitro model were investigated. RESULTS Mitochondrial score was decreased in DMD group. Significant differences were observed in 12 immune cell types in normal/DMD and high/low mitochondrial score groups. 9 hub genes were identified, with 7 validated. Among them, VDAC1 was selected for further study. Overexpression of VDAC1 in HS C2C12 myoblasts promoted cell proliferation, reduced apoptosis rate and the Bax expression (with concurrent Bcl2 upregulation), diminished LDH release to reduce cytotoxicity, decreased intracellular ROS levels to alleviate oxidative stress, inhibited the expression of autophagy (LC3) and atrophy (Atrogin-1 and MuRF-1) markers, and promoted differentiation. CONCLUSION In conclusion, VDAC1 may participate in the myoblast proliferation and myotube atrophy by influencing mitochondrial function, which may serve as a new target for DMD treatment.
Collapse
Affiliation(s)
- Wenjuan Wu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; Department of Neurology, Hebei Children's Hospital, The Key Laboratory of Pediatric Epilepsy and Neurology of Hebei Province, Shijiazhuang 050031, China
| | - Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; The Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang 050000, China; Neurological Laboratory of Hebei Province, Shijiazhuang 050000, China.
| | - Baoguang Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; Department of Neurology, Hebei Children's Hospital, The Key Laboratory of Pediatric Epilepsy and Neurology of Hebei Province, Shijiazhuang 050031, China
| |
Collapse
|
6
|
Zhou N, Ma L, Shi W, Reiter RJ, Lin J, Zhang Y, Hu D, Ren J, Xu K. Akt mitigates ER stress-instigated cardiac dysfunction via regulation of ferroptosis and mitochondrial integrity in a DHODH-dependent manner. Life Sci 2025; 371:123591. [PMID: 40164331 DOI: 10.1016/j.lfs.2025.123591] [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: 01/08/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
ER stress evokes various types of cell death and myocardial dysfunction. This study aimed to discern the involvement of ferroptosis in chronic Akt activation-offered benefit, if any, against ER stress-triggered cardiac remodeling and contractile anomalies. Cardiac-selective expression of active mutant of Akt (AktOE) and wild-type (WT) mice were challenged with the ER stress instigator tunicamycin (1 mg/kg, 48 h) prior to assessment of cardiac morphology and function. Tunicamycin insult prompted cardiac remodeling (interstitial fibrosis), deranged echocardiographic (higher LVESD, dropped ejection fraction and fractional shortening), cardiomyocyte mechanical and intracellular Ca2+ features alongside mitochondrial injury (collapsed mitochondrial membrane potential and ultrastructural change), oxidative stress, compromised Akt-GSK3β signaling, ER stress (upregulated GRP78 and Gadd153), carbonyl formation, apoptosis and ferroptosis (decreased GPX4, SLC7A11). Intriguingly, tunicamycin-evoked anomalies (except GRP78 and Gadd153) were abrogated by Akt activation. Chronic Akt activation negated tunicamycin-induced downregulation of ferric flavin enzyme dihydroorotate dehydrogenase (DHODH), which catalyzes the fourth step of pyrimidine ab initio biosynthesis, and conversion of dihydroorotic acid to orotate. ER stress-induced myocardial anomalies were reversed by the newly identified PI3K activator triptolide, DHODH activator menaquinone-4 and pyrimidine booster coenzyme Q. In vitro experiment revealed that Akt activation- or triptolide-evoked beneficial responses against tunicamycin-induced cardiomyocyte anomalies were cancelled off by DHODH inhibitor BAY2402234 or ferroptosis inducer erastin. These findings support that chronic Akt activation rescues ER stress-evoked myocardial derangements through DHODH-dependent control of ferroptosis and mitochondrial homeostasis.
Collapse
Affiliation(s)
- Na Zhou
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Li Ma
- Heart Center, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Wanting Shi
- Child Healthcare Department, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, China
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, TX 78229, USA
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Dandan Hu
- Child Healthcare Department, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.
| |
Collapse
|
7
|
Dai L, Huang L, Li L, Tang L, Shi Y, Han X. Unraveling the role of HDAC3 as an immunotherapy prognostic biomarker and therapeutic target in advanced non-small cell lung cancer. Respir Res 2025; 26:214. [PMID: 40506719 PMCID: PMC12160119 DOI: 10.1186/s12931-025-03275-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 05/14/2025] [Indexed: 06/16/2025] Open
Abstract
Background This study investigates HDAC3 as a potential immunotherapy biomarker in advanced non-small cell lung cancer (aNSCLC), focusing on its association with treatment response to immune checkpoint inhibitors (ICIs). Methods We employed a multi-phase approach in 78 aNSCLC patients with 138 plasma samples, starting with a discovery phase that identified differential autoantibodies (AAbs) using proteomic analysis in responders and non-responders. In the validation phase, we assessed AAb levels at multiple time points. Additionally, immunohistochemistry and multiple immunofluorescence (n = 21) were used to validate HDAC3 expression in FFPE samples, single-cell RNA sequencing (n = 26) was performed to explore gene expression differences, cell and animal experiments were performed. Results We identified 127 differential AAbs, with five key AAbs (HDAC3, METTL21C, HSPB3, SPACA7, and SPPL2B) consistently linked to prognosis pre- and post-treatment (p < 0.05). A risk score model based on these AAbs effectively predicted progression-free survival. Furthermore, HDAC3 expression correlated with significant pathway enrichments and was associated with higher TGFβ1, PD-L1 infiltration and lower CD8+ T cells infiltration (p < 0.05). HDAC3 knockdown significantly inhibited cell proliferation, impaired colony formation, and induced G0/G1 phase arrest in lung cancer cells. Preclinical models demonstrated that RGFP966, an HDAC3 inhibitor, combined with anti–PD-1 therapy enhanced CD8+ T cell infiltration (p < 0.05). Conclusion Our findings underscore HDAC3’s role as a biomarker for predicting ICI response in aNSCLC and suggest its potential as a therapeutic target, paving the way for future studies on HDAC3-targeted therapies to improve immunotherapy outcomes. Clinical trial number not applicable. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-025-03275-w.
Collapse
Affiliation(s)
- Liyuan Dai
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Liling Huang
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Xiaohong Han
- Clinical Pharmacology Research Center,NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, Peking Union Medical College Hospital,Chinese Academy of Medical Sciences & Peking Union Medical College, No.1, Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
8
|
Thu NQ, Oh JH, Tien NTN, Park SM, Yen NTH, Phat NK, Hung TM, Nguyen HT, Nguyen DN, Yoon S, Kim DH, Long NP. The lipidome landscape of amiodarone toxicity: An in vivo lipid-centric multi-omics study. Toxicol Appl Pharmacol 2025; 499:117341. [PMID: 40216313 DOI: 10.1016/j.taap.2025.117341] [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: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Amiodarone is an effective therapy for arrhythmias, its prolonged management may lead to significant adverse drug reactions. Amiodarone-induced hepatotoxicity is described by phospholipidosis, hepatic steatosis, cholestatic hepatitis, and cirrhosis. However, the systemic and hepatic lipidome disturbances and underlying toxicological mechanisms remain comprehensively elucidated. Untargeted lipidomics were utilized to analyze serum and liver samples from the rats orally administered a daily dose of amiodarone of either 100 or 300 mg/kg for one week. Changes in the expression of hepatic lipid-related genes were also examined utilizing transcriptomics. We found a higher magnitude of lipidome alterations in the 300 mg/kg than those in the 100 mg/kg groups. Treated animals showed elevated abundances of phosphatidylcholines, ether-linked phosphatidylcholines, sphingomyelins, and ceramides, and decreased levels of triacylglycerols, ether-linked triacylglycerols, and fatty acids. We also found 199 lipid-related differentially expressed hepatic genes between the 300 mg/kg group versus controls, implying lipid metabolism and signaling pathways disturbances. Specifically, elevation of serum phosphatidylcholines and ether-linked phosphatidylcholines, as well as hepatic bismonoacylglycerophosphates might be associated with reduced expression of phospholipase genes and elevated expression of glycerophospholipid biosynthesis genes, possibly driving phospholipidosis. Perturbations of sphingolipid metabolism might also be the key events for amiodarone-induced toxicity. Alterations in gene expression levels related to lipid storage and metabolism, mitochondria functions, and energy homeostasis were also found. Collectively, our study characterized the sophisticated perturbations in the lipidome and transcriptome of amiodarone-treated rats and suggested potential mechanisms responsible for amiodarone-induced hepatotoxicity.
Collapse
Affiliation(s)
- Nguyen Quang Thu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Se-Myo Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Ky Phat
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Tran Minh Hung
- School of Medicine, Tan Tao University, Long An 850000, Vietnam
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Duc Ninh Nguyen
- Comparative Pediatrics, Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| |
Collapse
|
9
|
Xavier S, Nguyen V, Khairnar V, Phan A, Yang L, Nelson MS, Shukla RP, Wang J, Li A, Geng H, Lee J, Sadras T, Pham LV, Weisenburger DD, Chan WC, Lang KS, Shouse GP, Danilov AV, Song JY, Parekh S, Müschen M, Ngo VN. CEACAM1 as a mediator of B-cell receptor signaling in mantle cell lymphoma. Nat Commun 2025; 16:4967. [PMID: 40436855 PMCID: PMC12120064 DOI: 10.1038/s41467-025-60208-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 05/19/2025] [Indexed: 06/01/2025] Open
Abstract
B-cell receptor (BCR) signaling plays an important role in the pathogenesis of mantle cell lymphoma (MCL), but the detailed mechanisms are not fully understood. In this study, through a genome-wide loss-of-function screen, we identify carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) as an essential factor in a subset of MCL tumors. Our signal transduction studies reveal that CEACAM1 plays a critical role in BCR activation through involvement in two dynamic processes. First, following BCR engagement, CEACAM1 co-localizes to the membrane microdomains (lipid rafts) by anchoring to the F-actin cytoskeleton through the adaptor protein filamin A. Second, CEACAM1 recruits and increases the abundance of SYK in the BCR complex leading to BCR activation. These activities of CEACAM1 require its cytoplasmic tail and the N-terminal ectodomain. Considering that previous studies have extensively characterized CEACAM1 as an ITIM-bearing inhibitory receptor, our findings regarding its activating role are both surprising and context-dependent, which may have implications for BCR-targeting therapies.
Collapse
MESH Headings
- Lymphoma, Mantle-Cell/metabolism
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/pathology
- Humans
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Signal Transduction
- Cell Adhesion Molecules/metabolism
- Cell Adhesion Molecules/genetics
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Syk Kinase/metabolism
- Membrane Microdomains/metabolism
- Cell Line, Tumor
- Animals
- Filamins/metabolism
- Mice
- Actin Cytoskeleton/metabolism
Collapse
Affiliation(s)
- Serene Xavier
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Vivian Nguyen
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Vishal Khairnar
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - An Phan
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Lu Yang
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA
| | - Michael S Nelson
- Light Microscopy and Digital Imaging Core, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Ravi P Shukla
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jinhui Wang
- Integrative Genomics Core, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Aimin Li
- Department of Pathology, City of Hope Medical Center, Duarte, CA, USA
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Jaewoong Lee
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale University, New Haven, CT, USA
- School of Biosystems and Biomedical Sciences, College of Health Science, Korea University, Seoul, Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Korea
| | - Teresa Sadras
- Olivia Newton-John Cancer Research Institute, Heidelberg, Melbourne, VIC, Australia
| | - Lan V Pham
- Oncology Discovery, Abbvie Inc., South San Francisco, CA, USA
| | - Dennis D Weisenburger
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wing C Chan
- Department of Pathology, City of Hope Medical Center, Duarte, CA, USA
| | - Karl S Lang
- Institute of Immunology, University Hospital Essen, Essen, Germany
| | - Geoffrey P Shouse
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Alexey V Danilov
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Medical Center, Duarte, CA, USA
| | - Joo Y Song
- Department of Pathology, City of Hope Medical Center, Duarte, CA, USA
| | - Samir Parekh
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Markus Müschen
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Vu N Ngo
- Department of Systems Biology, City of Hope Comprehensive Cancer Center, Monrovia, CA, USA.
| |
Collapse
|
10
|
Lopes C, Brandão A, Teixeira MR, Dinis-Ribeiro M, Pereira C. Saliva-derived transcriptomic signature for gastric cancer detection using machine learning and leveraging publicly available datasets. Sci Rep 2025; 15:18491. [PMID: 40425785 PMCID: PMC12116728 DOI: 10.1038/s41598-025-96864-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/01/2025] [Indexed: 05/29/2025] Open
Abstract
Saliva, a non-invasive, self-collected liquid biopsy, holds promise for early gastric cancer (GC) screening. This study aims to assess the potential of saliva as a proxy for malignant gastric transformation and its diagnostic value through transcriptomic profiling. Leveraging transcriptomic data from the Gene Expression Omnibus (GEO), we constructed and validated predictive models through machine learning algorithms within the tidymodels framework. Tissue-based models were validated on independent tissue datasets, and subsequently applied to saliva. Additionally, an independent saliva-derived model was created and evaluated using sensitivity, specificity, accuracy, area under the curve (AUC), and likelihood ratio (LR) metrics. Tissue-derived models demonstrated excellent performance, with AUC values exceeding 0.9, but did not translate effectively to saliva, suggesting distinct molecular landscapes between tissue and saliva in GC. The saliva-specific model using support vector machine (SVM) achieved the highest performance, with an AUC of 0.87 (95% CI 0.72-0.97), a sensitivity of 0.79 (95% CI 0.58-0.95) and a specificity of 0.70 (95% CI 0.40-0.90). While saliva may not mirror tissue gene expression profile, it represents a promising non-invasive predictive tool for the early detection of GC. Further research is warranted to optimize saliva-derived molecular signatures, increasing their sensitivity and specificity for early cancer detection and advance the use of liquid biopsies in personalized medicine for improved screening, diagnostic and prognostic capabilities.
Collapse
Affiliation(s)
- Catarina Lopes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Center for Health Technology and Services Research (CINTESIS@RISE), University of Porto, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
| | - Andreia Brandão
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
| | - Manuel R Teixeira
- ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal
- Cancer Genetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Laboratory Genetics, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal
- Department of Gastroenterology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Carina Pereira
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), Porto, Portugal.
| |
Collapse
|
11
|
Delgado-Chaves FM, Spurny F, Laske T, Oubounyt M, Baumbach J. DRaCOon: a novel algorithm for pathway-level differential co-expression analysis in transcriptomics. BMC Bioinformatics 2025; 26:137. [PMID: 40419963 DOI: 10.1186/s12859-025-06162-9] [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: 11/21/2024] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Understanding the molecular mechanisms underlying diseases is crucial for more precise, personalized medicine. Pathway-level differential co-expression analysis, a powerful approach for transcriptomics, identifies condition-specific changes in gene-gene interaction networks, offering targeted insights. However, a key challenge is the lack of robust methods and benchmarks specifically for evaluating algorithms' ability to identify disrupted gene-gene associations across conditions. We introduce DRaCOoN (Differential Regulatory and Co-expression Networks), a Python package and web tool for pathway-level differential co-expression analysis. DRaCOoN uniquely integrates multiple association and differential metrics, with a novel, computationally efficient permutation test for significance assessment. Crucially, DRaCOoN also provides a benchmarking framework for comprehensive method evaluation. Extensive benchmarking on simulated data and three real-world datasets (bone healing, colorectal cancer, and head/neck carcinoma) showed that DRaCOoN, particularly with an entropy-based association measure and the s differential metric, consistently outperforms eight other methods. It remains highly accurate in balanced datasets, robust to varying gene perturbation levels, and identifies biologically relevant regulatory changes. Furthermore, DRaCOoN serves as both a powerful tool and a benchmarking framework for elucidating disease mechanisms from transcriptomics data, advancing precision medicine by uncovering critical gene regulatory alterations.
Collapse
Affiliation(s)
- Fernando M Delgado-Chaves
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22607, Hamburg, Hamburg, Germany.
| | - Ferdinand Spurny
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22607, Hamburg, Hamburg, Germany
| | - Tanja Laske
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22607, Hamburg, Hamburg, Germany
- Viral Systems Modeling, Leibniz Institute of Virology, Martinistraße 52, 20251, Hamburg, Hamburg, Germany
| | - Mhaned Oubounyt
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22607, Hamburg, Hamburg, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22607, Hamburg, Hamburg, Germany
| |
Collapse
|
12
|
Yang X, Chen Q. Integrative Transcriptomic Profiling Identifies TNF and IL1B as Candidate Key Early-Response Genes in Macrophages Infected with Smooth Brucella Using a Comprehensive Bioinformatic Approach. BIOLOGY 2025; 14:579. [PMID: 40427768 PMCID: PMC12109160 DOI: 10.3390/biology14050579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/01/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Smooth Brucella are the main pathogenic bacteria that threaten human health and food safety. The early stage of smooth Brucella and macrophage interaction is an important phase, and smooth Brucella species elicit a dramatic transcriptional response in infected macrophages. However, the key transcriptional events are still obscure. This study aimed to identify key candidate response pathways and genes in macrophages infected with smooth Brucella at the early interaction stage. Three gene expression profiles including GSE21117, GSE5202, and GSE8385 were retrieved from the NCBI GEO database, and were integrated using comprehensive bioinformatics methods including gene set enrichment analysis, differentially expressed gene analysis, protein and protein interaction (PPI) network construction, and transcription factor prediction. The results showed that 16 up-regulated and 22 down-regulated pathways were identified, including six up-regulated immune-related pathways. A total of 41 up-regulated and four down-regulated genes were identified, and a PPI network including 31 nodes and 134 edges was constructed based on the interactive information of 45 dysregulated genes. A highly correlated module comprising 19 nodes and 103 edges was identified based on the topological features of the whole PPI network. Seven centrality analyses revealed that Tnf and Il1b were essential genes in the highly correlated module, and that the two essential genes were simultaneously enriched in eight significantly up-regulated pathways (including two immune-related pathways). Bcl3 was predicted as a transcription factor in the highly correlated module, and may play regulatory roles in the transcription of Tnf and Il1b genes. The present study identified Tnf and IL1b as candidate key response genes in infected macrophages at the early stage of smooth Brucella and macrophage interaction, which contributes to a deeper understanding of the early key transcriptional events in macrophages infected with smooth Brucella species.
Collapse
Affiliation(s)
| | - Qiang Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| |
Collapse
|
13
|
Swenson H, Ittner E, Werner L, Rönnerman EW, Mateoiu C, Kovács A, Dahm-Kähler P, Saed GM, Nemes S, Karlsson P, Parris TZ, Helou K. Integrative analysis of epigenetic and transcriptional interrelations identifies histotype-specific biomarkers in early-stage ovarian carcinoma. J Ovarian Res 2025; 18:103. [PMID: 40390000 PMCID: PMC12087105 DOI: 10.1186/s13048-025-01676-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a deadly and heterogenous disease comprising five major histotypes: clear cell carcinoma (CCC), endometrioid carcinoma (EC), low- and high-grade serous carcinoma (LGSC, HGSC), and mucinous carcinoma (MC). Despite this heterogeneity, EOC is often treated as a homogenous disease, and reliable screening tests are lacking. Although progress has been made, there is a pressing need for biomarkers to refine patient stratification, guide treatment, and improve outcomes. Here, we elucidated the relationship between DNA methylation and gene expression patterns in EOC to identify histotype-specific biomarkers. METHODS Differential DNA methylation and gene expression analyses were performed for 86 early-stage EOC samples after histopathological reclassification stratified by histotype. The correlation between DNA methylation and gene expression was examined, and histotype-specific biomarkers were identified. Hierarchical clustering and predictive machine learning modeling were employed to assess the performance of the histotype-specific biomarkers using four external cohorts. RESULTS EOC histotypes exhibited distinct epigenetic, transcriptional, and functional profiles, with candidate histotype-specific biomarkers such as CTSE and VCAN effectively distinguishing CCC, HGSC, and MC on the transcriptional level. Gene expression for the candidate biomarkers was found to be reproducible across external cohorts, with histotype-specific differences remaining homogenous. CONCLUSIONS This study identified promising histotype-specific biomarkers for EOC using integrative transcriptomic and epigenomic analysis. Furthermore, these findings indicate that additional stratification or potential reclassification of the EC histotype is warranted in future studies.
Collapse
Affiliation(s)
- Hugo Swenson
- Department of Oncology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden.
| | - Ella Ittner
- Department of Oncology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Lucas Werner
- Department of Oncology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Werner Rönnerman
- Department of Clinical Pathology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Claudia Mateoiu
- Department of Clinical Pathology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pernilla Dahm-Kähler
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Ghassan M Saed
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Per Karlsson
- Department of Oncology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Sahlgrenska Academy, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
14
|
Pius-Sadowska E, Kulig P, Niedźwiedź A, Baumert B, Rogińska D, Łuczkowska K, Sobuś A, Parczewski M, Kawa M, Paczkowska E, Machaliński B. The micro-RNA expression profile predicts the severity of SARS-CoV-2 infection. Sci Rep 2025; 15:17139. [PMID: 40382351 DOI: 10.1038/s41598-025-01229-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Although much is known about the pathophysiology of severe COVID-19, there are still areas that remain to be determined. It is well established that the pivotal molecular event is a hyperinflammatory response also referred to as a cytokine storm. The aim of this retrospective cohort study was to determine miRNAs which might be predictive for the admission to the intensive care unit (ICU). We analyzed blood samples from 210 COVID-19 patients and the control group consisted of 80 healthy individuals. Results revealed the miRNA expression pattern has the potential to predict the severity of COVID-19, reflecting the clinical symptoms of the infection, such as the need for oxygen therapy and concomitant pneumonia. In particular, low expression of miRNAs miR106a-5p, miR17-5p, miR181a-5p, miR191-5p, miR20a-5p and miR451a, especially in the initial phase of the disease, is associated with an unfavorable clinical course of SARS-CoV-2 infection (admission to the ICU).
Collapse
Affiliation(s)
- Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Piotr Kulig
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Niedźwiedź
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bartłomiej Baumert
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Sobuś
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, Arkońska 4 Street, 71-455, Szczecin, Poland
| | - Miłosz Kawa
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland.
| |
Collapse
|
15
|
Jopek K, Tyczewska M, Blatkiewicz M, Olechnowicz A, Szyszka M, Stelcer E, Ciesiółka S, Jopek M, Malendowicz LK, Ruciński M. Profile of Rat Adrenal microRNAs Induced by Gonadectomy and Testosterone or Estradiol Replacement. Int J Mol Sci 2025; 26:4543. [PMID: 40429687 PMCID: PMC12111343 DOI: 10.3390/ijms26104543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Sex-related differences in the structure and function of the adrenal cortex in mature rats are well recognized, largely driven by the action of sex hormones on the hypothalamic-pituitary-adrenal axis (HPA). By replacing testosterone or estradiol in gonadectomized rats, we aimed to elucidate the regulation of micro RNA (miRNA) profiles by sex hormones and their role in physiological adrenal function, providing new insights into gene expression modulation in the adrenal gland. This paper focuses on the description of miRNA profiles using the microarray technique. In our study, we observed significant sex differences in miRNA and mRNA expression levels. These differences are as follows: miRNA expression profiles Male C vs. Female C-0 down, 25 up-regulated, while mRNA profiles were 43 down and 27 up-regulated. Moreover, we observed the most significant differences in miRNA profiles between orchiectomized male rats supplemented with testosterone (ORX + T) and ovariectomized female rats treated with estradiol (OVX + E). Furthermore, we described changes in target gene expression and biological processes regulated by miRNAs. The processes most differentially expressed between the ORX + T and OVX + E groups are those related to the metabolism and synthesis of sterol compounds, the positive and negative regulation of metabolic processes in cells, e.g., cholesterol metabolism, response to various external factors, e.g., hormones, regulation of processes related to cell motility. We also identified several miRNAs, such as miR-370, miR-377, and miR-503, that exhibited interesting changes in their expression after testosterone or estradiol replacement. These results contribute to a deeper understanding of adrenal physiology.
Collapse
Affiliation(s)
- Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Marianna Tyczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
- Department of Anatomy and Histology, University of Zielona Gora, Licealna Street 9, 65-417 Zielona Gora, Poland
| | - Małgorzata Blatkiewicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Anna Olechnowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Marta Szyszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Ewelina Stelcer
- Department of Biochemistry and Biotechnology, Poznan University of Life Sciences, 60-637 Poznan, Poland;
| | - Sylwia Ciesiółka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Maria Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Ludwik K. Malendowicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| | - Marcin Ruciński
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (K.J.); (M.T.); (M.B.); (A.O.); (M.S.); (S.C.); (M.J.); (M.R.)
| |
Collapse
|
16
|
Zhang C, Li S, Guo J, Pan T, Zhang Y, Gao Y, Pan J, Liu M, Yang Q, Yu J, Xu J, Li Y, Li X. Multi-dimensional characterization of cellular states reveals clinically relevant immunological subtypes and therapeutic vulnerabilities in ovarian cancer. J Transl Med 2025; 23:519. [PMID: 40340848 PMCID: PMC12063340 DOI: 10.1186/s12967-025-06521-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/22/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Diverse cell types and cellular states in the tumor microenvironment (TME) are drivers of biological and therapeutic heterogeneity in ovarian cancer (OV). Characterization of the diverse malignant and immunology cellular states that make up the TME and their associations with clinical outcomes are critical for cancer therapy. However, we are still lack of knowledge about the cellular states and their clinical relevance in OV. METHODS We manually collected the comprehensive transcriptomes of OV samples and characterized the cellular states and ecotypes based on a machine-learning framework. The robustness of the cellular states was validated in independent cohorts and single-cell transcriptomes. The functions and regulators of cellular states were investigated. Meanwhile, we thoroughly examined the associations between cellular states and various clinical factors, including clinical prognosis and drug responses. RESULTS We depicted and characterized an immunophenotypic landscape of 3,099 OV samples and 80,044 cells based on a machine learning framework. We identified and validated 32 distinct transcriptionally defined cellular states from 12 cell types and three cellular communities or ecotypes, extending the current immunological subtypes in OV. Functional enrichment and upstream transcriptional regulator analyses revealed cancer hallmark-related pathways and potential immunological biomarkers. We further investigated the spatial patterns of identified cellular states by integrating the spatially resolved transcriptomes. Moreover, prognostic landscape and drug sensitivity analysis exhibited clinically relevant immunological subtypes and therapeutic vulnerabilities. CONCLUSION Our comprehensive analysis of TME helps leveraging various immunological subtypes to highlight new directions and targets for the treatment of cancer.
Collapse
Affiliation(s)
- Can Zhang
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Si Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Jiyu Guo
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Tao Pan
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Ya Zhang
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Yueying Gao
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Jiwei Pan
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Meng Liu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Qingyi Yang
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China
| | - Jinyang Yu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| | - Yongsheng Li
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150081, China.
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150040, Heilongjiang, China.
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xia Li
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou, 571199, China.
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
17
|
Salem F, ElGamal A, Zhang Z, Kong W. Integrative multi-transcriptomic analysis uncovers core genes and potential defense mechanisms in rice-Magnoporthe oryzae interaction. PLANT CELL REPORTS 2025; 44:114. [PMID: 40332586 DOI: 10.1007/s00299-025-03490-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025]
Abstract
KEY MESSAGE Multiple transcriptomic comprehensive analyses highlight key genes and cast new light on multifaceted pathways that may be important arenas in rice innate immunity against Magnoporthe oryzae blast disease. Magnaporthe oryzae (MOR) poses a significant threat to rice production worldwide. However, defense mechanisms in rice against MOR remain inadequately defined. In this study, a multi-transcriptomic integrative analysis on 441 samples from diverse microarrays and RNA-seq sets was conducted to reveal critical factors in rice defense against MOR infection. A robust pattern of 3534 upregulated genes and 2920 repressed genes was commonly identified across all MOR-infected arrays and RNA-seq profiles. Interestingly, enrichment analysis revealed a consistent triggering of endoplasmic reticulum (ER)-related mechanisms and citric acid cycle (TCA) influx in rice response to MOR infection across all the transcriptome profiles, suggesting their critical role in modulating rice immunity against the pathogen. By contrast, chloroplast and photosynthesis pathways were frequently repressed across all the profiles. Among ER-related mechanisms, the phagosome pathway involved in the activation of NADPH oxidase was highly triggered in early response to MOR infection. Moreover, WGCNA analysis highlighted four key co-expressed gene modules and 80 significant hub genes associated with MOR infection. Among the core genes, Sec61 gene involved in the ER-translocation process was identified along with OsMFP (peroxisomal oxidation gene) and OSAHH gene (involved in cyclic-trans-methylation). Furthermore, MPK6, WRKY24, NUP35, and NPR1 genes were observed as core co-expressed genes, suggesting their significance in regulating rice immunity against MOR. Our findings elucidate key genes and multifaceted mechanisms in rice-MOR interaction, proposing new informative clues that can be exploited to improve rice resistance against blast disease.
Collapse
Affiliation(s)
- Fatma Salem
- College of Plant Protection, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
- Faculty of Agriculture, Fayoum University, Fayoum, 63514, Egypt.
| | - Ahmed ElGamal
- Virus and Phytoplasma Research Department, Plant Pathology Research Institute, Agricultural Research Center, Giza, 12619, Egypt
| | - Zujian Zhang
- College of Agriculture, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| | - Weiwen Kong
- College of Plant Protection, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
18
|
Erady C, Bethlehem R, Bullmore E, Lynall ME. Systematic review and mega-analysis of the peripheral blood transcriptome in depression implicates dysregulation of lymphoid cells and histones. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.01.25326802. [PMID: 40385445 PMCID: PMC12083620 DOI: 10.1101/2025.05.01.25326802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Background Depression has been associated with transcriptomic changes in peripheral blood. However, the contribution of specific immune cell subsets or pathways remains unclear, and findings have been variable across previous studies, which have not tended to account for sample cellular composition. Methods We performed a systematic review of peripheral blood transcriptome studies in depression. For the five datasets meeting criteria (total N=6,011), we performed harmonized reprocessing and cell-composition-adjusted differential gene and transcript analyses, followed by a bias- and inflation-adjusted weighted Z-score mega-analysis. We investigated the biological pathways and cell subsets implicated by the results. We also performed a sex-stratified gene network mega-analysis using consensus weighted gene co-expression network analysis (WGCNA). Results Few genes showed robust differential gene expression (DGE) in depression. Depression was reproducibly associated with decreases in replication-dependent histones, and with a decrease in oxidative phosphorylation pathways in females only. Cell source analyses implicated lymphoid cells (T cells and NK cells) as likely contributors to the depression differential expression signature. WGCNA mega-analysis revealed multiple consensus modules associated with depression, with a PUF60-related module upregulated in both female and male depression in sex-stratified analyses. Two genes predicted to be causally relevant to depression by transcriptome-wide association studies (GPX4 and GYPE) showed significant DGE. Conclusions These results are convergent with immunogenetic evidence implicating lymphoid cell dysregulation in depression, while also highlighting histone alterations as a key molecular signature in depression. They also indicate the importance of large-scale datasets for biomarker discovery in the context of heterogeneous disorders like depression.
Collapse
Affiliation(s)
| | | | - Ed Bullmore
- Department of Psychiatry, University of Cambridge, UK
| | - Mary-Ellen Lynall
- Department of Psychiatry, University of Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, UK
| |
Collapse
|
19
|
Zhang Y, Liu H, Zhang P, Ye B, Ying H, Yang H, Zhang J, Zhang N, Li K, Wei T, Jiang A, Lin A, Luo P. THER: Integrative Web Tool for Tumour Hypoxia Exploration and Research. Cell Prolif 2025:e70053. [PMID: 40312874 DOI: 10.1111/cpr.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/20/2025] [Accepted: 04/16/2025] [Indexed: 05/03/2025] Open
Affiliation(s)
- Yasi Zhang
- Donghai County People's Hospital, Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huiying Liu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - BiCheng Ye
- Liver Disease Center of Integrated Traditional Chinese and Western Medicine, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Nanjing, China
| | - Haoxuan Ying
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hong Yang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Hongshan, China
| | - Kailai Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University, Second Military Medical University, Shanghai, China
| | - Anqi Lin
- Donghai County People's Hospital, Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Donghai County People's Hospital, Jiangnan University Smart Healthcare Joint Laboratory, Donghai County People's Hospital (Affiliated Kangda College of Nanjing Medical University), Lianyungang, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Duan L, Yin H, Liu J, Wang W, Huang P, Liu L, Shen J, Wang Z. Maternal COVID-19 infection associated with offspring neurodevelopmental disorders. Mol Psychiatry 2025; 30:2108-2118. [PMID: 39521839 DOI: 10.1038/s41380-024-02822-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/20/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Maternal COVID-19 infection increases the incidence of neurodevelopmental disorders (NDDs) in offspring, although the underlying mechanisms have not been elucidated. This study demonstrated that COVID-19 infection during pregnancy disrupted the balance of maternal and fetal immune environments, driving alterations in astrocytes, endothelial cells, and excitatory neurons. A risk score was established using 47 unique genes in the single-cell transcriptome of gestational mothers. The high risk score in CD4 proliferating T cell level served as an indicator for increased risk of offspring NDDs. Summary-based Mendelian randomization and phenome-wide association study analyses were conducted to identify the causal association of the transcriptional changes with the increased risk of offspring NDDs. Additionally, 10 drugs were identified as potential therapeutic candidates. Our findings support a model where the maternal COVID-19 infection changed the levels of CD4 proliferating T cells, leading to the alterations of astrocytes, endothelial cells, and excitatory neurons in offspring, contributing to the increased risk of NDDs in these individuals.
Collapse
Affiliation(s)
- Lian Duan
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Huamin Yin
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxin Liu
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Wenhang Wang
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Peijun Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Li Liu
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China
| | - Jingling Shen
- Institute of Life Sciences, College of Life and Environmental Sciences, Wenzhou University, Wenzhou, 325035, China.
| | - Zhendong Wang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
21
|
Arbet J, Yamaguchi TN, Shiah YJ, Hugh-White R, Wiggins A, Oh J, Gebo T, Foucal A, Lesurf R, Jung CH, Dang RMA, Agrawal R, Livingstone J, Salcedo A, Yao CQ, Espiritu SMG, Houlahan KE, Yousif F, Heisler LE, Papenfuss AT, Fraser M, Pope B, Kishan A, Berlin A, Chua MLK, Corcoran NM, van der Kwast T, Hovens CM, Bristow RG, Boutros PC. The Landscape of Prostate Tumour Methylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637178. [PMID: 39990314 PMCID: PMC11844408 DOI: 10.1101/2025.02.07.637178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Prostate cancer is characterized by profound clinical and molecular heterogeneity. While its genomic heterogeneity is well-characterized, its epigenomic heterogeneity remains less understood. We therefore created a compendium of 3,001 multi-ancestry prostate methylomes spanning normal tissue through localized disease of all grades to poly-metastatic disease. A subset of 884 samples had multi-omic DNA and/or RNA characterization. We identify four epigenomic subtypes that risk-stratify patients and reflect distinct evolutionary trajectories. We demonstrate extensive regulatory interplay between DNA ploidy and DNA methylation, with transcriptional consequences that vary across genes and disease stages. We define the epigenetic dysregulation signatures of the 15 most important clinico-molecular features, creating predictive models for each. For example, we identify specific epigenetic features that predict patient outcome and that are synergistic with clinico-genomic prognostic features. These results define a complex interplay between tumour genetics and epigenetics that converges to modify gene-expression programs and clinical presentation.
Collapse
|
22
|
Elghaish RA, Attallah NE, Khaled H, Mekawy AS, Elserafy M, Badr E. A computational framework for identifying cytoskeletal genes associated with age-related diseases. Sci Rep 2025; 15:14590. [PMID: 40287491 PMCID: PMC12033331 DOI: 10.1038/s41598-025-97363-y] [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: 06/05/2024] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
The cytoskeleton comprises polymers from protein filaments shaped in an organized structure. This structure contributes significantly to the cell's function and viability. Decades of research have implicated that the cytoskeleton's dynamic nature is associated with downstream signaling events that further regulate cellular activity and control aging and neurodegeneration. This study aims to investigate the transcriptional changes of the cytoskeletal genes and their regulators in five age-related diseases: Hypertrophic Cardiomyopathy (HCM), Coronary Artery Disease (CAD), Alzheimer's disease (AD), Idiopathic Dilated Cardiomyopathy (IDCM), and Type 2 Diabetes Mellitus (T2DM). An integrative approach of machine learning-based models and differential expression analysis was employed to identify potential biomarkers based on the cytoskeletal genes. Multiple machine-learning algorithms were used, where the Support Vector machines (SVM) classifier achieved the highest accuracy. The study highlighted 17 genes involved in the cytoskeleton's structure and regulation associated with age-related diseases. The results provide a holistic overview of the role of transcriptionally dysregulated cytoskeletal genes in age-related diseases. This study pinpoints cytoskeletal genes and regulators of the cytoskeleton that can be utilized as potential markers and drug targets.
Collapse
Affiliation(s)
- Reem A Elghaish
- University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Nayera E Attallah
- University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Hesham Khaled
- University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Asmaa S Mekawy
- University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Menattallah Elserafy
- University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, 12578, Egypt.
| | - Eman Badr
- University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
- Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
23
|
Shahriari AG, Tahmasebi A, Ghodoum Parizipour MH, Soltani Z, Tahmasebi A, Shahid MS. The crucial role of mitochondrial/chloroplast-related genes in viral genome replication and host defense: integrative systems biology analysis in plant-virus interaction. Front Microbiol 2025; 16:1551123. [PMID: 40336839 PMCID: PMC12055828 DOI: 10.3389/fmicb.2025.1551123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025] Open
Abstract
Plant viruses participate as biotrophic parasites in complex interactions with their hosts, resulting in the regulation of a diverse range of chloroplast/mitochondria-related genes that are essential for mediating immune responses. In this study, integrative systems biology approaches were applied to identify chloroplast/mitochondrial genes during viral infections caused by a wide number of viruses in Arabidopsis thaliana, tobacco (Nicotiana tabacum L.), and rice (Oryza sativa L.). These findings indicated that 1.5% of the DEGs were common between Arabidopsis/tobacco and Arabidopsis/rice, whereas 0.1% of the DEGs were shared among all species. Approximately 90% of common DEGs are uniquely associated with chloroplasts and mitochondria in the host defense against viral infection and replication. The functions of WRKY, NAC, and MYB transcription factors in imparting resistance to viral infections can be established. Promoter analysis revealed that AP2/EREBP, DOF, and C2H2 zinc finger factors included the most frequent binding sites and played a more important role in plant-viral interactions. Comparative analysis revealed several miRNAs with defensive functions including miRNA156, miRNA160, and miRNA169. The PPI network revealed several key hub genes mostly related to chloroplasts/mitochondria, including ZAT6, CML37, CHLI, DREB, F27B13.20, and ASP2 with upregulation, also PLGG1, PSBY, APO2, POR, ERF, and CSP with downregulation. Moreover, novel hub genes with unknown functions, such as AT2G41640 and AT3G57380 have been identified. This study represents the first preliminary systems biology approach to elucidate the roles of chloroplast/mitochondria-related genes in Arabidopsis, tobacco, and rice against viral challenges by introducing valuable candidate genes for enhanced genetic engineering programs to develop virus-resistant crop varieties.
Collapse
Affiliation(s)
- Amir Ghaffar Shahriari
- Department of Agriculture and Natural Resources, Higher Education Center of Eghlid, Eghlid, Iran
| | - Aminallah Tahmasebi
- Department of Agriculture, Minab Higher Education Center, University of Hormozgan, Bandar Abbas, Iran
| | - Mohamad Hamed Ghodoum Parizipour
- Department of Plant Protection, Faculty of Agriculture, Agricultural Sciences and Natural Resources University of Khuzestan, Mollasani, Iran
| | - Zahra Soltani
- Institute of Biotechnology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Ahmad Tahmasebi
- Institute of Biotechnology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Muhammad Shafiq Shahid
- Department of Plant Sciences, College of Agricultural and Marine Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
24
|
Zhou Y, Li X, Wang Z, Ng L, He R, Liu C, Liu G, Fan X, Mu X, Zhou Y. Machine learning-driven prediction model for cuproptosis-related genes in spinal cord injury: construction and experimental validation. Front Neurol 2025; 16:1525416. [PMID: 40337173 PMCID: PMC12057486 DOI: 10.3389/fneur.2025.1525416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Spinal cord injury (SCI) severely affects the central nervous system. Copper homeostasis is closely related to mitochondrial regulation, and cuproptosis is a novel form of cell death associated with mitochondrial metabolism. This study aimed to explore the relationship between SCI and cuproptosis and construct prediction models. Methods Gene expression data of SCI patient samples from the GSE151371 dataset were analyzed. The differential expression and correlation of 13 cuproptosis-related genes (CRGs) between SCI and non-SCI samples were identified, and the ssGSEA algorithm was used for immunological infiltration analysis. Unsupervised clustering was performed based on differentially expressed CRGs, followed by weighted gene co-expression network analysis (WGCNA) and enrichment analysis. Three machine learning models (RF, LASSO, and SVM) were constructed to screen candidate genes, and a Nomogram model was used for verification. Animal experiments were carried out on an SCI rat model, including behavioral scoring, histological staining, electron microscopic observation, and qRT-PCR. Results Seven CRGs showed differential expression between SCI and non-SCI samples, and there were significant differences in immune cell infiltration levels. Unsupervised clustering divided 38 SCI samples into two clusters (Cluster C1 and Cluster C2). WGCNA identified key modules related to the clusters, and enrichment analysis showed involvement in pathways such as the Ribosome and HIF-1 signaling pathway. Four candidate genes (SLC31A1, DBT, DLST, LIAS) were obtained from the machine learning models, with SLC31A1 performing best (AUC = 0.958). Animal experiments confirmed a significant decrease in the behavioral scores of rats in the SCI group, pathological changes in tissue sections, and differential expression of candidate genes in the SCI rat model. Discussion This study revealed a close association between SCI and cuproptosis. Abnormal expression of the four candidate genes affects mitochondrial function, energy metabolism, oxidative stress, and the immune response, which is detrimental to the recovery of neurological function in SCI. However, this study has some limitations, such as unidentified SRGs, a small sample size. Future research requires more in vitro and in vivo experiments to deeply explore regulatory mechanisms and develop intervention methods.
Collapse
Affiliation(s)
- Yimin Zhou
- Department of Orthopedics, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xin Li
- Postdoctoral Research Workstation, Orthopedic Hospital, Chonqqing University of Chinese Medicine, Chongqing, China
| | - Zixiu Wang
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Liqi Ng
- Institute of Orthopaedics and Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| | - Rong He
- College of Integrated Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chaozong Liu
- Institute of Orthopaedics and Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, London, United Kingdom
| | - Gang Liu
- Department of Orthopedics, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Fan
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Xiaohong Mu
- Department of Orthopedics, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Yu Zhou
- Postdoctoral Research Workstation, Orthopedic Hospital, Chonqqing University of Chinese Medicine, Chongqing, China
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Wang Y, Wei A, Su Z, Shi Y, Li X, He L. Characterization of lactylation-based phenotypes and molecular biomarkers in sepsis-associated acute respiratory distress syndrome. Sci Rep 2025; 15:13831. [PMID: 40263316 PMCID: PMC12015483 DOI: 10.1038/s41598-025-96969-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/01/2025] [Indexed: 04/24/2025] Open
Abstract
Sepsis-associated acute respiratory distress syndrome (ARDS) is a heterogeneous disease with high morbidity and mortality. Lactylation plays a crucial role in sepsis and sepsis-induced lung injury. This study aimed to identify distinct lactylation-based phenotypes in patients with sepsis-associated ARDS and determine relevant molecular biomarkers. We analyzed blood transcriptome and clinical data from patients with sepsis-associated ARDS and calculated the lactylation activity. KEGG pathway analysis, drug sensitivity prediction, and immune cell infiltration analysis were performed. Candidate molecular biomarkers were identified by intersecting the feature genes extracted from four machine learning models. Lactylation activity showed significant heterogeneity among patients with sepsis-associated ARDS, which enabled the classification into low- and high-lactylation activity phenotypes. Patients with high-lactylation experienced longer hospital stays and higher mortality rates, as well as distinct signaling pathways, drug responses, and circulating immune cell abundances. Six key markers (ALDOB, CCT5, EP300, PFKP, PPIA, and SIRT1) were identified to differentiate the two lactylation activity phenotypes, all significantly correlated with circulating immune cell populations. This study revealed significant heterogeneity in lactylation activity phenotypes among patients with sepsis-associated ARDS and identified potential biomarkers to facilitate the application of these phenotypes in clinical practice.
Collapse
Affiliation(s)
- Yiheng Wang
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China.
| | - An Wei
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Zixuan Su
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Yunyi Shi
- Department of Ophthalmology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China
| | - Xinqiu Li
- Department of Anesthesiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, 69 Chuanshan Road, Shigu District, Hengyang, 421001, Hunan Province, China
| | - Lixian He
- Department of Ophthalmology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, China.
| |
Collapse
|
26
|
Li J, Yang Y, Cui Z. Identification of shared important genes associated with ferroptosis across different etiologies of acute lung injury. Sci Rep 2025; 15:13561. [PMID: 40253492 PMCID: PMC12009320 DOI: 10.1038/s41598-025-98936-7] [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: 11/03/2024] [Accepted: 04/15/2025] [Indexed: 04/21/2025] Open
Abstract
Acute lung injury (ALI) of different etiologies has shared pathophysiologic process, from which we speculated that ALI of different etiologies may share common molecular features. While the shared genetic characteristics of ALI remain unclear. In this paper, we aimed to identify shared ferroptosis-associated and bottleneck genes from acute lung injury of different etiologies. Firstly, we extracted five groups of gene sets related to three distinct models of ALI from the Gene Expression Omnibus (GEO) database. Then, through the utilization of weighted gene co-expression network analysis (WGCNA), we identified 3 significant gene modules and ascertained 7 shared co-expressed genes affected by these models. Subsequently, through the utilization of differential gene expression analysis and protein-protein interaction network analysis for the 3 gene modules, the shared bottleneck gene Slc7a11 was identified. Moreover, the 7 shared co-expressed genes subjected to these three ALI models were used to identify shared ferroptosis-associated genes via the FerrDb database. Finally, the key gene Slc7a11 was confirmed and validated. In addition, we observed that Slc7a11 is both a driver and a suppressor gene in the FerrDb database. Interestingly, we found the expression level of Slc7a11 was significantly upregulated in the three ALI models. Experimentally, we confirmed the expression of Slc7a11 in rat ALI tissues by using immunofluorescence staining and real-time polymerase chain reaction (qRT-PCR) assays. Collectively, our findings complement the exploration of the shared pathogenesis of ALI. There are genetic features shared by ALI of different etiology and the increased expression of Slc7a11 was identified in the three different etiologies of ALI, which can improve our understanding of the shared molecular mechanisms underlying ALI.
Collapse
Affiliation(s)
- Jing Li
- Department of Burns and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Emergency Surgery, Xinxiang Central Hospital, Xinxiang, 453000, China
| | - Yanming Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhengjun Cui
- Department of Burns and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
27
|
Nocito MC, Hantel C, Lerario AM, Mastrorocco F, De Martino L, Musicco C, Perrotta ID, Scalise M, Indiveri C, Giannattasio S, Val P, Lanzino M, Pezzi V, Casaburi I, Sirianni R. A targetable antioxidant defense mechanism to EZH2 inhibitors enhances tumor cell vulnerability to ferroptosis. Cell Death Dis 2025; 16:291. [PMID: 40229247 PMCID: PMC11997205 DOI: 10.1038/s41419-025-07607-y] [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: 07/05/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
Epigenetic changes are present in all human cancers and are responsible for switching on or off genes, thus controlling tumor cell transcriptome. These changes occur through DNA methylation, histone modifiers and readers, chromatin remodelers, and microRNAs. The histone H3 methyl-transferase EZH2 gene is overexpressed in several cancer types, including adrenocortical carcinoma (ACC), a rare cancer still lacking a targeted therapy. EZH2 inhibitors (EZH2i) have been tested in several clinical trials, but their effectiveness was limited by the toxic effects of the therapeutic doses. We tested several EZH2i on ACC cells, and observed a significant reduction in cell growth only with doses much higher than those required to prevent H3 methylation. We found that all tested EZH2i doses affected lipid metabolism genes, ROS, and glutathione production. Transcript changes correlated with metabolic data, which suggested the effects of EZH2i on ferroptosis. We found that EZH2i dose-dependently increased SLC7A11/glutathione axis and glutathione peroxidase-4 (GPX4), required to counteract lipid peroxidation and ferroptosis. A GPX4 inhibitor synergized with EZH2i, making low doses - which otherwise do not affect cell viability - able to significantly reduce ACC cell growth in vitro and in vivo. Importantly, we found that the anti-ferroptosis defense mechanism induced by EZH2i is a common response for several aggressive tumor phenotypes, uncovering a general co-targetable mechanism that could limit EZH2i effectiveness. Correcting this antioxidant response by ferroptosis inducers may be a new combination therapy that will easily find clinical applications.
Collapse
Affiliation(s)
- Marta C Nocito
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091, Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307, Dresden, Germany
| | - Antonio M Lerario
- Departments of Molecular and Integrative Physiology and Internal Medicine, University of Michigan, Medical School, 48104, Ann Arbor, MI, USA
| | - Fabrizio Mastrorocco
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy (CNR), 70126, Bari, Italy
| | - Luca De Martino
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy (CNR), 70126, Bari, Italy
| | - Clara Musicco
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy (CNR), 70126, Bari, Italy
| | - Ida D Perrotta
- Centre for Microscopy and Microanalysis (CM2), Department of Biology, Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Mariafrancesca Scalise
- Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Cesare Indiveri
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy (CNR), 70126, Bari, Italy
- Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Sergio Giannattasio
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council of Italy (CNR), 70126, Bari, Italy
| | - Pierre Val
- France iGReD (Institute of Genetics, Reproduction and Development), CNRS UMR 6293, Inserm U1103, Université Clermont Auvergne, 28 Place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Marilena Lanzino
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
- Centro Sanitario, University of Calabria, Ponte P. Bucci, 87036, Rende, Italy.
| | - Vincenzo Pezzi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| | - Ivan Casaburi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Rosa Sirianni
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| |
Collapse
|
28
|
Morris AB, Adelman MW, Bennion KB, Martinez CD, McCook KM, Woodworth MH, Langelier CR, Rouphael N, Scharer CD, Maier CL, Kraft CS, Ford ML. Fgl2 regulates FcγRIIB+CD8+ T cell responses during infection. JCI Insight 2025; 10:e186259. [PMID: 40197366 PMCID: PMC11981615 DOI: 10.1172/jci.insight.186259] [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: 08/22/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025] Open
Abstract
While the inhibitory receptor FcγRIIB has been shown to be upregulated on activated CD8+ T cells in both mice and humans, its effect on T cell fate during infection has not been fully elucidated. We identified an increase in FcγRIIB-expressing CD8+ T cells in patients with COVID-19 relative to healthy controls as well as in mouse models of viral infection. Despite its well-known role as an Fc receptor, FcγRIIB also ligates the immunosuppressive cytokine Fgl2, resulting in CD8+ T cell apoptosis. Both chronic LCMV infection in mice and COVID-19 in humans resulted in a significant increase in plasma Fgl2. Transfer of CD8+ T cells into a Fgl2-replete, but not Fgl2-devoid, environment resulted in elimination of FcγRIIB+, but not FcγRIIB-, CD8+ T cells. Similarly, plasma Fgl2 was directly proportional to CD8+ T cell lymphopenia in patients with COVID-19. RNA-Seq analysis demonstrated that Fgl2 was produced by murine virus-specific CD8+ T cells, with an increase in Fgl2 in CD8+ T cells elicited during chronic versus acute viral infection. Fgl2 was also upregulated in CD8+ T cells from patients with COVID-19 versus healthy controls. In summary, CD8+ T cell production of Fgl2 during viral infection underpinned an FcγRIIB-mediated loss of CD8+ T cell immunity in both mice and humans.
Collapse
Affiliation(s)
| | - Max W. Adelman
- Department of Medicine, Division of Infectious Diseases, Emory University, Atlanta, Georgia, USA
| | | | | | | | - Michael H. Woodworth
- Department of Medicine, Division of Infectious Diseases, Emory University, Atlanta, Georgia, USA
| | - Charles R. Langelier
- Department of Medicine, Division of Infectious Diseases, University of California, San Francisco, California, USA
| | - Nadine Rouphael
- Department of Medicine, Division of Infectious Diseases, Emory University, Atlanta, Georgia, USA
| | | | - Cheryl L. Maier
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia, USA
| | - Colleen S. Kraft
- Department of Medicine, Division of Infectious Diseases, Emory University, Atlanta, Georgia, USA
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant Center and
| |
Collapse
|
29
|
Bouchama A, Gomez M, Abdullah ML, Al Mahri S, Malik SS, Yezli S, Mohammad S, Lehe C, Abuyassin B, Hoehndorf R. Whole genome transcriptomic profiling reveals distinct sex-specific responses to heat stroke. J Appl Physiol (1985) 2025; 138:964-978. [PMID: 40066897 DOI: 10.1152/japplphysiol.00001.2025] [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: 01/02/2025] [Revised: 01/18/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
Heat-related mortality remains health challenges exacerbated by climate change, with sex-based differences in outcomes, yet underlying mechanisms remain poorly understood. This study examined transcriptomic responses to heat exposure in peripheral blood mononuclear cells from 19 patients with heat stroke (HS; 8 males, mean age 64.8 ± 6.6 yr; 11 females, mean age 49.7 ± 11 yr) and 19 controls (11 males, mean age 48.9 ± 9.6 yr; 8 females, mean age 44.9 ± 11.8 yr). At admission, gene expression revealed upregulation of heat shock protein genes, and pathway analysis demonstrated activation of heat shock and unfolded protein responses across both sexes consistent with proteotoxic stress. However, distinct metabolic, oxidative stress, cell cycle control, and immune responses were observed within each sex. Females displayed inhibition of protein synthesis, oxidative phosphorylation, and metabolic pathways, including glucose metabolism, indicative of a hypometabolic state. Males maintained metabolic activity precooling and enhanced adenosine triphosphate production postcooling. Females activated nuclear factor erythroid 2-related factor 2 (NRF2)-mediated oxidative stress responses and inhibited DNA replication and mitosis, potentially mitigating genomic instability, whereas these pathways showed limited regulation in males. Females promoted innate immunity via interleukin (IL)-6, inflammasome, and triggering receptor expressed on myeloid cells 1 (TREM1) signaling, whereas males showed suppression of both innate and adaptive immunity, including IL-12, Th1, and T-cell receptor pathways. Upstream analysis identified over 100 transcription factors in both sexes. Males primarily relied on transcriptional mechanisms, whereas females also exhibited translational regulation via La ribonucleoprotein 1 (LARP1), fragile X messenger ribonucleoprotein 1 (FMR1), insulin-like growth factor 2 mRNA binding protein 1 (IGF2BP1), and eukaryotic translation initiation factor 6 (EIF6). These findings suggest distinct, sex-specific molecular adaptations to heat stroke, underscoring the need for targeted therapeutic strategies to mitigate heat-induced morbidity and mortality.NEW & NOTEWORTHY Heat-related mortality continues to rise with climate change. Our transcriptomic analysis reveals distinct sex-specific metabolic strategies to heat stroke: females enter a hypometabolic state, an evolutionary adaptation that conserves energy, whereas males sustain metabolic activity. Transcription factors and a subset of translation regulators in females modulate proteostasis and bioenergetics, driving these sex-specific pathways. These novel findings highlight the critical need to consider sex-specific differences in heat-related illnesses and inform carefully targeted interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Abderrezak Bouchama
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Maria Gomez
- Computational Bioscience Research Center (CBRC) Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Mashan L Abdullah
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Saeed Al Mahri
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Shuja Shafi Malik
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Saber Yezli
- Biostatistics, Epidemiology and Scientific Computing Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sameer Mohammad
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Cynthia Lehe
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Bisher Abuyassin
- Experimental Medicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Robert Hoehndorf
- Computer, Electrical, and Mathematical Sciences & Engineering (CEMSE) Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| |
Collapse
|
30
|
Zheng W, Zhou C, Xue Z, Qiao L, Wang J, Lu F. Integrative analysis of a novel signature incorporating metabolism and stemness-related genes for risk stratification and assessing clinical outcomes and therapeutic responses in lung adenocarcinoma. BMC Cancer 2025; 25:591. [PMID: 40170009 PMCID: PMC11963273 DOI: 10.1186/s12885-025-13984-6] [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: 09/04/2024] [Accepted: 03/20/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Metabolism and stemness-related genes (msRGs) are critical in the development and progression of lung adenocarcinoma (LUAD). Nevertheless, reliable prognostic risk signatures derived from msRGs have yet to be established. METHODS In this study, we downloaded and analyzed RNA-sequencing and clinical data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. We employed univariate and multivariate Cox regression analyses, along with least absolute shrinkage and selection operator (LASSO) regression analysis, to identify msRGs that are linked to the prognosis of LUAD and to develop the prognostic risk signature. The prognostic value was evaluated using Kaplan-Meier analysis and log-rank tests. We generated receiver operating characteristic (ROC) curves to evaluate the predictive capability of the prognostic signature. To estimate the relative proportions of infiltrating immune cells, we utilized the CIBERSORT algorithm and the MCPCOUNTER method. The prediction of the half-maximal inhibitory concentration (IC50) for commonly used chemotherapy drugs was conducted through ridge regression employing the "pRRophetic" R package. The validation of our analytical findings was performed through both in vivo and in vitro studies. RESULTS A novel five-gene prognostic risk signature consisting of S100P, GPX2, PRC1, ARNTL2, and RGS20 was developed based on the msRGs. A risk score derived from this gene signature was utilized to stratify LUAD patients into high- and low-risk groups, with the former exhibiting significantly poorer overall survival (OS). A nomogram was constructed incorporating the risk score and other clinical characteristics, showcasing strong capabilities in estimating the OS rates for LUAD patients. Furthermore, we observed notable differences in the infiltration of various immune cell subtypes, as well as in responses to immunotherapy and chemotherapy, between the low-risk and high-risk groups. Results from gene set enrichment analysis (GSEA) and in vitro studies indicated that the prognostic signature gene ARNTL2 influenced the prognosis of LUAD patients, primarily through the activation of the PI3K/AKT/mTOR signaling pathway. CONCLUSIONS Utilizing this gene signature for risk stratification could help with clinical treatment management and improve the prognosis of LUAD patients.
Collapse
Affiliation(s)
- Wanrong Zheng
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Chuchu Zhou
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Zixin Xue
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Ling Qiao
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jianjun Wang
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Feng Lu
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China.
- Joint National Laboratory for Antibody Drug Engineering, Henan University, Kaifeng, China.
| |
Collapse
|
31
|
Mikołajczyk-Stecyna J, Zuk E, Chmurzynska A, Blatkiewicz M, Jopek K, Rucinski M. Exposure to a choline-deficient diet during pregnancy and lactation alters the liver transcriptome profile in offspring of dams with fatty liver. Clin Nutr ESPEN 2025; 66:9-23. [PMID: 39800134 DOI: 10.1016/j.clnesp.2025.01.028] [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: 10/28/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS The developmental origin of health and disease hypothesis shows that early adverse exposures can have lifelong health effects. Thus, the aim of this study was to analyze the impact of choline intake during pregnancy and/or lactation on gene expression profiles in the liver of 24-day-old male rat offspring from dams with non-alcoholic fatty liver disease (NAFLD). METHODS Phenotypic characteristic, histological examination and global transcriptome pattern of liver tissue specimens obtained from offspring of dams suffering from fatty liver, provided with proper choline intake during pregnancy and lactation (NN), fed a choline-deficient diet during both periods (DD), deprived of choline only during pregnancy (DN), or only during lactation (ND), was performed. The global gene expression profile was analyzed by using microarray approach (Affymetrix® Rat Gene 2.1 ST Array Strip). The relative expression of selected genes was validated by real-time polymerase chain reaction (qPCR). RESULTS The histological examination of rat liver sections indicated alternations typical for fatty liver in all analyzed groups with increased progression among groups deprived of choline. Choline deficiency in the maternal diet was associated with changes in body mass and composition but not with biochemical marker levels, except for the high density lipoprotein fraction of cholesterol (HDL). Enhanced expression of genes involved in oxidative stress, cell proliferation, activation of catabolic processes related to hepatocyte dysfunction and cell membrane composition were simultaneously observed in all choline-deficient groups. CONCLUSIONS An adequate amount of choline in the diet of a mother with fatty liver during pregnancy and/or lactation can regulate gene expression in the offspring's liver and contribute to a milder stage of the disease in the progeny. Moreover, proper choline supply during the postpartum period is as crucial as during the prenatal period.
Collapse
Affiliation(s)
- Joanna Mikołajczyk-Stecyna
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland.
| | - Ewelina Zuk
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland
| | - Agata Chmurzynska
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Wojska Polskiego 31, 60-624 Poznań, Poland
| | | | - Karol Jopek
- University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Marcin Rucinski
- University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| |
Collapse
|
32
|
Xu Y, Cao X, Zhou H, Xu H, Chen B, Bai H. Identifying potential prognosis markers in relapsed multiple myeloma via integrated bioinformatics analysis and biological experiments. Curr Res Transl Med 2025; 73:103495. [PMID: 39818172 DOI: 10.1016/j.retram.2025.103495] [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: 01/14/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Almost all multiple myeloma (MM) patients will eventually develop disease that has relapsed with or become refractory to current therapeutic regimes. However, the pervious clinical parameters have been proved inaccurate for defining MM relapse, and molecular targets have become the focuses of interests. Prognostic predictions based on molecular targets have been more effective to this day. Our research was performed to demonstrate hub genes involving relapsed MM by bioinformatics and biological experiments. METHODS AND RESULTS The integrated bioinformatics analysis in baseline and relapsed MM patients were executed. Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were utilized to analyze biologic functions of up-regulated differentially expressed genes (DEGs). Four hub genes (CENPE, ASPM, TOP2A and FANCI) were adopted for construction of relapsed gene score model (RGS), and RGS model was evaluated in two testing sets. The CENPE inhibitor GSK923295 had anti-myeloma effect, including promoting cell death, cell cycle arrest and DNA damage of MM cell lines. CONCLUSION Through bioinformatics analysis, we found that the four hub genes (CENPE, ASPM, TOP2A and FANCI) were associated to cell cycle, nuclear division, mitosis and spindle. Our research provided proof-of-concept that RGS model could be utilized to estimate recurrence risk and prognosis for patients, and targeting CENPE contributed to developing novel therapeutic pattern for MM.
Collapse
Affiliation(s)
- Yong Xu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinya Cao
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - He Zhou
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Xu
- School of Medicine, Southeast University, Nanjing, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hua Bai
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
33
|
Nguyen NTB, Gevers S, Kok RNU, Burgering LM, Neikes H, Akkerman N, Betjes MA, Ludikhuize MC, Gulersonmez C, Stigter ECA, Vercoulen Y, Drost J, Clevers H, Vermeulen M, van Zon JS, Tans SJ, Burgering BMT, Rodríguez Colman MJ. Lactate controls cancer stemness and plasticity through epigenetic regulation. Cell Metab 2025; 37:903-919.e10. [PMID: 39933514 DOI: 10.1016/j.cmet.2025.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 11/04/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025]
Abstract
Tumors arise from uncontrolled cell proliferation driven by mutations in genes that regulate stem cell renewal and differentiation. Intestinal tumors, however, retain some hierarchical organization, maintaining both cancer stem cells (CSCs) and cancer differentiated cells (CDCs). This heterogeneity, coupled with cellular plasticity enabling CDCs to revert to CSCs, contributes to therapy resistance and relapse. Using genetically encoded fluorescent reporters in human tumor organoids, combined with our machine-learning-based cell tracker, CellPhenTracker, we simultaneously traced cell-type specification, metabolic changes, and reconstructed cell lineage trajectories during tumor organoid development. Our findings reveal distinctive metabolic phenotypes in CSCs and CDCs. We find that lactate regulates tumor dynamics, suppressing CSC differentiation and inducing dedifferentiation into a proliferative CSC state. Mechanistically, lactate increases histone acetylation, epigenetically activating MYC. Given that lactate's regulation of MYC depends on the bromodomain-containing protein 4 (BRD4), targeting cancer metabolism and BRD4 inhibitors emerge as a promising strategy to prevent tumor relapse.
Collapse
Affiliation(s)
- Nguyen T B Nguyen
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Sira Gevers
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Rutger N U Kok
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Lotte M Burgering
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Hannah Neikes
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen 6525 GA, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Ninouk Akkerman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Marlies C Ludikhuize
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Can Gulersonmez
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Edwin C A Stigter
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Yvonne Vercoulen
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Jarno Drost
- Oncode Institute, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Hans Clevers
- Oncode Institute, Utrecht, the Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen 6525 GA, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | | | - Sander J Tans
- AMOLF, Amsterdam, the Netherlands; Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, the Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Maria J Rodríguez Colman
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
34
|
Fukuda Y, Kawaguchi K, Fukushima A. AtSRGA: A shiny application for retrieving and visualizing stress-responsive genes in Arabidopsis thaliana. PLANT PHYSIOLOGY 2025; 197:kiaf105. [PMID: 40112239 DOI: 10.1093/plphys/kiaf105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
Abiotic and biotic stresses pose serious threats to plant productivity. Elucidating the gene regulatory networks involved in plant stress responses is essential for developing future breeding programs and innovative agricultural products. Here, we introduce the AtSRGA (Arabidopsis thaliana Stress-Responsive Gene Atlas), a user-friendly application facilitating the retrieval of stress-responsive genes in Arabidopsis (Arabidopsis thaliana). The application was developed using 1,131 microarrays and 1,050 RNA sequencing datasets obtained from public databases. These datasets correspond to 11 stress-related conditions, namely abscisic acid, cold, drought, heat, high-light, hypoxia, osmotic stress, oxidative stress, salt, wounding, and Pseudomonas syringae pv. tomato DC3000. Using a modified meta-analysis technique known as the vote-counting method, we computed integrated scores to evaluate stress responsiveness for each condition across multiple studies. AtSRGA visualizes gene behavior under 11 stress conditions and offers an interactive, user-friendly interface accessible to all researchers. It presents a comprehensive heatmap of stress-responsive genes, facilitating the comparative analysis of individual stress responses and groups of genes responding to multiple stresses. We validated the expression patterns of several high-scoring genes of unknown function under cold and heat stress using RT-qPCR, thus demonstrating that our application helps select targets to understand stress-responsive gene networks in Arabidopsis. AtSRGA will improve the screening of stress-responsive genes in Arabidopsis, thereby supporting the advancement of plant science toward a sustainable society.
Collapse
Affiliation(s)
- Yusuke Fukuda
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto, Kyoto 606-8522, Japan
| | - Kohei Kawaguchi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto, Kyoto 606-8522, Japan
| | - Atsushi Fukushima
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto, Kyoto 606-8522, Japan
- RIKEN, Center for Sustainable Resource Science, 1-7-22 Suehiro, Tsurumi, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
35
|
Bhattacharjee K, Sengupta A, Kumar R, Ghosh A. Identification of key hub genes in pancreatic ductal adenocarcinoma: an integrative bioinformatics study. FRONTIERS IN BIOINFORMATICS 2025; 5:1536783. [PMID: 40226632 PMCID: PMC11985535 DOI: 10.3389/fbinf.2025.1536783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) poses a significant health threat characterized by poor clinical outcomes, largely attributable to late detection, chemotherapy resistance, and the absence of tailored therapies. Despite progress in surgical, radiation, and chemotherapy treatments, 80% of PDAC patients do not benefit optimally from systemic therapy, often due to asymptomatic presentation or disease regression upon diagnosis. The disease's progression is influenced by complex interactions involving immunological, genetic, and environmental factors, among others. However, the precise molecular mechanisms underlying PDAC remain incompletely understood. A major challenge in elucidating PDAC's origins lies in deciphering the genetic variations governing its network. PDAC exhibits heterogeneity, manifesting diverse genetic compositions, cellular attributes, and behaviors across patients and within tumors. This diversity complicates diagnosis, treatment strategies, and prognostication. Identification of "Differentially Expressed Genes" (DEGs) between PDAC and healthy controls is vital for addressing these challenges. These DEGs serve as the foundation for constructing the PDAC protein interaction network, with their network properties being assessed for further insights. Our analysis revealed five key hub genes (KHGs): EGF, SRC, SDC1, ICAM1 and CEACAM5. The KHGs were predominantly enriched in pathways such as: ErbB signaling pathway, Rap1 signaling pathway, etc. Acknowledging the therapeutic promise and biomarker importance of PDAC KHGs, we have also pinpointed approved medications for the identified key genes. Nevertheless, it is crucial to conduct experimental validation on KHGs to confirm their effectiveness within the PDAC context. Overall, this study identified potential key hub genes implicated in the progression of PDAC, offering significant guidance for personalized clinical decision-making and molecular-targeted therapy for PDAC patients.
Collapse
Affiliation(s)
| | - Avik Sengupta
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana, India
| | - Aryya Ghosh
- Department of Chemistry, Ashoka University, Sonipat, Haryana, India
| |
Collapse
|
36
|
Liu J, Yan M, Chen L, Yu W, Lü Y. Construction and evaluation of a diagnostic model for Alzheimer's disease based on mitophagy-related genes. Sci Rep 2025; 15:10632. [PMID: 40148430 PMCID: PMC11950216 DOI: 10.1038/s41598-025-89980-4] [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: 11/11/2024] [Accepted: 02/10/2025] [Indexed: 03/29/2025] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Mitophagy fulfills crucial functions in neurodegenerative disorders and neuronal survival but the relationship between mitophagy and AD is unclear. Mitophagy correlation scores between AD samples and control samples were calculated using single-sample GSEA (ssGSEA) based on two datasets from gene expression omnibus (GEO) database. Mitophagy-related genes (MRGs) and differentially expressed genes (DEGs) in AD screened by WGCNA and "limma" package were intersected to take common genes. These overlapping genes were further compressed and used for diagnostic modeling by adopting the recursive feature elimination (RFE) and LASSO analysis. The reliability of the diagnostic model was verified based on the receiver operating characteristic (ROC) curve. Then, a transcription factor (TF)-mRNA regulatory network of these key genes was established. Lastly, ssGSEA was employed to examine the relationship between the identified genes and cellular pathways and immune cell infiltration. AD samples had notably lower mitophagy correlation scores than control samples. A total of 12 MRGs in the module with the greatest mitophagy connection with AD patients were identified. Functional enrichment analysis revealed that the DEGs were significantly enriched in synaptic function-related pathways. Based on GSE122063, a diagnostic prediction model was created and validated using two mitophagy-related genes (YWHAZ and NDE1), showing an area under ROC curve (AUC) greater than 0.7. This confirmed that the diagnostic model had a high predictive value. The TF-mRNA network showed that four TFs, namely, FOXC1, FOXL1, HOXA5 and GATA2, were regulated by both YWHAZ and NDE1 genes. Immune infiltration analysis revealed that NDE1 promoted the infiltration of most immune cells, while YWHAZ mainly inhibited the infiltration of most immune cells. The current findings improved our understanding of mitophagy in AD, contributing to future research and treatment development in AD.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Mengyu Yan
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Lihua Chen
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weihua Yu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China.
| | - Yang Lü
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
37
|
Gao X, Fan Y, Wang G, Xu J, Deng R, Song J, Sun B, Wang Y, Wu Z, Jia R, Huang J, He H, Gao L, Zhang Y, Sun N, Wu B. Combined analysis of single-cell and bulk transcriptome sequencing data identifies critical glycolysis genes in idiopathic pulmonary arterial hypertension. J Transl Med 2025; 23:373. [PMID: 40140873 PMCID: PMC11948795 DOI: 10.1186/s12967-025-06373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Abnormal glycolytic metabolism plays a significant role in pulmonary vascular remodeling in idiopathic pulmonary arterial hypertension (IPAH), yet the specific mechanisms remain unclear. The primary objective of this study is to investigate the key regulatory mechanisms of glycolysis in IPAH. METHODS Bulk and single-cell sequencing data obtained from IPAH patient tissue samples were downloaded from the GEO database. scMetabolism and AUCcell analyses of the IPAH single-cell sequencing data were carried out to quantify the glycolytic metabolic activity and identify the main cell types regulating glycolysis, respectively. The ssGSEA method was used to assess the glycolytic activity in each bulk sample within the bulk sequencing data. Differential analysis, weighted gene co-expression network analysis (WGCNA), and protein-protein interaction (PPI) network analysis were conducted to identify key genes associated with glycolysis in IPAH samples. Single-cell sequencing and a monocrotaline (MCT)-induced model of PH in rats were utilized to validate the expression of these key genes. RESULTS Single-cell sequencing data indicated that IPAH patients displayed increased glycolytic activity, which was primarily regulated by fibroblasts. Similarly, bulk transcriptomic data revealed a significant increase in glycolytic activity in IPAH patients. Differential analysis, WGCNA, PPI network analysis, and integrated single-cell analysis further identified insulin-like growth factor-1 (IGF1), lysyl-tRNA synthetase (KARS), caspase-3 (CASP3), and cyclin-dependent kinase inhibitor 2 A (CDKN2A) as key genes associated with fibroblast-mediated glycolysis in IPAH patients. Differential expression of IGF1, KARS, CASP3, and CDKN2A was also observed in our in vivo model of PH. CONCLUSION Our study identifies IGF1, KARS, CASP3, and CDKN2A as key regulatory genes in glycolysis in IPAH, which provides the basis for the development of targeted therapies.
Collapse
Affiliation(s)
- Xuan Gao
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Youli Fan
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Guijia Wang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Jiangjiang Xu
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Runwei Deng
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Jiangwei Song
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Binfeng Sun
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Yongbing Wang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Zixuan Wu
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Ruyi Jia
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Jing Huang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Huiyu He
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China
| | - Lei Gao
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Yihao Zhang
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China
| | - Na Sun
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China.
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China.
| | - Bingxiang Wu
- Department of Cardiology, The Key Laboratory of Myocardial Ischemia, Ministry of Education, The Second Affiliated Hospital of Harbin Medical University, NO.246 Xuefu Road, Nangang District, Harbin, 150086, China.
- Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150086, China.
| |
Collapse
|
38
|
Zhang T, Zhou H. Machine Learning and Weighted Gene Coexpression Network-Based Identification of Biomarkers Predicting Immune Profiling and Drug Resistance in Lung Adenocarcinoma. Int J Genomics 2025; 2025:9923294. [PMID: 40161493 PMCID: PMC11955064 DOI: 10.1155/ijog/9923294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Background: The prognosis for lung adenocarcinoma (LUAD) is poor, and the recurrence rate is high. Thus, to evaluate patients' prognoses and direct therapy choices, new prognostic markers are desperately needed. Methods: First, gene modules associated with LUAD were identified by weighted gene coexpression network analysis (WGCNA) analysis. The expression profiles obtained were intersected with the differential expressed genes taken between LUAD samples and paracancerous samples. Afterward, stepwise regression analysis and the LASSO were used to compress the genes further, and a risk model was created. Furthermore, a nomogram based on risk scores and clinical features was created to validate the model. After that, the distinctions between the pertinent biological processes and signaling pathways among the various subgroups were investigated. Additionally, drug sensitivity testing, immunotherapy, immune infiltration analysis, and enrichment analysis were carried out. Finally, the biological role of ANLN in LUAD was explored by qPCR, cell scratch assay, and transwell. Results: A total of 257 intersected genes were obtained by taking the intersection of the differential genes between 2866 LUAD samples and paraneoplastic samples with the module genes after we screened two particular modules that had the strongest link with LUAD by WGCNA. ANLN, CASS4, and NMUR1 were found to be distinctive genes for the development of risk models after the intersecting genes were screened to find 176 genes linked to the prognosis for LUAD. Based on risk assessments, high- and low-risk groups of LUAD patients were divided. Low-risk patients exhibited a significantly higher overall survival (OS) than those in the high-risk group. Expression of model genes correlates with infiltration of the vast majority of immune cells. Significant differences in the biological pathways, immune microenvironment, and abundance of immune cell infiltration were found between the two groups. The drug sensitivity study showed that patients in the high-risk group had higher IC50 values for BMS-754807_2171 and Doramapimod_10424. Finally, in vitro experiments demonstrated that knocking down ANLN noticeably inhibited the viability, migration, and invasion of A549 cells. Conclusion: This study may provide a theoretical reference for future exploration of potential diagnostic and prognostic biomarkers for LUAD.
Collapse
Affiliation(s)
- Tian Zhang
- Pharmacy Department, Xiangxi Autonomous Prefecture People's Hospital, Jishou, China
| | - Han Zhou
- Pharmacy Department, Xiangxi Autonomous Prefecture People's Hospital, Jishou, China
| |
Collapse
|
39
|
Turky MA, Youssef I, El Amir A. Identifying behavior regulatory leverage over mental disorders transcriptomic network hubs toward lifestyle-dependent psychiatric drugs repurposing. Hum Genomics 2025; 19:29. [PMID: 40102990 PMCID: PMC11921594 DOI: 10.1186/s40246-025-00733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND There is a vast prevalence of mental disorders, but patient responses to psychiatric medication fluctuate. As food choices and daily habits play a fundamental role in this fluctuation, integrating machine learning with network medicine can provide valuable insights into disease systems and the regulatory leverage of lifestyle in mental health. METHODS This study analyzed coexpression network modules of MDD and PTSD blood transcriptomic profile using modularity optimization method, the first runner-up of Disease Module Identification DREAM challenge. The top disease genes of both MDD and PTSD modules were detected using random forest model. Afterward, the regulatory signature of two predominant habitual phenotypes, diet-induced obesity and smoking, were identified. These transcription/translation regulating factors (TRFs) signals were transduced toward the two disorders' disease genes. A bipartite network of drugs that target the TRFS together with PTSD or MDD hubs was constructed. RESULTS The research revealed one MDD hub, the CENPJ, which is known to influence intellectual ability. This observation paves the way for additional investigations into the potential of CENPJ as a novel target for MDD therapeutic agents development. Additionally, most of the predicted PTSD hubs were associated with multiple carcinomas, of which the most notable was SHCBP1. SHCBP1 is a known risk factor for glioma, suggesting the importance of continuous monitoring of patients with PTSD to mitigate potential cancer comorbidities. The signaling network illustrated that two PTSD and three MDD biomarkers were co-regulated by habitual phenotype TRFs. 6-Prenylnaringenin and Aflibercept were identified as potential candidates for targeting the MDD and PTSD hubs: ATP6V0A1 and PIGF. However, habitual phenotype TRFs have no leverage over ATP6V0A1 and PIGF. CONCLUSION Combining machine learning and network biology succeeded in revealing biomarkers for two notoriously spreading disorders, MDD and PTSD. This approach offers a non-invasive diagnostic pipeline and identifies potential drug targets that could be repurposed under further investigation. These findings contribute to our understanding of the complex interplay between mental disorders, daily habits, and psychiatric interventions, thereby facilitating more targeted and personalized treatment strategies.
Collapse
Affiliation(s)
| | - Ibrahim Youssef
- Faculty of Engineering, Biomedical Engineering Department, Cairo University, Giza, 12613, Egypt
| | - Azza El Amir
- Faculty of Science, Biotechnology Department, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
40
|
Qu G, Liu K, Xu W, Li D. Integrated analysis and experimental validation reveal the prognostic and immunological features associated with coagulation in hepatocellular carcinoma. Sci Rep 2025; 15:8626. [PMID: 40074769 PMCID: PMC11904193 DOI: 10.1038/s41598-025-85491-4] [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: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 03/14/2025] Open
Abstract
Coagulation is intensively related to various tumors, which affects their progression and prognosis. However, research on the impact of coagulation-associated genes (CAGs) on hepatocellular carcinoma (HCC) occurrence, prognosis, and immune microenvironment is limited. Consequently, our research aims to uncover how CAGs affect the prognosis and immune microenvironments of HCC. We integrated gene expression data and clinical information from three datasets (GSE14520, GSE76427, and TCGA-LIHC). 281 CAGs were obtained from the coagulation-related pathway (hsa04610). We obtained three CAG patterns through a consensus clustering algorithm. Afterward, differential analyses of prognosis, biological processes, immune infiltration, and functional and pathway enrichment were conducted on the three CAG patterns. We intersected CAGs with differentially expressed genes in GSE76427 and then conducted Cox regression analysis to obtain the prognostic genes in HCC. Glycerol-3-phosphate dehydrogenase 2 (GPD2) was selected for further analyses. TCGA-LIHC samples with different GPD2 expression levels were analyzed for prognosis, DNA methylation, immune infiltration, and drug sensitivity. The expression level of GPD2 was verified through quantitative real-time PCR (qPCR) and immunohistochemistry. The wound-healing and Transwell assays were used to analyze the tumor cell migration and the Matrigel invasion and apoptosis assays were performed to determine cell invasion and apoptosis. Three CAG patterns were obtained through an unsupervised consensus clustering algorithm. CAGclusterA held the best prognosis compared to the other two clusters. The CAGclusterC was characterized by poor prognosis and abundant immune cell infiltration. The TCGA-LIHC dataset, as an internal validation, also yielded similar subtype classifications. Afterward, we identified the GPD2 gene, which significantly affected the prognosis of HCC and was positively correlated with the tumor progression. The upregulation of GPD2 expression was closely related to tumorigenic signatures and immune escape. The qPCR confirmed the upregulation of GPD2 expression in HCC tumor cell lines, compared to normal liver cell lines. Immunohistochemical staining confirmed the high expression of GPD2 in HCC tumor tissues compared to normal tissues. Regulating the expression level of GPD2 can inhibit the proliferation, migration, invasion, and induce apoptosis of HCC cells. Our study comprehensively elucidated the coagulation characteristics in HCC and identified a promising oncogenic gene GPD2. Exploring targeted strategies based on coagulation-related characteristics and biomarkers may shed light on HCC treatment.
Collapse
Affiliation(s)
- Guangzhen Qu
- Department of Interventional Radiology, Beijing Chao-Yang Hospital Affiliated with Capital Medical University, Beijing, 100020, China
| | - Kun Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Weiyu Xu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
41
|
Shi S, Hong X, Zhang Y, Chen S, Huang X, Zheng G, Hu B, Lu M, Li W, Zhong Y, Sun G, Ouyang Y. Exploring The Role of TOP2A in the Intersection of Pathogenic Mechanisms Between Rheumatoid Arthritis and Idiopathic Pulmonary Fibrosis Based on Bioinformatics. J Inflamm Res 2025; 18:3449-3468. [PMID: 40093950 PMCID: PMC11910056 DOI: 10.2147/jir.s497734] [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: 09/24/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background Rheumatoid arthritis (RA) and idiopathic pulmonary fibrosis (IPF) share a common pathogenic mechanism, but the underlying mechanisms remain ambiguous. Our study aims at exploring the genetic-level pathogenic mechanism of these two diseases. Methods We carried out bioinformatics analysis on the GSE55235 and GSE213001 datasets. Machine learning was employed to identify candidate genes, which were further verified using the GSE92592 and GSE89408 datasets, as well as quantitative real-time PCR (qRT-PCR). The expression levels of TOP2A in RA and IPF in vitro models were confirmed using Western blotting and qRT-PCR. Furthermore, we explored the influence of TOP2A on the occurrence and development of RA and IPF by using the selective inhibitor PluriSIn #2 in an in vitro model. Finally, an in vivo model of RA and IPF was constructed to assess TOP2A expression levels via immunohistochemistry. Results Our bioinformatics analysis suggests a potential intersection in the pathogenic mechanisms of RA and IPF. We have identified 7 candidate genes: CXCL13, TOP2A, MMP13, MMP1, LY9, TENM4, and SEMA3E. Our findings reveal that the expression level of TOP2A is significantly elevated in both in vivo and in vitro models of RA and IPF. Additionally, our research indicates that PluriSIn #2 can effectively restrain inflammatory factors, extracellular matrix deposition, migration, invasion, the expression and nuclear uptake of p-smad2/3 protein in RA and IPF in vitro models. Conclusion There is a certain correlation between RA and IPF at the genetic level, and the molecular mechanisms of their pathogenesis overlap, which might be the reason for the progression of RA. Among the candidate genes we identified, TOP2A may influence the occurrence and development of RA and IPF through the TGF-β/Smad signal pathway. This could be beneficial to the study of the pathogenesis and treatment of RA and IPF.
Collapse
Affiliation(s)
- Shoujie Shi
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Xin Hong
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Yue Zhang
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Shuilin Chen
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Xiangfei Huang
- Anesthesiology Department, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Guihao Zheng
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Bei Hu
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Meifeng Lu
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Weihua Li
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Yanlong Zhong
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Guicai Sun
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| | - Yulong Ouyang
- Department of Sports Medicine, Orthopaedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi Province, 330006, People's Republic of China
| |
Collapse
|
42
|
Liu FF, Li K. Molecular characterization underlying IFN-α2 treatment in polycythemia vera: a transcriptomic overview. Mol Cell Biochem 2025:10.1007/s11010-025-05238-7. [PMID: 40029555 DOI: 10.1007/s11010-025-05238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
Polycythemia vera (PV) is the most common chronic myeloproliferative neoplasm (MPN) in adults. Pegylated interferon-α2 (IFN-α2) is an effective and safe drug for the treatment of PV. However, the mechanisms of its action in PV are still not fully understood. Using the WGCNA and Limma algorithm, we found a subset of IFN-α2 sensitive genes and four gene co-expression modules. Meanwhile, we also found 820 genes were differentially expressed in PV compared with healthy controls. By integrating the above results, several differentially expressed genes (DEGs) that were up- or down-regulated in PV but showed opposite alterations in the IFN-α2-treated group were found. These genes were mainly related to three types of biological processes (metal ion homeostasis, metabolic/catabolic process, and Jak-STAT signaling pathway), the dysfunctions of which were prevalent in PV. Moreover, we applied another threshold-free analysis method to compare global gene expression between IFN-α2 treated PV, PV, and control groups. Results showed the transcriptome changes of PV versus controls were negatively correlated with that of IFN-α2 treated versus untreated PV, indicating IFN-α2 treatment could partially reverse the dysregulated gene expression profile due to PV pathology. In summary, interferon may alleviate the progression of PV through multiple pathways. The findings may be of assistance in understanding the molecular basis underlying this treatment.
Collapse
Affiliation(s)
- Fang-Fang Liu
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, People's Republic of China
| | - Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Hankou District, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
43
|
Schmidt ENC, Evert BO, Pregler BEF, Melhem A, Hsieh M, Raspe M, Strobel H, Roos J, Pietsch T, Schuss P, Fischer‐Posovszky P, Westhoff M, Hölzel M, Herrlinger U, Vatter H, Waha A, Schneider M, Potthoff A. Tonabersat enhances temozolomide-mediated cytotoxicity in glioblastoma by disrupting intercellular connectivity through connexin 43 inhibition. Mol Oncol 2025; 19:878-898. [PMID: 39680504 PMCID: PMC11887680 DOI: 10.1002/1878-0261.13786] [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: 10/09/2023] [Revised: 10/24/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastoma cells rely on connexin 43 (Cx43)-based gap junctions (GJs) for intercellular communication, enabling them to integrate into a widely branched malignant network. Although there are promising prospects for new targeted therapies, the lack of clinically feasible GJ inhibitors has impeded their adoption in clinical practice. In the present study, we investigated tonabersat (TO), a blood-brain-barrier-penetrating drug with GJ-inhibitory properties, in regard to its potential to disassemble intercellular connectivity in glioblastoma networks. Fluorescence-guided measurements of calcein cell-to-cell transfer were used to study functional intercellular connectivity. Specific DNA fragmentation rates of propidium iodide-stained nuclei were measured as a surrogate readout for cell death using flow cytometry. CRISPR/Cas9-mediated gene editing of Cx43 served as a validation tool of cellular effects related to Cx43 GJ inhibition. 3' mRNA sequencing was performed for molecular downstream analysis. We found that TO reduced intercellular GJ-mediated cytosolic traffic and yielded a significant reduction of tumor microtube (TM) length. TO-mediated inhibition of cellular tumor networks was accompanied by a synergistic effect for temozolomide-induced cell death. CRISPR/Cas9 Cx43-knockout revealed similar results, indicating that TO-mediated inhibitory effects rely on the inhibition of Cx43-based GJs. Gene set enrichment analyses found that GJ-mediated synergistic cytotoxic effects were linked to a significant upregulation of cell death signaling pathways. In conclusion, TO disrupts TM-based network connectivity via GJ inhibition and renders glioblastoma cells more susceptible to cytotoxic therapy. Given its previous use in clinical trials for migraine therapy, TO might harbor the potential of bridging the idea of a GJ-targeted therapeutic approach from bench to bedside.
Collapse
Affiliation(s)
- Elena N. C. Schmidt
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | | | - Barbara E. F. Pregler
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Ahmad Melhem
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Meng‐Chun Hsieh
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Markus Raspe
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Hannah Strobel
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
| | - Julian Roos
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
| | | | - Patrick Schuss
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Present address:
Department of NeurosurgeryBG Klinikum Unfallkrankenhaus Berlin BGGermany
| | - Pamela Fischer‐Posovszky
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
- German Center for Child and Adolescent Health (DZKJ), partner site UlmGermany
| | - Mike‐Andrew Westhoff
- Department of Pediatrics and Adolescent MedicineUniversity Medical Center UlmGermany
| | - Michael Hölzel
- Institute of Experimental OncologyUniversity Hospital BonnGermany
| | - Ulrich Herrlinger
- Department of Neurooncology, Center for Neurology and Center of Integrated Oncology ABCDUniversity Hospital BonnGermany
| | - Hartmut Vatter
- Department of NeurosurgeryUniversity Hospital BonnGermany
| | - Andreas Waha
- Department of NeuropathologyUniversity Hospital BonnGermany
| | - Matthias Schneider
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
| | - Anna‐Laura Potthoff
- Department of NeurosurgeryUniversity Hospital BonnGermany
- Brain Tumor Translational Research GroupUniversity Hospital BonnGermany
- Department of NeuropathologyUniversity Hospital BonnGermany
| |
Collapse
|
44
|
Liu X, Liu X, Wan B, Ge Y, Hu H, Yu H, Zhao M, Li H, Zhu J. Identification of the Hub Gene LDB3 in Stanford Type A Aortic Dissection Based on Comprehensive Bioinformatics Analysis. J Cell Mol Med 2025; 29:e70471. [PMID: 40099963 PMCID: PMC11916769 DOI: 10.1111/jcmm.70471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
Stanford type A aortic dissection (TAAD) is a life-threatening disease. This study explored the role of LIM domain binding 3 (LDB3) in TAAD progression. Four datasets from the Gene Expression Omnibus were analyzed to identify TAAD-related hub genes. LDB3 single nucleotide polymorphisms (SNPs) were assessed in the UK Biobank. Western blotting and immunofluorescence detected LDB3 expression in angiotensin II (Ang II) stimulated human aortic vascular smooth muscle cells (HA-VSMC), human samples, and a murine model. Bioinformatics identified tissue inhibitor of metalloproteinase-1 (TIMP1) and LDB3 as TAAD hub genes. TIMP1 was expressed in macrophages, mesenchymal cells, and smooth muscle cells, while LDB3 was mostly expressed in smooth muscle cells. Validation showed TIMP1 was upregulated and LDB3 downregulated in TAAD. Six LDB3 SNPs were associated with aortic aneurysm and dissection in the UK Biobank. In human and murine samples, LDB3 expression was reduced in diseased tissues and co-localized with smooth muscle. Ang II-stimulated HA-VSMC exhibited LDB3 reduction and altered intercellular connections. The aforementioned findings suggest that the newly identified gene LDB3 is crucial in the progression of TAAD.
Collapse
Affiliation(s)
- Xinyi Liu
- Department of Cardiovascular Surgery, Beijing Aortic Disease CenterBeijing Anzhen Hospital of Capital Medical UniversityBeijingChina
| | - Xing Liu
- Department of Cardiovascular SurgeryXinqiao Hospital of Army Medical UniversityChongqingChina
| | - Bin Wan
- Max Planck Insititute for Human Cognitive and Brain SciencesLeipzigGermany
- Institute of Neuroscience and Medicine (INM‐7: Brain and Behavior)Research Centre JülichJülichGermany
| | - Yipeng Ge
- Department of Cardiovascular Surgery, Beijing Aortic Disease CenterBeijing Anzhen Hospital of Capital Medical UniversityBeijingChina
| | - Haiou Hu
- Department of Cardiovascular Surgery, Beijing Aortic Disease CenterBeijing Anzhen Hospital of Capital Medical UniversityBeijingChina
| | - Hong Yu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Meng Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Huadong Li
- Department of Cardiovascular Surgery Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Aortic Disease CenterBeijing Anzhen Hospital of Capital Medical UniversityBeijingChina
| |
Collapse
|
45
|
Wu Y, Lin C, Qian Y, Huang X, Xu Y, Li J, He Y, Xie C, Su H. Identification of immune subtypes associated with CD8+ T cell-related genes providing new treatment strategies of esophageal carcinoma. Front Immunol 2025; 16:1512230. [PMID: 40083549 PMCID: PMC11903738 DOI: 10.3389/fimmu.2025.1512230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Background CD8+ T lymphocytes greatly affect the efficacy of immunotherapy, displaying promising potential in various tumors. Here, we aimed to identify immune subtypes associated with CD8+ T cell-related genes to predict the efficacy of treatment in esophageal cancer (ESCA). Methods We obtained 13 immune cell-related datasets from the Gene Expression Omnibus (GEO) database and removed batch effects. Weighted correlation network analysis (WGCNA) and co-expression analysis were performed to identify highly correlated CD8+ T cell genes. Cox analysis was used to process ESCA clinical information, and the immune clusters (ICs) were constructed through consensus cluster analysis. Furthermore, we constructed an immune risk score model to predict the prognosis of ESCA based on these CD8+ T cell genes. This model was verified using the IMvigor210 dataset, and we functionally validated the immune risk score model in vitro. Results The results revealed significant correlations between CD8+ T cell-related genes and immune-related pathways. Three ICs were identified in ESCA, with IC3 demonstrating the most favorable prognosis. The final 6-gene prognostic risk model exhibited stable predictive performance in datasets across different platforms. Compared with that in normal esophageal epithelial (HEEC cells), CHMP7 in the 6-gene prognostic risk model was upregulated in KYSE150 and TE-1 cells. Si-CHMP7 transfection led to a decrease in tumor cell migration, invasion, and proliferation, accompanied by an accelerated apoptotic process. Conclusions Collectively, we identified the immune subtypes of CD8+ T cell-related genes with different prognostic significance. We designated CHMP7 in the 6-gene prognostic risk model as a potential target to improve tumor cell prognosis. These insights provide a strong basis for improving prognosis and facilitating more personalized and accurate treatment decisions for the immunotherapy of ESCA.
Collapse
Affiliation(s)
- Youyi Wu
- Department Oncology Radiotherapy, The Third Affiliated Hospital of Wenzhou Medical University, Rui’an People Hospital, Ruian, Zhejiang, China
| | - Chen Lin
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuchen Qian
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yajing Xu
- Department of Radiation Oncology Wenzhou Central Hospital Theorem Hospital Affiliated of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Youdi He
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huafang Su
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
Chen S, Zhang W, Liu Y, Huang R, Zhou X, Wei X. Revolutionizing the treatment of intervertebral disc degeneration: an approach based on molecular typing. J Transl Med 2025; 23:227. [PMID: 40001145 PMCID: PMC11863857 DOI: 10.1186/s12967-025-06225-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is a significant cause of global disability, reducing labor productivity, increasing the burden on public health, and affecting socio-economic well-being. Currently, there is a lack of recognized clinical approaches for molecular classification and precision therapy. METHODS Chondrocyte differentiation and prognosis-related genes were extracted from single-cell RNA sequencing and multi-omics data in the Gene Expression Omnibus (GEO) database through chondrocyte trajectory analysis and non-parametric tests. Subsequently, a precise IVDD risk stratification system was developed using ConsensusClusterPlus analysis. The clinical significance of molecular typing was demonstrated through case-control trials involving IVDD patients. Specific inhibitors of molecular typing were predicted using the pRRophetic package in R language and then validated in vitro. RESULTS A stratified model for IVDD, considering chondrocyte differentiation and demonstrating high clinical relevance, was developed using a set of 44 chondrocyte fate genes. Extensive analyses of multi-omics data confirmed the clinical relevance of this model, indicating that cases in the High Chondrocyte Scoring Classification (HCSC) group had the most favorable prognosis, whereas those in the Low Chondrocyte Scoring Classification (LCSC) group had the worst prognosis. Additionally, clinical case-control studies provided evidence of the utility of IVDD molecular typing in translational medicine. A gene expression-based molecular typing approach was used to create a matrix identifying potential inhibitors specific to each IVDD subtype. In vitro experiments revealed that gefitinib, a drug designed for LCSC, not only had protective effects on chondrocytes but also could induce the conversion of LCSC into the HCSC subgroup. Therefore, IVDD molecular typing played a critical role in assisting clinicians with risk stratification and enabling personalized treatment decisions. CONCLUSION The results of the study have provided a comprehensive and clinically relevant molecular typing for IVDD, involving a precise stratification system that offers a new opportunity for customizing personalized treatments for IVDD.
Collapse
Affiliation(s)
- Shaofeng Chen
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China
- Department of Orthopaedic Surgery, China Coast Guard Hospital, Zhejiang, China
| | - Wei Zhang
- Department of Burn Surgery, Changhai Hospital, Shanghai, China
- Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- BGI research, BGI-Hangzhou, 310012, Hangzhou, China
| | - Runzhi Huang
- Department of Burn Surgery, Changhai Hospital, Shanghai, China.
- Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Xiaoyi Zhou
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China.
| | - Xianzhao Wei
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China.
| |
Collapse
|
47
|
Ceranski AK, Carreño-Gonzalez MJ, Ehlers AC, Hanssen KM, Gmelin N, Geyer FH, Kolodynska Z, Vinca E, Faehling T, Poeller P, Ohmura S, Cidre-Aranaz F, Schulze A, Grünewald TGP. Refined culture conditions with increased physiological relevance uncover oncogene-dependent metabolic signatures in Ewing sarcoma spheroids. CELL REPORTS METHODS 2025; 5:100966. [PMID: 39922188 PMCID: PMC11955266 DOI: 10.1016/j.crmeth.2025.100966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/23/2024] [Accepted: 01/17/2025] [Indexed: 02/10/2025]
Abstract
Ewing sarcoma (EwS) cell line culture largely relies on standard techniques, which do not recapitulate physiological conditions. Here, we report on a feasible and cost-efficient EwS cell culture technique with increased physiological relevance employing an advanced medium composition, reduced fetal calf serum, and spheroidal growth. Improved reflection of the transcriptional activity related to proliferation, hypoxia, and differentiation in EwS patient tumors was detected in EwS cells grown in this refined in vitro condition. Moreover, transcriptional signatures associated with the oncogenic activity of the EwS-specific FET::ETS fusion transcription factors in the refined culture condition were shifted from proliferative toward metabolic gene signatures. The herein-presented EwS cell culture technique with increased physiological relevance provides a broadly applicable approach for enhanced in vitro modeling relevant to advancing EwS research and the validity of experimental results.
Collapse
Affiliation(s)
- A Katharina Ceranski
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Martha J Carreño-Gonzalez
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Anna C Ehlers
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Kimberley M Hanssen
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Nadine Gmelin
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Florian H Geyer
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Zuzanna Kolodynska
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Endrit Vinca
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Tobias Faehling
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Philipp Poeller
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Shunya Ohmura
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Florencia Cidre-Aranaz
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Thomas G P Grünewald
- Hopp-Children's Cancer Center (KiTZ) Heidelberg, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
48
|
Barbera MC, Guarrera L, Re Cecconi AD, Cassanmagnago GA, Vallerga A, Lunardi M, Checchi F, Di Rito L, Romeo M, Mapelli SN, Schoser B, Generozov EV, Jansen R, de Geus EJC, Penninx B, van Dongen J, Craparotta I, Piccirillo R, Ahmetov II, Bolis M. Increased ectodysplasin-A2-receptor EDA2R is a ubiquitous hallmark of aging and mediates parainflammatory responses. Nat Commun 2025; 16:1898. [PMID: 39988718 PMCID: PMC11847917 DOI: 10.1038/s41467-025-56918-3] [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: 01/07/2024] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
Intensive efforts have been made to identify features that could serve as biomarkers of aging. Yet, drug-based interventions aimed at lessening the detrimental effects of getting older are lacking. This is largely attributable to tissue-specificity, sex-related differences, and to the difficulty of identifying actionable targets, which continues to pose a significant challenge. Here, we implement a bioinformatics approach revealing that aging-associated increase of the transmembrane Ectodysplasin-A2-Receptor is a prominent tissue-independent alteration occurring in humans and other species, and is particularly pronounced in models of accelerated aging. We show that strengthening of the Ectodysplasin-A2-Receptor signalling axis in myogenic precursors and differentiated myotubes suffices to trigger potent parainflammatory responses, mirroring aspects of aging-driven sarcopenia. Intriguingly, obesity, insulin-resistance, and aging-related comorbidities, such as type-2-diabetes, result in heightened levels of the Ectodysplasin-A2 ligand. Our findings suggest that targeting the Ectodysplasin-A2 surface receptor represents a promising pharmacological strategy to mitigate the development of aging-associated phenotypes.
Collapse
Affiliation(s)
- Maria Chiara Barbera
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Luca Guarrera
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Andrea David Re Cecconi
- Laboratory of Muscle Pathophysiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Giada Andrea Cassanmagnago
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
- Institute of Oncology Research, Bellinzona, Switzerland
- Università Della Svizzera Italiana (USI), Faculty of Biomedical Sciences, Bellinzona, Switzerland
| | - Arianna Vallerga
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Martina Lunardi
- Laboratory of Muscle Pathophysiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Francesca Checchi
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Laura Di Rito
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Margherita Romeo
- Laboratory of Human Pathology in Model Organism, Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Sarah Natalia Mapelli
- Department of Research in Inflammation and Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Benedikt Schoser
- Friedrich-Baur-Institute, Department of Neurology, LMU Klinikum, Ludwig-Maximilians University, Munich, Germany
| | - Edward V Generozov
- Department of Molecular Biology and Genetics, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Rick Jansen
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Brenda Penninx
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health, Mental Health Program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress Program, Amsterdam, The Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ilaria Craparotta
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Rosanna Piccirillo
- Laboratory of Muscle Pathophysiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy
| | - Ildus I Ahmetov
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, L3 5AF, UK
- Department of Physical Education, Plekhanov Russian University of Economics, Moscow, Russia
- Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, Russia
| | - Marco Bolis
- Computational Oncology Unit, Department of Oncology, Istituto di Ricerche Farmacologiche 'Mario Negri' IRCCS, Via Mario Negri 2, 20156, Milano, Italy.
- Institute of Oncology Research, Bellinzona, Switzerland.
- Università Della Svizzera Italiana (USI), Faculty of Biomedical Sciences, Bellinzona, Switzerland.
- Swiss Institute of Bioinformatics, Bioinformatics Core Unit, Bellinzona, TI 6500, Switzerland.
| |
Collapse
|
49
|
Yue X, Chen X, Zang Y, Wu J, Chen G, Tan H, Yang K. Bioinformatics analysis reveals key mechanisms of oligodendrocytes and oligodendrocyte precursor cells regulation in spinal cord Injury. Sci Rep 2025; 15:6400. [PMID: 39984610 PMCID: PMC11845783 DOI: 10.1038/s41598-025-90489-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: 10/11/2024] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
Despite extensive research, spinal cord injuries (SCI), which could cause severe sensory, motor and autonomic dysfunction, remain largely incurable. Oligodendrocytes and oligodendrocyte precursor cells (ODC/OPC) play a crucial role in neural morphological repair and functional recovery following SCI. We performed single-cell sequencing (scRNA-seq) on 59,558 cells from 39 mouse samples, combined with microarray data from 164 SCI samples and 3 uninjured samples. We further validated our findings using a large clinical cohort consisting of 38 SCI patients, 10 healthy controls, and 10 trauma controls, assessed with the American Spinal Cord Injury Association (ASIA) scale. We proposed a novel SCI classification model based on the expression of prognostic differentially expressed ODC/OPC differentiation-related genes (PDEODGs). This model includes three types: Low ODC/OPC Score Classification (LOSC), Median ODC/OPC Score Classification (MOSC), and High ODC/OPC Score Classification (HOSC). Considering the relationship between these subtypes and prognosis, we speculated that enhancing ODC/OPC differentiation and inhibiting inflammatory infiltration may improve outcomes. Additionally, we identified potential treatments for SCI that target key genes within these subtypes, offering promising implications for therapy.
Collapse
Affiliation(s)
- Xi Yue
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xunling Chen
- Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Yang Zang
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinliang Wu
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guanhao Chen
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Tan
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kerong Yang
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
50
|
Bouwman W, Raymakers R, van der Poll T, van de Stolpe A. Comparison Between Signal Transduction Pathway Activity in Blood Cells of Sepsis Patients and Laboratory Models. Cells 2025; 14:311. [PMID: 39996782 PMCID: PMC11854017 DOI: 10.3390/cells14040311] [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: 09/20/2024] [Revised: 10/18/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Sepsis represents a serious disease burden that lacks effective treatment. Drug development for sepsis requires laboratory models that adequately represent sepsis patients. Simultaneous Transcriptome-based Activity Profiling of Signal Transduction Pathway (STAP-STP) technology quantitatively infers STP activity from mRNA levels of target genes of the STP-associated transcription factor. Here, we used STAP-STP technology to compare STP activities between sepsis patients and lipopolysaccharide (LPS)-based models. Activity scores of Androgen Receptor (AR), TGFβ, NFκB, JAK-STAT1/2, and JAK-STAT3 STPs were calculated based on publicly available transcriptome data. Peripheral blood mononuclear cells (PBMCs) from patients with Gram-negative sepsis, nor PBMCs stimulated with LPS in vitro, showed altered STP activity. Increased NFκB, JAK-STAT1/2, and JAK-STAT3 STP activity was found in whole blood stimulated with LPS in vitro, and in whole blood obtained after intravenous injection of LPS in humans in vivo; AR and TGFβ STP activity only increased in the in vivo LPS model. These results resembled previously reported STP activity in whole blood of sepsis patients. We provide the first comparison of STP activity between patients with sepsis and laboratory model systems. Results are of use for the refinement of sepsis model systems for rational drug development.
Collapse
Affiliation(s)
- Wilbert Bouwman
- Center of Experimental and Molecular Medicine & Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | | - Tom van der Poll
- Center of Experimental and Molecular Medicine & Division of Infectious Diseases, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | | |
Collapse
|