1
|
Hong Y, Zhang X, Rong W, Hu C, Jiang Y, Xu J, Wen H, Feng F, Naman CB, Shen H, He S, Ding L, Cui W. Uncovering the therapeutic potentials of marine-derived natural compounds with small amounts for neurological disorders. Gene 2025; 957:149465. [PMID: 40189165 DOI: 10.1016/j.gene.2025.149465] [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: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
The discovery of novel drugs from the ocean is a relatively recent development. However, for most studies of marine drugs, the priority is the discovery of compounds with new structures. Normally, only small amounts (< 1 mg) of new compounds could be extracted from marine microbes, and it is difficult to evaluate the therapeutic potentials of these newly-identified marine-derived natural compounds by traditional cell- or animal-based phenotypic screenings. Genes play a crucial role in determining the phenotype of diseases and the action of drugs. By comparing genomic expression associated with disease conditions and compound treatments, it is possible to predict the potential of a certain compound to counteract a specific type of disease. In this study, marine-derived natural compounds-induced genomic changes in cells were collected either from public databases or by using RNA-seq analysis. The therapeutic potentials of representative marine-derived natural compounds, namely phycocyanobilin, cycloheximide and NBU-1, a newly-identified natural compound extracted from a marine sponge-associated Streptomyces, on bipolar disorder (BD), Parkinson's disease (PD) and Alzheimer's disease (AD), were predicted by gene set enrichment analysis (GSEA), respectively. The anti-neurological disorder activity of these marine-derived natural compounds were further validated in methamphetamine-induced rats mimicking manic phase of BD, 6-OHDA-treated PC12 cells mimicking PD neurotoxicity and β-amyloid oligomer-incubated SH-SY5Y cells mimicking AD neuronal loss. Our study provides not only new insights for pharmacological applications of the marine-derived natural compounds here studied, but also a method for predicting and evaluating therapeutic potentials of newly-identified marine-derived natural compounds with small quantities.
Collapse
Affiliation(s)
- Yirui Hong
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China
| | - Xinyu Zhang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China; College of Food Science and Engineering, Ningbo University, Zhejiang 315211, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China
| | - Wenni Rong
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China
| | - Chenwei Hu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China
| | - Yujie Jiang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China
| | - Jiayi Xu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China
| | - Huimin Wen
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China; College of Food Science and Engineering, Ningbo University, Zhejiang 315211, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China
| | - Fangjian Feng
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China; College of Food Science and Engineering, Ningbo University, Zhejiang 315211, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China
| | - C Benjamin Naman
- Department of Science and Conservation, San Diego Botanic Garden, CA 92024, USA
| | - Haowei Shen
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China
| | - Shan He
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China; Ningbo Institute of Marine Medicine, Peking University, Zhejiang 315800, China
| | - Lijian Ding
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China; College of Food Science and Engineering, Ningbo University, Zhejiang 315211, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China.
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Zhejiang 315211, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China; Institute of One Health Science (IOHS), Ningbo University, Zhejiang, 315211, China.
| |
Collapse
|
2
|
Shi J, Zhang M, Hu Y, Liu J, Li K, Sun X, Chen S, Liu J, Ye L, Fan J, Jia J. Differences in transcriptome characteristics and drug repositioning of Alzheimer's disease according to sex. Neurobiol Dis 2025; 210:106909. [PMID: 40220916 DOI: 10.1016/j.nbd.2025.106909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Previous studies have shown significant sex differences in AD with regarding its epidemiology, pathophysiology, clinical presentation, and treatment response. However, the transcriptome variances associated with sex in AD remain unclear. METHODS RNA sequencing (RNA-seq) and transcriptomic analyses were performed on peripheral blood samples from total of 54 patients, including male AD patients (n = 15), female AD patients (n = 10), male MCI patients (n = 7), female MCI patients (n = 11), male healthy controls (n = 6), female healthy controls (n = 5). The snRNA-seq dataset (GSE167494, GSE157827) of prefrontal cortex tissues was obtained from the Gene Expression Omnibus (GEO). We conducted an investigation into differentially expressed genes and pathways in the peripheral blood cells as well as prefrontal cortex tissues of both male and female AD patients with consideration to sex-related factors. Additionally, we analyzed the distribution and characteristics of cells in the cerebral cortex as well as the interaction and communication between cells of male and female AD patients. Connectivity Map (CMap) was utilized for predicting and screening potential sex-specific drugs for AD. RESULTS The transcriptome profile and associated biological processes in the peripheral blood of male and female AD and MCI patients exhibit discernible differences, including upregulation of BASP1 in AD male patients and arousing TNS1 in AD female patients. The distribution of various cell types in the prefrontal cortex tissues differs between male and female AD patients, like neuron and oligodendrocyte decreased and endothelial cell and astrocyte increased in female compared with male, while a multitude of genes exhibit significant differential expression. The results of cell communication analysis, such as collagen signaling pathway, suggest that sex disparities impact intercellular interactions within prefrontal cortex tissues among individuals with AD. By drug repositioning, several drugs, including torin-2 and YM-298198, might have the potential to therapeutic value of MCI or AD, while drugs like homoharringtonine and teniposide have potential opposite effects in different sexes. CONCLUSION The characteristics of the transcriptome in peripheral blood and single-cell transcriptome in the prefrontal cortex exhibit significant differences between male and female patients with AD, which providing a basis for future sex stratified treatment of AD.
Collapse
Affiliation(s)
- Jingqi Shi
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China
| | - Minghua Zhang
- Medical Supplies Center of PLA General Hospital, Beijing 100853, China
| | - Yazhuo Hu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China
| | - Jing Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China
| | - Ke Li
- Geriatric Neurological Department of the Second Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Xuan Sun
- Geriatric Neurological Department of the Second Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Siyu Chen
- Geriatric Neurological Department of the Second Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jianwei Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China
| | - Ling Ye
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China
| | - Jiao Fan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China.
| | - Jianjun Jia
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
3
|
Choi Y, Lee H, Beck BR, Lee B, Lee JH, Kim S, Chun SH, Won HS, Ko YH. Repurposing of the Syk inhibitor fostamatinib using a machine learning algorithm. Exp Ther Med 2025; 29:110. [PMID: 40242601 PMCID: PMC12001310 DOI: 10.3892/etm.2025.12860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 02/14/2025] [Indexed: 04/18/2025] Open
Abstract
TAM (TYRO3, AXL, MERTK) receptor tyrosine kinases (RTKs) have intrinsic roles in tumor cell proliferation, migration, chemoresistance, and suppression of antitumor immunity. The overexpression of TAM RTKs is associated with poor prognosis in various types of cancer. Single-target agents of TAM RTKs have limited efficacy because of an adaptive feedback mechanism resulting from the cooperation of TAM family members. This suggests that multiple targeting of members has the potential for a more potent anticancer effect. The present study used a deep-learning based drug-target interaction (DTI) prediction model called molecule transformer-DTI (MT-DTI) to identify commercially available drugs that may inhibit the three members of TAM RTKs. The results showed that fostamatinib, a spleen tyrosine kinase (Syk) inhibitor, could inhibit the three receptor kinases of the TAM family with an IC50 <1 µM. Notably, no other Syk inhibitors were predicted by the MT-DTI model. To verify this result, this study performed in vitro studies with various types of cancer cell lines. Consistent with the DTI results, this study observed that fostamatinib suppressed cell proliferation by inhibiting TAM RTKs, while other Syk inhibitors showed no inhibitory activity. These results suggest that fostamatinib could exhibit anticancer activity as a pan-TAM inhibitor. Taken together, these findings demonstrated that this artificial intelligence model could be effectively used for drug repurposing and repositioning. Furthermore, by identifying its novel mechanism of action, this study confirmed the potential for fostamatinib to expand its indications as a TAM inhibitor.
Collapse
Affiliation(s)
| | - Heejin Lee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Bo Ram Beck
- Deargen Inc., Daejeon 35220, Republic of Korea
| | - Bora Lee
- Deargen Inc., Daejeon 35220, Republic of Korea
| | - Ji Hyun Lee
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seoree Kim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang Hoon Chun
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hye Sung Won
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Yoon Ho Ko
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
4
|
Liu H, Lin S, Chen PX, Min J, Liu XY, Guan T, Yang CY, Xiao XJ, Xiong DH, Sun SJ, Nie L, Gong H, Wu XS, He XF, Liu J. Integrated bioinformatics analysis to develop diagnostic models for malignant transformation of chronic proliferative diseases. BLOOD SCIENCE 2025; 7:e00226. [PMID: 40201199 PMCID: PMC11977743 DOI: 10.1097/bs9.0000000000000226] [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: 11/05/2024] [Accepted: 02/16/2025] [Indexed: 04/10/2025] Open
Abstract
The combined analysis of dual diseases can provide new insights into pathogenic mechanisms, identify novel biomarkers, and develop targeted therapeutic strategies. Polycythemia vera (PV) is a chronic myeloproliferative neoplasm associated with a risk of acute myeloid leukemia (AML) transformation. However, the chronic nature of disease transformation complicates longitudinal high-throughput sequencing studies of patients with PV before and after AML transformation. This study aimed to develop a diagnostic model for malignant transformation of chronic proliferative diseases, addressing the challenges of early detection and intervention. Integrated public datasets of PV and AML were analyzed to identify differentially expressed genes (DEGs) and construct a weighted correlation network. Machine-learning algorithms screen genes for potential biomarkers, leading to the development of diagnostic models. Clinical specimens were collected to validate gene expression. cMAP and molecular docking predicted potential drugs. In vitro experiments were performed to assess drug efficacy in PV and AML cells. CIBERSORT and single-cell RNA-sequencing (scRNA-seq) analyses were used to explore the impact of hub genes on the tumor microenvironment. We identified 24 genes shared between PV and AML, which were enriched in immune-related pathways. Lactoferrin (LTF) and G protein-coupled receptor 65 (GPR65) were integrated into a nomogram with a robust predictive power. The predicted drug vemurafenib inhibited proliferation and increased apoptosis in PV and AML cells. TME analysis has linked these biomarkers to macrophages. Clinical samples were used to confirm LTF and GPR65 expression levels. We identified shared genes between PV and AML and developed a diagnostic nomogram that offers a novel avenue for the diagnosis and clinical management of AML-related PV.
Collapse
Affiliation(s)
- Hua Liu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Sheng Lin
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Pei-Xuan Chen
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Juan Min
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xia-Yang Liu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Ting Guan
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Chao-Ying Yang
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xiao-Juan Xiao
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - De-Hui Xiong
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Sheng-Jie Sun
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Ling Nie
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Han Gong
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xu-Sheng Wu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Xiao-Feng He
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| | - Jing Liu
- Shenzhen Health Development Research and Data Management Center, Shenzhen 518028, China
- Molecular Biology Research Center, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410013, China
| |
Collapse
|
5
|
Zhang J, Qin X, Qin Y, Qi F, Wang Y, Sun J, Yan L, Sun W, Guo X. Proteomic- and metabolomic-based mechanisms of androgen-mediated right ventricular maladaptive remodeling under pressure overload. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167768. [PMID: 40049298 DOI: 10.1016/j.bbadis.2025.167768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Right ventricular (RV) maladaptive remodeling has been demonstrated to be more severe in males than in females under similar afterload, with androgen potentially involved. However, the mechanism remains unknown. METHODS We performed RV proteomics and metabolomics in male and castrated rats with pulmonary artery banding (PAB) or sham surgery. The core pathway was tested in other sets of male, castrated male, and testosterone-replaced rats with and without pathway inhibitors administration and in RV remodeling patients. Metabolite verification was carried out by matching secondary spectra. RESULTS With the same extent of increases in RV afterload, male PAB rats exhibited more pronounced RV hypertrophy and fibrosis than castrated PAB rats (p < 0.05). The omics analysis indicated that pathways and functions related to oxidative stress were exhibited in the male group, with the platelet-derived growth factor (PDGF) pathway being among them. More proteins and metabolites associated with fatty acid metabolism were downregulated in males. Correlation analysis showed that PDGF receptor beta (PDGFRB) and signal transducer and activator of transcription 3 (STAT3) were negatively correlated with carnitine and reactive oxygen species scavenging metabolites only in male rats. The activation of the PDGF pathway was verified in testosterone-replaced PAB rats and male patients with RV remodeling. Treatments with PDGFRB inhibitor and STAT3 inhibitor could reverse RV maladaptive remodeling in male and testosterone-replaced PAB rats but not in castrated ones. CONCLUSIONS Androgen might exacerbate RV maladaptive remodeling via intensified oxidative stress and insufficient energy supply, with activating the PDGFRB-STAT3 signaling being one of the possible pathways.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Qi
- Proteomics Center, Core Facility of Instrument, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yufei Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiameng Sun
- Proteomics Center, Core Facility of Instrument, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Sun
- Proteomics Center, Core Facility of Instrument, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Hatoum AS, Gorelik AJ, Blaydon L, Huggett SB, Chi T, Baranger DAA, Miller AP, Johnson EC, Agrawal A, Bogdan R. Psychiatric genome-wide association study enrichment shows promise for future psychopharmaceutical discoveries. COMMUNICATIONS MEDICINE 2025; 5:176. [PMID: 40379965 DOI: 10.1038/s43856-025-00877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Innovation in psychiatric therapeutics has stagnated on known mechanisms. Psychiatric genome-wide association studies (GWAS) have identified hundreds of genome-wide significant (GWS) loci that have rapidly advanced our understanding of disease etiology. However, whether these results can be leveraged to improve clinical treatment for specific psychiatric disorders remains poorly understood. METHODS In this proof-of-principal evaluation of GWAS clinical utility, we test whether the targets of drugs used to treat Attention Deficit Hyperactivity Disorder (ADHD), Bipolar Disorder (BiP), Generalized Anxiety Disorder (GAD), Major Depressive Disorder (MDD), Post-Traumatic Stress Disorder (PTSD), Schizophrenia (SCZ), Substance Use Disorders (SUDs), and insomnia (INS), are enriched for GWAS meta-analysis findings. RESULTS The genes coding for treatment targets of medications used to SCZ, BiP, MDD, and SUDs (but not ADHD, PTSD, GAD, or INSOM) are enriched for GWS loci identified in their respective GWAS (ORs: 2.78-27.63; all ps <1.15e-3). Enrichment is largely driven by the presence of a GWS locus or loci within a gene coding for a drug target (i.e., proximity matching). Broadly, additional annotation (i.e., functional: Combined Annotation Dependent Depletion [CADD] scores, regulomeDB scores, eQTL, chromatin loop, and gene region; statistical: effect size of genome-wide significant SNPs; Z-score of SNPs; number of drug targets implicated by GWAS), with the exception of weighting by the largest SNP effect size, does not further improve enrichment across disorders. Evaluation of prior smaller GWAS reveal that more recent larger GWAS improve enrichment. CONCLUSIONS GWAS results may assist in the prioritization of medications for future psychopharmaceutical research.
Collapse
Affiliation(s)
- Alexander S Hatoum
- Washington University in St. Louis, Department of Psychological & Brain Sciences, St. Louis, MO, USA.
- Washington University School of Medicine, Department of Psychiatry, St. Louis, MO, USA.
- Washington University School of Medicine, AI and Health Institute, St. Louis, MO, USA.
| | - Aaron J Gorelik
- Washington University in St. Louis, Department of Psychological & Brain Sciences, St. Louis, MO, USA
| | - Lauren Blaydon
- Washington University in St. Louis, Department of Psychological & Brain Sciences, St. Louis, MO, USA
| | | | - Tingying Chi
- St. Louis Behavioral Medicine Institute, St. Louis, WA, USA
| | - David A A Baranger
- Washington University in St. Louis, Department of Psychological & Brain Sciences, St. Louis, MO, USA
- Washington University School of Medicine, Department of Psychiatry, St. Louis, MO, USA
| | - Alex P Miller
- Washington University School of Medicine, Department of Psychiatry, St. Louis, MO, USA
- Indiana University School of Medicine, Department of Psychiatry, Indianapolis, IN, USA
| | - Emma C Johnson
- Washington University School of Medicine, Department of Psychiatry, St. Louis, MO, USA
| | - Arpana Agrawal
- Washington University School of Medicine, Department of Psychiatry, St. Louis, MO, USA
- Washington University School of Medicine, AI and Health Institute, St. Louis, MO, USA
| | - Ryan Bogdan
- Washington University in St. Louis, Department of Psychological & Brain Sciences, St. Louis, MO, USA
- Washington University School of Medicine, AI and Health Institute, St. Louis, MO, USA
| |
Collapse
|
7
|
Zheng Q, Deng S, Chen X, Wang Y, Yang Y. Macrophage inhibition in the alleviation of nonalcoholic steatohepatitis caused by bariatric surgery. Genes Immun 2025:10.1038/s41435-025-00334-6. [PMID: 40374920 DOI: 10.1038/s41435-025-00334-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 04/21/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
The incidence of nonalcoholic steatohepatitis (NASH) is increasing worldwide, and effective treatment is urgently needed. To understand the molecular mechanisms behind the effectiveness of bariatric surgery in treating NASH, we integrated single-cell and bulk RNA sequencing data to identify the role of liver macrophage polarization in alleviating NASH and screen possible drugs for treatment. Analysis revealed that bariatric surgery alleviates NASH by inhibiting liver M1 macrophage polarization with 12 differentially expressed M1 macrophage-related genes. Additionally, 56 potentially effective drugs were predicted for NASH treatment. These findings shed light on the effectiveness of bariatric surgery in treating NASH and offer potential drug candidates for further exploration.
Collapse
Affiliation(s)
- Qianwen Zheng
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Shizhou Deng
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiyu Chen
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yayun Wang
- Specific Lab for Mitochondrial Plasticity Underlying Nervous System Diseases, National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yanling Yang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
8
|
Wiedmer T, Teoh ST, Christodoulaki E, Wolf G, Tian C, Sedlyarov V, Jarret A, Leippe P, Frommelt F, Ingles-Prieto A, Lindinger S, Barbosa BMG, Onstein S, Klimek C, Garcia J, Serrano I, Reil D, Santacruz D, Piotrowski M, Noell S, Bueschl C, Li H, Chi G, Mereiter S, Oliveira T, Penninger JM, Sauer DB, Steppan CM, Viollet C, Klavins K, Hannich JT, Goldmann U, Superti-Furga G. Metabolic mapping of the human solute carrier superfamily. Mol Syst Biol 2025:10.1038/s44320-025-00106-4. [PMID: 40355754 DOI: 10.1038/s44320-025-00106-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Solute carrier (SLC) transporters govern most of the chemical exchange across cellular membranes and are integral to metabolic regulation, which in turn is linked to cellular function and identity. Despite their key role, individual functions of the SLC superfamily members were not evaluated systematically. We determined the metabolic and transcriptional profiles upon SLC overexpression in knock-out or wild-type isogenic cell backgrounds for 378 SLCs and 441 SLCs, respectively. Targeted metabolomics provided a fingerprint of 189 intracellular metabolites, while transcriptomics offered insights into cellular programs modulated by SLC expression. Beyond the metabolic profiles of 102 SLCs directly related to their known substrates, we identified putative substrates or metabolic pathway connections for 71 SLCs without previously annotated bona fide substrates, including SLC45A4 as a new polyamine transporter. By comparing the molecular profiles, we identified functionally related SLC groups, including some with distinct impacts on osmolyte balancing and glycosylation. The assessment of functionally related human genes presented here may serve as a blueprint for other systematic studies and supports future investigations into the functional roles of SLCs.
Collapse
Affiliation(s)
- Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Shao Thing Teoh
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Eirini Christodoulaki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Chengzhe Tian
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Vitaly Sedlyarov
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Abigail Jarret
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Philipp Leippe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Fabian Frommelt
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Sabrina Lindinger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Barbara M G Barbosa
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Svenja Onstein
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Christoph Klimek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Julio Garcia
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Iciar Serrano
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Daniela Reil
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Diana Santacruz
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Mary Piotrowski
- Pfizer Worldwide Research and Development, Groton, CT, 06340, USA
| | - Stephen Noell
- Pfizer Worldwide Research and Development, Groton, CT, 06340, USA
| | - Christoph Bueschl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Huanyu Li
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gamma Chi
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Mereiter
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030, Vienna, Austria
| | - Tiago Oliveira
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030, Vienna, Austria
| | - Josef M Penninger
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), 1030, Vienna, Austria
- Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, V6T 1Z3, Vancouver, Canada
| | - David B Sauer
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Claire M Steppan
- Pfizer Worldwide Research and Development, Groton, CT, 06340, USA
| | - Coralie Viollet
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88400, Biberach, Germany
| | - Kristaps Klavins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - J Thomas Hannich
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Ulrich Goldmann
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria.
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria.
- Fondazione Ri.MED, Palermo, Italy.
| |
Collapse
|
9
|
Alishvandi A, Barancheshemeh M, Firuzpour F, Aram C, Kamali MJ, Keikha M. Decoding virulence and resistance in Klebsiella pneumoniae: Pharmacological insights, immunological dynamics, and in silico therapeutic strategies. Microb Pathog 2025; 205:107691. [PMID: 40355055 DOI: 10.1016/j.micpath.2025.107691] [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: 02/21/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Klebsiella pneumoniae (K. pneumoniae) has become a serious global health concern due to its rising virulence and antibiotic resistance. As one of the leading members of ESKAPE pathogens, it plays a major role in a wide range of infections that cause pneumonia, urinary tract infections, and bacteremia, especially in immunocompromised and hospitalized patients. The recent increase in multidrug-resistant (MDR) and hypervirulent (hvKP) strains due to the production of extended-spectrum beta-lactamases (ESBLs) and carbapenemases, has greatly limited therapeutic options that highlights the need for novel approaches to combat the pathogen. This review outlines the virulence mechanisms, profiles of antibiotic resistance, and immune evasion strategies in K. pneumoniae. Also, it points out the role of capsular polysaccharides, lipopolysaccharides, and fimbriae in host colonization and immune evasion. Additionally, the review discusses the emerging therapeutic strategies of vaccine development, computational drug discovery, and the use of artificial intelligence (AI). The progress achieved in reverse vaccinology and structural biology enables the identification of new drug and vaccine targets, whereas AI and machine learning (ML) stand out as powerful candidates for high-throughput screening and drug design. However, challenges with antigenic variability, safety, and the need to collaborate globally still exist. This review focuses on the need for interdisciplinary approaches involving molecular biology and immunology with computational sciences to address K. pneumoniae infections and provide appropriate therapies in the era of antibiotic resistance.
Collapse
Affiliation(s)
- Ali Alishvandi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran; Department of Immunology, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Faezeh Firuzpour
- Research Committee, Babol University of Medical Sciences, Babol, Iran; Cancer Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Cena Aram
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mohammad Javad Kamali
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Masoud Keikha
- Tropical and Communicable Diseases Research Center, Iranshahr University of Medical Sciences, Iranshahr, Iran; Department of Medical Microbiology, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| |
Collapse
|
10
|
Qiu Q, Yang Z, Zhao J, Zhang R, Zheng S, Wang C, Xu H, Deng H, Zhao K, Liu M. Integrative analysis of cuproptosis-related lncRNAs for prognostic risk assessment and tumor immune microenvironment evaluation in laryngeal squamous cell carcinoma. Int J Biol Macromol 2025; 306:141846. [PMID: 40058422 DOI: 10.1016/j.ijbiomac.2025.141846] [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: 07/17/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 05/11/2025]
Abstract
Long non-coding RNAs (lncRNAs) play a pivotal role in tumor prognostic models. This study targets cuproptosis-related lncRNAs in laryngeal squamous cell carcinoma (LSCC) using RNA-seq data from The Cancer Genome Atlas (TCGA)-LSCC. Differentially expressed genes were identified, and Pearson correlation analysis pinpointed cuproptosis-related lncRNAs. Combining clinical data, a prognostic risk model was constructed using LASSO Cox regression and Cox proportional hazards analyses. Nine lncRNAs (LINCO1473, SNHG12, AC007938.3, AC040970.1, AC023669.1, AL158166.2, GIHCG, AC007240.3, and AC011370.1) were identified, with GIHCG showing significant correlation with LSCC prognosis. GIHCG's competing endogenous RNAs (ceRNA) and co-expression networks (CEN) were established, revealing sensitivity to drugs like BMS-509744, YM155, and KU-55933. Silencing of GIHCG inhibited migration, invasion, EMT, and other biological processes in LSCC cells, suggesting GIHCG as a potential therapeutic target. Moreover, METTL16-mediated m6A methylation regulates GIHCG expression. In conclusion, this study successfully established a prognostic model comprising nine cuproptosis-related lncRNAs, accurately predicting LSCC prognosis, and highlighted the crucial role of GIHCG as a novel nucleic acid biomarker in regulating LSCC progression.
Collapse
Affiliation(s)
- Qibing Qiu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zhe Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jiandong Zhao
- Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, Beijing 100853, China
| | - Rongqi Zhang
- Beijing Institute of Technology, Beijing 100081, China
| | - Shikang Zheng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Cheng Wang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Haiming Xu
- Medical School of Chinese PLA, Beijing 100853, China
| | - Haihua Deng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China
| | - Kai Zhao
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China.
| | - Mingbo Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China; Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, Beijing 100853, China; Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya 572013, China.
| |
Collapse
|
11
|
Marino GB, Evangelista JE, Clarke DJB, Ma'ayan A. L2S2: chemical perturbation and CRISPR KO LINCS L1000 signature search engine. Nucleic Acids Res 2025:gkaf373. [PMID: 40308216 DOI: 10.1093/nar/gkaf373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/10/2025] [Accepted: 04/22/2025] [Indexed: 05/02/2025] Open
Abstract
As part of the Library of Integrated Network-Based Cellular Signatures (LINCS) NIH initiative, 248 human cell lines were profiled with the L1000 assay to measure the effect of 33 621 small molecules and 7508 single-gene CRISPR knockouts. From this massive dataset, we computed 1.678 million sets of up- and down-regulated genes. These gene sets are served for search by the LINCS L1000 Signature Search (L2S2) web server application. With L2S2, users can identify small molecules and single gene CRISPR KOs that produce gene expression profiles similar or opposite to their submitted single or up/down gene sets. L2S2 also includes a consensus search feature that ranks perturbations across all cellular contexts, time points, and concentrations. To demonstrate the utility of L2S2, we crossed the L2S2 gene sets with gene sets collected for the RummaGEO resource. The analysis identified clusters of differentially expressed genes that match drug classes, tissues, and diseases, pointing to many opportunities for drug repurposing and drug discovery. Overall, the L2S2 web server application can be used to further the development of personalized therapeutics while expanding our understanding of complex human diseases. The L2S2 web server application is available at https://l2s2.maayanlab.cloud.
Collapse
Affiliation(s)
- Giacomo B Marino
- Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - John E Evangelista
- Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Daniel J B Clarke
- Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
12
|
Qiu X, Huang W, Liang J, Chen H, Sha W, Lyu Y, Chen K, Yang H, Zhang Q. Predicting prognosis, immune landscape, and drug targets with a novel signature for hepatocellular carcinoma. Technol Health Care 2025; 33:1367-1380. [PMID: 40331560 DOI: 10.1177/09287329241296358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
BackgroundDespite advances in therapeutics, hepatocellular carcinoma (HCC) remains one of the most malignant types of digestive tract cancers with a poor prognosis. Pyroptosis is a form of programmed cell death induced by inflammatory caspases. Recent studies have identified pyroptosis, a form of programmed cell death induced by inflammatory caspases, as playing a role in tumorigenesis and cancer progression. However, the functions and mechanisms of pyroptosis in HCC are barely explored.MethodsGene expression and clinical data were derived from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. A prognostic signature and nomogram were constructed by on differentially expressed genes and clinical data. Pathway enrichment and immune cell infiltration were further analyzed. Potential drugs to modulate the pathways were explored.ResultsIn this study, a pyroptosis-related gene signature was developed and identified to be significantly correlated with the survival of HCC patients. Additionally, a nomogram on the basis of pyroptosis-related genes was constructed with distinct prognostic values. Furthermore, the pyroptosis-related gene signature might correlate with immune-related pathways and the regulation of the immune microenvironment, and several compounds (KIN001-220, TPCA-1, LY-303511, physostigmine, vemurafenib, etc.) could potentially reverse the pathogenic gene-expression patterns.Conclusions: Our study provides evidence that pyroptosis is involved in HCC development, progression and immune microenvironment, which is promising in predicting the prognosis and developing targeted therapy.
Collapse
Affiliation(s)
- Xinqi Qiu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Cancer Prevention Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
| | - Wentao Huang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| | - Jun Liang
- Geriatric Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510080, China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Yanlin Lyu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Kequan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510080, China
| | - Hongwei Yang
- Department of Medical Ultrasound, The First affiliated hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qingfang Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China
| |
Collapse
|
13
|
Liu Y, Sun W, Liu L, Cheng J, Li J, Huang Z, Ouyang M. Elevated KIF2C Expression Drives Osteosarcoma Progression by Modulating the Wnt/β-Catenin Signaling Pathway and Contributing to an Immunosuppressive Tumor Microenvironment. Cancer Med 2025; 14:e70915. [PMID: 40292920 PMCID: PMC12035763 DOI: 10.1002/cam4.70915] [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: 01/14/2025] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Although kinesin family member 2C (KIF2C) is implicated in various cancers, its role in osteosarcoma (OS) and the associated inflammatory microenvironment remains unclear. METHODS Publicly available datasets were analyzed to determine KIF2C expression, diagnostic value, and prognostic relevance in OS. In vitro (proliferation, colony formation, apoptosis, migration, invasion) and in vivo assays assessed its biological functions. KEGG enrichment and GSVA explored underlying pathways. ssGSEA, ESTIMATE algorithms, and single-cell sequencing evaluated the immune context, and molecular docking and molecular dynamics identified potential inhibitory compounds. RESULTS KIF2C was significantly overexpressed in OS, effectively distinguishing OS from normal tissues. Elevated KIF2C levels correlated with poor survival outcomes. Silencing KIF2C suppressed OS cell proliferation, migration, invasion, and in vivo tumor growth, while promoting apoptosis; conversely, overexpression of KIF2C had the opposite effect. Mechanistically, co-immunoprecipitation results indicated that KIF2C can bind to β-catenin to regulate the Wnt/β-catenin pathway. Furthermore, high KIF2C expression was associated with an immunosuppressive tumor microenvironment characterized by immune exhaustion. Molecular docking and molecular dynamics suggested butein as a candidate small-molecule inhibitor targeting KIF2C-related oncogenic mechanisms. CONCLUSION KIF2C drives OS progression by enhancing Wnt/β-catenin signaling and fostering an immunosuppressive microenvironment. Targeting KIF2C may offer new therapeutic approaches in managing OS.
Collapse
Affiliation(s)
- Ya‐Yun Liu
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Wu Sun
- Discipline of Chinese and Western Integrative MedicineJiangxi University of Chinese MedicineNanchangPeople's Republic of China
| | - Lin Liu
- Department of NursingJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Jin‐Hui Cheng
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Jing‐Tang Li
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Zu‐Tai Huang
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Min Ouyang
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| |
Collapse
|
14
|
Jiang J, Xu H, Liu M, Guo J, Li J, Li J, Bi H, Wang Y, Wang Z. AMPK in Chemoradiotherapy-Induced Oral Mucositis. J Oral Pathol Med 2025; 54:325-333. [PMID: 40091685 DOI: 10.1111/jop.13626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/25/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Oral mucositis (OM) is a prevalent adverse effect of radiotherapy and chemotherapy, significantly impacting cancer patients' well-being and potentially increasing mortality rates. Understanding OM's pathogenesis and identifying effective preventative and therapeutic agents are clinically crucial. METHODS This study analyzed RNA-Seq data from the GEO database, focusing on OM samples post-radiotherapy and chemotherapy. Differential gene expression analysis between OM and non-OM groups, followed by gene ontology (GO) enrichment analysis of differentially expressed genes (DEGs), was conducted. LASSO regression identified five potential biomarkers, and CIBERSORT assessed immune infiltration in OM samples. Correlations between biomarkers and immune infiltration were explored, and the connectivity map (CMAP) screened potential therapeutic drugs. The top 10 drugs were validated through molecular docking. RESULTS A total of 47 DEGs were identified, primarily involved in mitotic sister chromatid separation according to GO enrichment analysis. CIBERSORT analysis revealed significant changes in B cell naive and dendriform cells (DCs) resting content in the OM group. PRKAA2, encoding the AMP-activated protein kinase (AMPK) catalytic subunit, showed a negative correlation with DC resting content. Molecular docking from CMAP identified aloisine and teniposide as potential agents for OM induced by radiotherapy and chemotherapy. CONCLUSION AMPK emerges as a crucial regulator in OM post radiotherapy and chemotherapy, implicating sister chromatid separation, where DCs may play a pivotal role. Aloisine and Teniposide appear promising for OM prevention or treatment associated with these treatments.
Collapse
Affiliation(s)
- Junjie Jiang
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Hao Xu
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Mingyue Liu
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Jiwei Guo
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Jing Li
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Jianwen Li
- Department of Tumor Radiotherapy, WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Hengtai Bi
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Yousen Wang
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Zhiliang Wang
- WeiFang People's Hospital, Shandong Second Medical University, Weifang, China
| |
Collapse
|
15
|
Zhu F, Yan HHN, Tong Y, Zhao Y. Exploring WNT pathway dysregulation in serrated colorectal cancer for improved diagnostic and therapeutic strategies. Front Genet 2025; 16:1586867. [PMID: 40357363 PMCID: PMC12066562 DOI: 10.3389/fgene.2025.1586867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Background Serrated colorectal cancer (SCC) is a rare and aggressive subtype of colorectal cancer. Identifying SCC is crucial due to its high mortality rate and limited therapeutic options. Traditional methods to identify BRAF hotspot mutations and MLH1 methylation are insufficient in clinical practice. This study aims to explore the WNT pathway alterations in the CRC and to develop a WNT-derived subtyping model to identify SCC patients by using multi-OMICs data. Methods We included multi-omics data of 1751 colorectal cancer patients from the TCGA and GEO databases, and single-cell transcriptome data of 33 normal and cancer tissues from the SMC study cohort. The comprehensive study process incorporated unsupervised clustering, enrichment analysis, and statistical analysis. Results In this study, we investigated WNT pathway alterations in SCC by integrating both bulk and single-cell data into the multi-OMICs framework. The SCC subtype demonstrated significant WNT pathway heterogeneity and a more stable genomic structure. These findings support the development of a WNT-derived subtyping model that accurately identifies SCC patients across different CRC cohorts. In addition, the SCC subtype also presented a distinct immune microenvironment characterized by CD8+ T cell exhaustion. Finally, we utilized drug perturbation data to explore the potential drug targets for this severe cancer subtype. Conclusion We developed a WNT-derived subtyping method to identify SCC from canonical CRC, which enhances the molecular understanding of this severe cancer subtype and provides potential therapeutic strategies. Our findings suggest that SCC patients may benefit from the HSP90 inhibitor NVP-AUY922, highlighting its potential as a targeted therapy.
Collapse
Affiliation(s)
- Fengzhi Zhu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Helen Hoi Ning Yan
- Department of Pathology, School of Clinical Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Yin Tong
- Department of Pathology, School of Clinical Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Yueliang Zhao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Csendes G, Sanz G, Szalay KZ, Szalai B. Benchmarking foundation cell models for post-perturbation RNA-seq prediction. BMC Genomics 2025; 26:393. [PMID: 40269681 PMCID: PMC12016270 DOI: 10.1186/s12864-025-11600-2] [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: 12/08/2024] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
Accurately predicting cellular responses to perturbations is essential for understanding cell behaviour in both healthy and diseased states. While perturbation data is ideal for building such predictive models, its availability is considerably lower than baseline (non-perturbed) cellular data. To address this limitation, several foundation cell models have been developed using large-scale single-cell gene expression data. These models are fine-tuned after pre-training for specific tasks, such as predicting post-perturbation gene expression profiles, and are considered state-of-the-art for these problems. However, proper benchmarking of these models remains an unsolved challenge. In this study, we benchmarked two recently published foundation models, scGPT and scFoundation, against baseline models. Surprisingly, we found that even the simplest baseline model-taking the mean of training examples-outperformed scGPT and scFoundation. Furthermore, basic machine learning models that incorporate biologically meaningful features outperformed scGPT by a large margin. Additionally, we identified that the current Perturb-Seq benchmark datasets exhibit low perturbation-specific variance, making them suboptimal for evaluating such models. Our results highlight important limitations in current benchmarking approaches and provide insights into more effectively evaluating post-perturbation gene expression prediction models.
Collapse
|
17
|
Yu P, Feng S, Wang W, Cao W, Cao Y. Mechanism of Dahuang-Dangshen drug pairs in the treatment of HCC based on network pharmacology, bioinformatics, molecular docking and experimental verification. Med Oncol 2025; 42:174. [PMID: 40261593 DOI: 10.1007/s12032-025-02738-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
The anti-hepatocellular carcinoma (HCC) effect of the active ingredients of natural Chinese herbal medicine has become a hot topic at home and abroad. A list of studies evidence the therapeutic efficacy of Dahuang (DH) or Dangshen (DS) against HCC, but the combination effect of DH and DS in HCC treatment is rarely reported and the molecular mechanism of the drug pairs is not yet clear. Therefore, in this study, the combined effects and potential mechanisms of DH and DS drug pairs were investigated through network pharmacology, bioinformatics, molecular docking, and a series of pharmacological experiments, including the MTT assay, clone formation, wound healing, JC-1 staining, and western blotting. In total, 140 intersection targets between the DH-DS drug pairs and HCC were identified. In the PPI network, the top ten hub targets with the highest node connection values were VEGFA, AKT, CTNNB1, EGFR, TNF, CASP3, HRAS, SRC, JUN, and ESR1. GO functional and KEGG pathway enrichment analysis involved 289 biological processes, 33 cellular components, 57 molecular functions, and 143 signaling pathways. Bioinformatic analysis indicated that EGFR and AKT were promising candidate genes that can serve as diagnostic and prognostic biomarkers for HCC. β-sitosterol from the DH drug and luteolin from the DS drug were found as promising small molecules for HCC. The experimental results showed that the combination of β-sitosterol and luteolin was more potent in suppressing cell proliferation, migration and inducing cell apoptosis when compared to β-sitosterol or luteolin alone. The western blot and molecular docking studies demonstrated that the potential mechanism may be related to the EGFR/AKT signaling pathway. In summary, the combination of DH and DS may resist cell proliferation, migration and promote apoptosis activity through the EGFR/AKT signaling pathway, which provides new insights for further exploring plant extract treatment for HCC.
Collapse
Affiliation(s)
- Pan Yu
- The First Affiliated Hospital of Anhui, University of Science and Technology, Huainan, 232001, China.
- College of Public Health, Anhui University of Science and Technology, Hefei, 230000, China.
| | - Shuhui Feng
- College of Public Health, Anhui University of Science and Technology, Hefei, 230000, China
| | - Weifan Wang
- Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Products, College of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Weiya Cao
- College of Public Health, Anhui University of Science and Technology, Hefei, 230000, China
- Joint Research Center of Occupational Medicine and Health, Institute of Grand Health, Hefei Comprehensive National Science Center, Hefei, 230000, China
| | - Yongchang Cao
- College of Public Health, Anhui University of Science and Technology, Hefei, 230000, China
| |
Collapse
|
18
|
Song Y, Ren S, Wu S, Liu W, Hu C, Feng S, Chen X, Tu R, Gao F. Glucocorticoid promotes metastasis of colorectal cancer via co-regulation of glucocorticoid receptor and TET2. Int J Cancer 2025; 156:1572-1582. [PMID: 39661335 DOI: 10.1002/ijc.35285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Glucocorticoids (GCs), commonly used for anti-inflammatory and cancer treatments, have been linked to the promotion of cancer metastasis. Yet, the molecular mechanisms behind this potential remain poorly understood. Clarifying these mechanisms is crucial for a nuanced understanding and potential refinement of GC therapies in the context of cancer treatment. In HEK293T cells, co-immunoprecipitation (Co-IP) and chromatin immunoprecipitation sequencing (ChIP-seq) were used with antibodies of glucocorticoid receptor (GR) and ten-eleven translocation enzymes (TET) family proteins (TET1, TET2, TET3). Drug repositioning was performed through the Connectivity Map database, using common target genes of GR and TET2 in HEK293 and HCT116 cell lines and differentially expressed genes (DEGs) of colorectal cancer (CRC). Cell migration and invasion were tested in CRC cell lines with varying GR expression, that is, HCT116 and HT29 cell lines. Dexamethasone (Dex) treatment resulted in a significant difference in cell migration rates in two CRC cell lines with disparate GR expression levels. Co-IP and ChIP-seq analyses substantiated the interaction between GR and TET family proteins in HEK293T cells. Belinostat, the selected compound, was successfully validated for its potential to counteract the effects of GC-induced invasion in CRC cells in vitro. Transcriptomic analyses of Belinostat-treated HCT116 cells revealed down-regulation of target genes associated with cancer metastasis. This study provides valuable insights into the molecular mechanisms underlying GC-induced metastasis, introducing newly repositioned compounds that could serve as potential adjuvant therapy to GC treatment. Furthermore, it opens avenues for exploring novel drug candidates for CRC treatment.
Collapse
Affiliation(s)
- Yanwei Song
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Shuqiang Ren
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Shumei Wu
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | | | - Chenghao Hu
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Siting Feng
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xinyu Chen
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Rui Tu
- E-GENE Co., Ltd, Shenzhen, China
| | - Fei Gao
- HIM-BGI Omics Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, China
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Kołodziejczak-Guglas I, Simões RLS, de Souza Santos E, Demicco EG, Lazcano Segura RN, Ma W, Wang P, Geffen Y, Storrs E, Petralia F, Colaprico A, da Veiga Leprevost F, Pugliese P, Ceccarelli M, Noushmehr H, Nesvizhskii AI, Kamińska B, Priebe W, Lubiński J, Zhang B, Lazar AJ, Kurzawa P, Mesri M, Robles AI, Ding L, Malta TM, Wiznerowicz M. Proteomic-based stemness score measures oncogenic dedifferentiation and enables the identification of druggable targets. CELL GENOMICS 2025:100851. [PMID: 40250426 DOI: 10.1016/j.xgen.2025.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/13/2024] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Cancer progression and therapeutic resistance are closely linked to a stemness phenotype. Here, we introduce a protein-expression-based stemness index (PROTsi) to evaluate oncogenic dedifferentiation in relation to histopathology, molecular features, and clinical outcomes. Utilizing datasets from the Clinical Proteomic Tumor Analysis Consortium across 11 tumor types, we validate PROTsi's effectiveness in accurately quantifying stem-like features. Through integration of PROTsi with multi-omics, including protein post-translational modifications, we identify molecular features associated with stemness and proteins that act as active nodes within transcriptional networks, driving tumor aggressiveness. Proteins highly correlated with stemness were identified as potential drug targets, both shared and tumor specific. These stemness-associated proteins demonstrate predictive value for clinical outcomes, as confirmed by immunohistochemistry in multiple samples. The findings emphasize PROTsi's efficacy as a valuable tool for selecting predictive protein targets, a crucial step in customizing anti-cancer therapy and advancing the clinical development of cures for cancer patients.
Collapse
Affiliation(s)
- Iga Kołodziejczak-Guglas
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Renan L S Simões
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Emerson de Souza Santos
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON M5G 1X5, Canada
| | - Rossana N Lazcano Segura
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yifat Geffen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antonio Colaprico
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Pietro Pugliese
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Houtan Noushmehr
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, MI 48202, USA
| | - Alexey I Nesvizhskii
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland
| | - Bing Zhang
- Lester and Sue Smith Breast Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paweł Kurzawa
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-514 Poznań, Poland
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Li Ding
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tathiane M Malta
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil.
| | - Maciej Wiznerowicz
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Department of Oncology, Institute of Oncology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-659 Poznań, Poland.
| |
Collapse
|
20
|
Yates J, Van Allen EM. New horizons at the interface of artificial intelligence and translational cancer research. Cancer Cell 2025; 43:708-727. [PMID: 40233719 PMCID: PMC12007700 DOI: 10.1016/j.ccell.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
Artificial intelligence (AI) is increasingly being utilized in cancer research as a computational strategy for analyzing multiomics datasets. Advances in single-cell and spatial profiling technologies have contributed significantly to our understanding of tumor biology, and AI methodologies are now being applied to accelerate translational efforts, including target discovery, biomarker identification, patient stratification, and therapeutic response prediction. Despite these advancements, the integration of AI into clinical workflows remains limited, presenting both challenges and opportunities. This review discusses AI applications in multiomics analysis and translational oncology, emphasizing their role in advancing biological discoveries and informing clinical decision-making. Key areas of focus include cellular heterogeneity, tumor microenvironment interactions, and AI-aided diagnostics. Challenges such as reproducibility, interpretability of AI models, and clinical integration are explored, with attention to strategies for addressing these hurdles. Together, these developments underscore the potential of AI and multiomics to enhance precision oncology and contribute to advancements in cancer care.
Collapse
Affiliation(s)
- Josephine Yates
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland; ETH AI Center, ETH Zurich, Zurich, Switzerland; Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard University, Boston, MA, USA; Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
21
|
Killick R, Hooper C, Fernandes C, Elliott C, Aarsland D, Kjosavik SR, Østerhus R, Williams G. Transcription-Driven Repurposing of Cardiotonic Steroids for Lithium Treatment of Severe Depression. Cells 2025; 14:575. [PMID: 40277900 PMCID: PMC12025515 DOI: 10.3390/cells14080575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Lithium is prescribed as a mood stabilizer for bipolar disorder and severe depression. However, the mechanism of action of lithium is unknown and there are major side effects associated with prolonged medication. This motivates a search for safer alternative drug repurposing candidates. Given that the drug mechanism may be encoded in transcriptional changes, we generated the gene expression profile for acute lithium treatment of cortical neuronal cultures. We found that the lithium-associated transcription response harbors a significant component that is the reverse of that seen in human brain samples from patients with major depression, bipolar disorder, and a mouse model of depression. Interrogating publicly available drug-driven expression data, we found that cardiotonic steroids drive gene expression in a correlated manner to our acute lithium profile. An analysis of the psychiatric medication cohort of the Norwegian Prescription Database showed that cardiotonic prescription is associated with a lower incidence of lithium prescription. Our transcriptional and epidemiological observations point towards cardiotonic steroids as possible repurposing candidates for lithium. These observations motivate a controlled trial to establish a causal connection and genuine therapeutic benefit in the context of depression.
Collapse
Affiliation(s)
- Richard Killick
- Centre for Healthy Brain Aging, IoPPN, King’s College London, London SE5 9RT, UK
| | - Claudie Hooper
- IHU HealthAge, Gérontopôle, Department of Geriatrics, CHU Toulouse, 31059 Toulouse, France
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King’s College London, London SE5 8AF, UK
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King’s College London, London SE1 1UL, UK
| | - Christina Elliott
- Faculty of Medical Sciences, School of Biomedical, Nutritional and Sport Sciences, Newcastle University, Newcastle NE4 5TG, UK
| | - Dag Aarsland
- Centre for Healthy Brain Aging, IoPPN, King’s College London, London SE5 9RT, UK
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, 4011 Stavanger, Norway
| | - Svein R. Kjosavik
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, 4011 Stavanger, Norway
- General Practice and Care Coordination Research Group, Stavanger University Hospital, 4011 Stavanger, Norway
| | - Ragnhild Østerhus
- Centre for Age-Related Medicine (SESAM), Stavanger University Hospital, 4011 Stavanger, Norway
| | - Gareth Williams
- Wolfson SPaRC, IoPPN, King’s College London, London SE1 1UL, UK
| |
Collapse
|
22
|
Zhang X, Hong Y, Hu C, Zhai Y, Pan N, Ding L, Han W, Cui W. Chryxanthone A, an extracted substance from endophytic fungal Aspergillus versicolor, produces anti-oxidant neuroprotection possibly via the action on mTOR/CREB axis. Gene 2025; 944:149298. [PMID: 39884402 DOI: 10.1016/j.gene.2025.149298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Neurons are susceptible to oxidative stress due to the elevated reactive oxygen species (ROS) production and the limited antioxidant defense mechanisms. Therefore, it is possible to treat oxidative stress-related neurological disorders via the inhibition of oxidative stress. Chryxanthone A is an extracted substance derived from the endophytic fungal Aspergillus versicolor, with an atypical dihydropyran ring. However, it is unknown whether and how chryxanthone A could produce anti-oxidant protection. PURPOSES The activity and mechanisms underlying the anti-oxidant protection of chryxanthone A were explored in the study. STUDY DESIGN AND METHODS HT22 neuronal cells were used to evaluate the anti-oxidant protection of chryxanthone A. Comprehensive bioinformatic methods, including RNA-seq analysis, transcription factor prediction, CMap prediction and molecular docking analysis, were utilized to explore the molecular mechanisms how chryxanthone A prevented oxidative stress, which was confirmed by Western blotting analysis. RESULTS Chryxanthone A concentration-dependently prevented H2O2-induced cell death and increase in intracellular ROS in HT22 cells. Results from RNA-seq and bioinformatic analysis indicated that chryxanthone A might act on mTOR/CREB axis, possibly via binding to the Val2227 site within ATP binding pocket of mTOR. The action of chryxanthone A on H2O2-induced alteration of mTOR/CREB axis were further confirmed in HT22 cells. CONCLUSION These results suggested that chryxanthone A produced anti-oxidant protection via the action on mTOR/CREB axis, providing a support that chryxanthone A might be developed as a novel drug candidate for the treatment of oxidative stress-related disorders.
Collapse
Affiliation(s)
- Xinyu Zhang
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China; Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China; Department of Marine Pharmacy, Ningbo University, Zhejiang 315211, China
| | - Yirui Hong
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China
| | - Chenwei Hu
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China
| | - Yijie Zhai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100 Shaanxi, China
| | - Nanyi Pan
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China
| | - Lijian Ding
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Zhejiang 315211, China; Department of Marine Pharmacy, Ningbo University, Zhejiang 315211, China
| | - Wenbo Han
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100 Shaanxi, China.
| | - Wei Cui
- Translational Medicine Center of Pain, Emotion and Cognition, Health Science Center, Ningbo University, Ningbo 315211 Zhejiang, China.
| |
Collapse
|
23
|
Dolivo DM, Rodrigues AE, Mustoe TA, Galiano RD, Hong SJ. Ethacrynic Acid Mitigates Skin Fibrosis through Downregulation of S100 Family Damage-Associated Molecular Pattern Expression in the Epidermis. J Invest Dermatol 2025:S0022-202X(25)00396-3. [PMID: 40210113 DOI: 10.1016/j.jid.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/07/2025] [Accepted: 03/23/2025] [Indexed: 04/12/2025]
Affiliation(s)
- David M Dolivo
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Adrian E Rodrigues
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Thomas A Mustoe
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert D Galiano
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Seok Jong Hong
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
24
|
Xu Q, Ma L, Streuer A, Altrock E, Schmitt N, Rapp F, Klär A, Nowak V, Obländer J, Weimer N, Palme I, Göl M, Zhu HH, Hofmann WK, Nowak D, Riabov V. Machine learning-based in-silico analysis identifies signatures of lysyl oxidases for prognostic and therapeutic response prediction in cancer. Cell Commun Signal 2025; 23:169. [PMID: 40186284 PMCID: PMC11971788 DOI: 10.1186/s12964-025-02176-1] [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: 09/25/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Lysyl oxidases (LOX/LOXL1-4) are crucial for cancer progression, yet their transcriptional regulation, potential therapeutic targeting, prognostic value and involvement in immune regulation remain poorly understood. This study comprehensively evaluates LOX/LOXL expression in cancer and highlights cancer types where targeting these enzymes and developing LOX/LOXL-based prognostic models could have significant clinical relevance. METHODS We assessed the association of LOX/LOXL expression with survival and drug sensitivity via analyzing public datasets (including bulk and single-cell RNA sequencing data of six datasets from Gene Expression Omnibus (GEO), Chinese Glioma Genome Atlas (CGGA) and Cancer Genome Atlas Program (TCGA)). We performed comprehensive machine learning-based bioinformatics analyses, including unsupervised consensus clustering, a total of 10 machine-learning algorithms for prognostic prediction and the Connectivity map tool for drug sensitivity prediction. RESULTS The clinical significance of the LOX/LOXL family was evaluated across 33 cancer types. Overexpression of LOX/LOXL showed a strong correlation with tumor progression and poor survival, particularly in glioma. Therefore, we developed a novel prognostic model for glioma by integrating LOX/LOXL expression and its co-expressed genes. This model was highly predictive for overall survival in glioma patients, indicating significant clinical utility in prognostic assessment. Furthermore, our analysis uncovered a distinct LOXL2-overexpressing malignant cell population in recurrent glioma, characterized by activation of collagen, laminin, and semaphorin-3 pathways, along with enhanced epithelial-mesenchymal transition. Apart from glioma, our data revealed the role of LOXL3 overexpression in macrophages and in predicting the response to immune checkpoint blockade in bladder and renal cancers. Given the pro-tumor role of LOX/LOXL genes in most analyzed cancers, we identified potential therapeutic compounds, such as the VEGFR inhibitor cediranib, to target pan-LOX/LOXL overexpression in cancer. CONCLUSIONS Our study provides novel insights into the potential value of LOX/LOXL in cancer pathogenesis and treatment, and particularly its prognostic significance in glioma.
Collapse
Affiliation(s)
- Qingyu Xu
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany.
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Ling Ma
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Alexander Streuer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Eva Altrock
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Nanni Schmitt
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Felicitas Rapp
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Alessa Klär
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Julia Obländer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Nadine Weimer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Iris Palme
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Melda Göl
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Hong-Hu Zhu
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Chinese Institutes for Medical Research, Beijing, China
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| |
Collapse
|
25
|
Wei W, Zhang Z, Li B, Fu Z, Liu J. Deciphering the role of lncRNA-mediated ceRNA network in disuse osteoporosis: insights from bone marrow mesenchymal stem cells under simulated microgravity. Front Med (Lausanne) 2025; 12:1444165. [PMID: 40248073 PMCID: PMC12003301 DOI: 10.3389/fmed.2025.1444165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Background Disuse osteoporosis (DOP) poses a significant health risk during extended space missions. Although the importance of long non-coding RNA (lncRNA) in bone marrow mesenchymal stem cells (BMSCs) and orthopedic diseases is recognized, the precise mechanism by which lncRNAs contribute to DOP remains elusive. This research aims to elucidate the potential regulatory role of lncRNAs in DOP. Methods Sequencing data were obtained from Gene Expression Omnibus (GEO) datasets, including coding and non-coding RNAs. Positive co-expression pairs of lncRNA-mRNA were identified using weighted gene co-expression network analysis, while miRNA-mRNA expression pairs were derived from the prediction database. A mRNA-miRNA-lncRNA network was established according to the shared mRNA. Functional enrichment analysis was conducted for the shared mRNAs using genome ontology and KEGG pathways. Hub genes were identified through protein-protein interaction analysis, and connectivity map analysis was employed to identify potential therapeutic agents for DOP. Results Integration of 74 lncRNAs, 19 miRNAs, and 200 mRNAs yielded a comprehensive mRNA-miRNA-lncRNA network. Enrichment analysis highlighted endoplasmic reticulum stress and extracellular matrix (ECM) pathways as significant in the ceRNA network. PPI analysis revealed three hub genes (COL4A1, LAMC1, and LAMA4) and identified five lncRNA-miRNA-hub gene regulatory axes. Furthermore, three potential therapeutic compounds (SB-216763, oxymetholone, and flubendazole) for DOP were identified. Conclusion This study sheds light on the involvement of lncRNAs in the pathogenesis and treatment of DOP through the construction of a ceRNA network, linking protein-coding mRNA functions with non-coding RNAs.
Collapse
Affiliation(s)
- Wuzeng Wei
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Zhongli Zhang
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Bing Li
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Zhe Fu
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Jun Liu
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Zhou D, Cui Y, Liang T, Wu Z, Yan H, Li Y, Yin W, Lin Y, You Q. Pan-cancer analysis identifies CLEC12A as a potential biomarker and therapeutic target for lung adenocarcinoma. Cancer Cell Int 2025; 25:128. [PMID: 40181336 PMCID: PMC11967068 DOI: 10.1186/s12935-025-03755-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
C-type lectin domain family 12 member A (CLEC12A) is a type II transmembrane glycoprotein widely expressed in innate immune cells, where it plays a crucial role in immune modulation and has been implicated in cancer progression. However, its precise function in oncogenesis and immune infiltration remains incompletely understood. To investigate this, we utilized multiple databases to assess the mRNA and protein expression levels of CLEC12A across normal tissues and a broad spectrum of cancers. We also evaluated its prognostic and diagnostic significance in pan-cancer contexts. Furthermore, the relationship between CLEC12A expression and immune cell infiltration, immune checkpoints, and immune predictors was explored. In addition, Weighted Gene Co-Expression Network Analysis (WGCNA) and differential expression analysis were performed to examine the biological relevance of CLEC12A in lung adenocarcinoma (LUAD). We also leveraged various databases to predict CLEC12A's response to immunotherapy and drug sensitivity. Finally, in vitro experiments validated the functional role of CLEC12A in LUAD. Our comprehensive pan-cancer analysis revealed that CLEC12A exhibited distinct expression patterns across different cancer types, suggesting its potential as both a diagnostic and prognostic biomarker. Notably, CLEC12A expression was strongly correlated with immune cell infiltration, immune checkpoints, and immune predictors. Functional enrichment analysis highlighted that increased CLEC12A expression in LUAD was associated with a variety of immune-related biological processes and pathways. Moreover, CLEC12A showed significant predictive value for immunotherapy outcomes, and several drugs targeting CLEC12A were identified. In vitro experiments further demonstrated that CLEC12A overexpression inhibited the proliferation, migration, and invasion of LUAD cells. Taken together, our findings position CLEC12A as a promising candidate for cancer detection, prognosis, and as a therapeutic target, particularly in LUAD, where it may serve as a potential target for both immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Desheng Zhou
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yachao Cui
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tianxiang Liang
- The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zhenpeng Wu
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haiping Yan
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yingchang Li
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenguang Yin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China.
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China.
| | - Yunen Lin
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Qiang You
- Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China.
- Department of Biotherapy, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
27
|
Zhong Y, Zhang W, Zheng C, Wu H, Luo J, Yuan Z, Zhang H, Wang C, Feng H, Wang M, Zhang Q, Ju H, Wang G. Multi-omic analyses reveal PTPN6's impact on tumor immunity across various cancers. Sci Rep 2025; 15:11025. [PMID: 40164665 PMCID: PMC11958644 DOI: 10.1038/s41598-025-96302-1] [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: 01/02/2024] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6) plays a crucial regulatory role in cellular processes and has been implicated in oncogenesis. This pan-cancer analysis aimed to elucidate PTPN6's involvement across various cancer types, with a particular emphasis on its association with tumor immunity. We analyzed PTPN6 expression data from open access databases using various statistical techniques, including survival analysis, genetic heterogeneity analysis, immune profiling, single-cell analysis, drug sensitivity analysis, and protein interaction analysis. We also conducted in vitro experiments utilizing colorectal cancer cell lines to validate PTPN6's functional role. PTPN6 exhibited distinct expression patterns across cancers, and its prognostic significance was apparent in several cancer types, particularly in glioblastoma, sarcoma, and melanoma. We observed correlations between PTPN6 and immune genes/cell infiltration in these cancers, suggesting a potential role in modulating the tumor immune microenvironment. Single-cell analysis revealed that PTPN6 is predominantly localized in macrophages, B cells, and dendritic cells within the tumor microenvironment, implying its involvement in regulating immune cell function. Enrichment analysis highlighted PTPN6's role in immune-related pathways. Drug sensitivity analysis identified specific drugs, including PAC-1, SNX-2112, BELINOSTAT, VORINOSTAT, TPCA-1, and PHA-893,888, whose efficacy may be influenced by PTPN6 expression. Knocking down PTPN6 expression inhibited the proliferation and migration of colorectal cancer cells in vitro, confirming its oncogenic role in this cancer type. This pan-cancer analysis establishes PTPN6's multifaceted influence on tumor immunity and its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yuchen Zhong
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Weiyuan Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chaojing Zheng
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Hongyu Wu
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Jun Luo
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Ziming Yuan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Hao Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chunlin Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Haiyang Feng
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Meng Wang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Qian Zhang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Haixing Ju
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
28
|
Xu H, Li K, Liang X, Wang Z, Yang B. Multi-omics analysis to explore the molecular mechanisms related to keloid. Burns 2025; 51:107396. [PMID: 39874886 DOI: 10.1016/j.burns.2025.107396] [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: 07/10/2024] [Revised: 12/14/2024] [Accepted: 01/18/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Keloid is a benign skin tumor that result from abnormal wound healing and excessive collagen deposition. The pathogenesis is believed to be linked to genetic predisposition and immune imbalance, although the precise mechanisms remain poorly understood. Current therapeutic approaches may not consistently yield satisfactory outcomes and are often accompanied by potential side effects and risks. The high recurrence rate and refractory nature of keloid nodules present significant challenges and uncertainties in their management. Given the lack of effective treatment strategies, it is essential to identify key molecular pathways and potential therapeutic targets for keloid. OBJECTIVE This study aimed to identify the potential pathogenic mechanisms, hub genes, and immune cell involvement in keloid formation, with the goal of providing novel insights for targeted therapies. METHODS We utilized a combination of bulk RNA sequencing to analyze gene expression profiles in keloid tissues. Differentially expressed genes (DEGs) were identified and subjected to pathway enrichment analysis to reveal key biological processes involved in keloid pathogenesis. Mendelian randomization was performed to investigate the causal relationship between genetic factors and keloid formation, identifying potential hub genes. Immune infiltration analysis was conducted to determine the role of specific immune cells in keloid development. Subsequently, Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were performed to investigate the functional pathways associated with the hub genes. Network analysis was employed to identify transcription factors, miRNAs, and potential drugs in the Connectivity Map associated with the hub genes. Single-cell RNA sequencing was also used to identify cell-specific expression patterns of these genes. RESULTS Pathway enrichment analysis highlighted the association of keloid pathogenesis with cell proliferation and division, providing insights into the molecular processes involved. Mendelian randomization revealed that DUSP1 acts as an inhibitor of keloid formation, while HOXA5 promotes keloid pathogenesis. Immune infiltration analysis suggested that mast cells and macrophages play critical roles in the disease's progression. Based on hub gene analysis, the IL17 signaling pathway emerged as a key pathway implicated in keloid development. Further drug prediction models identified 9-methyl-5H-6-thia-4, 5-diaza-chrysene-6, 6-dioxide, zebularine, temozolomide and valproic acid targeting these hub genes. CONCLUSION DUSP1 and HOXA5 are hub genes in keloid pathogenesis, with DUSP1 acting as an inhibitor and HOXA5 as a promoter of disease progression. Targeting the regulatory networks associated with these genes could provide novel therapeutic strategies. Mast cells and macrophages are identified as critical immune cell types involved in the disease process. Additionally, the IL17 signaling pathway plays a crucial role in keloid development, highlighting its potential as a therapeutic target. These findings suggest that a multi-target approach focusing on these pathways could offer effective treatment options for keloid patients.
Collapse
Affiliation(s)
- Hailin Xu
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Keai Li
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xiaofeng Liang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyong Wang
- Department of Joint Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Duobao Road No.63, Liwan District, Guangzhou, Guangdong 510150, China.
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
29
|
Su H, Zhou X, Lin G, Luo C, Meng W, Lv C, Chen Y, Wen Z, Li X, Wu Y, Xiao C, Yang J, Lu J, Luo X, Chen Y, Tam PKH, Li C, Sun H, Pan X. Deciphering the Oncogenic Landscape of Hepatocytes Through Integrated Single-Nucleus and Bulk RNA-Seq of Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412944. [PMID: 39960344 PMCID: PMC11984907 DOI: 10.1002/advs.202412944] [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: 10/14/2024] [Revised: 01/01/2025] [Indexed: 04/12/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality, while the hepatocyte mechanisms driving oncogenesis remains poorly understood. In this study, single-nucleus RNA sequencing of samples from 22 HCC patients revealed 10 distinct hepatocyte subtypes, including beneficial Hep0, predominantly malignant Hep2, and immunosuppressive Hep9. These subtypes were strongly associated with patient prognosis, confirmed in TCGA-LIHC and Fudan HCC cohorts through hepatocyte composition deconvolution. A quantile-based scoring method is developed to integrate data from 29 public HCC datasets, creating a Quantile Distribution Model (QDM) with excellent diagnostic accuracy (Area Under the Curve, AUC = 0.968-0.982). QDM was employed to screen potential biomarkers, revealing that PDE7B functions as a key gene whose suppression promotes HCC progression. Guided by the genes specific to Hep0/2/9 subtypes, HCC is categorized into metabolic, inflammatory, and matrix classes, which are distinguishable in gene mutation frequencies, survival times, enriched pathways, and immune infiltration. Meanwhile, the sensitive drugs of the three HCC classes are identified, namely ouabain, teniposide, and TG-101348. This study presents the largest single-cell hepatocyte dataset to date, offering transformative insights into hepatocarcinogenesis and a comprehensive framework for advancing HCC diagnostics, prognostics, and personalized treatment strategies.
Collapse
Affiliation(s)
- Huanhou Su
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Xuewen Zhou
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Guanchuan Lin
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Chaochao Luo
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- College of Life SciencesShihezi UniversityShiheziXinjiang832003China
| | - Wei Meng
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Cui Lv
- Clinical Biobank CenterMicrobiome Medicine CenterDepartment of Laboratory MedicineGuangdong Provincial Clinical Research Center for Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Yuting Chen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Zebin Wen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Xu Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Yongzhang Wu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Changtai Xiao
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Jian Yang
- Department of Hepatobiliary Surgery IGeneral Surgery Center and Guangdong Provincial Clinical and Engineering Center of Digital MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Jiameng Lu
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Xingguang Luo
- Department of PsychiatryYale University School of MedicineNew HavenCT06510USA
| | - Yan Chen
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Paul KH Tam
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Chuanjiang Li
- Division of Hepatobiliopancreatic SurgeryDepartment of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Haitao Sun
- Clinical Biobank CenterMicrobiome Medicine CenterDepartment of Laboratory MedicineGuangdong Provincial Clinical Research Center for Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Xinghua Pan
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
- Key Laboratory of Infectious Diseases Research in South China (China Ministry Education)Southern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
30
|
Kort EJ, Sayed N, Liu C, Mondéjar-Parreño G, Forsberg J, Eugster E, Wu SM, Wu JC, Jovinge S. Olmesartan Restores LMNA Function in Haploinsufficient Cardiomyocytes. Circulation 2025; 151:1436-1448. [PMID: 40166828 PMCID: PMC12084018 DOI: 10.1161/circulationaha.121.058621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Gene mutations are responsible for a sizeable proportion of cases of heart failure. However, the number of patients with any specific mutation is small. Repositioning of existing US Food and Drug Administration-approved compounds to target specific mutations is a promising approach to efficient identification of new therapies for these patients. METHODS The National Institutes of Health Library of Integrated Network-Based Cellular Signatures database was interrogated to identify US Food and Drug Administration-approved compounds that demonstrated the ability to reverse the transcriptional effects of LMNA knockdown. Top hits from this screening were validated in vitro with patient-specific induced pluripotent stem cell-derived cardiomyocytes combined with force measurement, gene expression profiling, electrophysiology, and protein expression analysis. RESULTS Several angiotensin receptor blockers were identified from our in silico screen. Of these, olmesartan significantly elevated the expression of sarcomeric genes and rate and force of contraction and ameliorated arrhythmogenic potential. In addition, olmesartan exhibited the ability to reduce phosphorylation of extracellular signal-regulated kinase 1 in LMNA-mutant induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS In silico screening followed by in vitro validation with induced pluripotent stem cell-derived models can be an efficient approach to identifying repositionable therapies for monogenic cardiomyopathies.
Collapse
Affiliation(s)
- Eric J. Kort
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids (E.J.K.)
- Helen DeVos Children’s Hospital, Corewell Health, Grand Rapids, MI (E.J.K.)
| | - Nazish Sayed
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Division of Vascular Surgery, Department of Surgery, Stanford University, CA (N.S.)
| | - Chun Liu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
| | - Gema Mondéjar-Parreño
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
| | - Jens Forsberg
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
| | - Emily Eugster
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
| | - Sean M. Wu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, CA (S.M.W., J.C.W.)
| | - Joseph C. Wu
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University, CA (S.M.W., J.C.W.)
- Department of Radiology, Stanford University, CA (J.C.W.)
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Fredrik Meijer Heart and Vascular Institute, Spectrum Health and Van Andel Institute, Grand Rapids, MI (E.J.K., J.F., E.E., S.J.)
- Cardiovascular Institute, Stanford University, CA (N.S., C.L., G.M.-P., S.M.W., J.C.W., S.J.)
- Skåne University Hospital, Lund University, Lund, Sweden (S.J.)
| |
Collapse
|
31
|
Park S, Haam K, Heo H, Kim D, Kim M, Jung H, Cha S, Kim M, Lee H. Integrative transcriptomic analysis identifies emetine as a promising candidate for overcoming acquired resistance to ALK inhibitors in lung cancer. Mol Oncol 2025; 19:1155-1169. [PMID: 39540457 PMCID: PMC11977641 DOI: 10.1002/1878-0261.13738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/02/2024] [Accepted: 09/10/2024] [Indexed: 11/16/2024] Open
Abstract
Anaplastic lymphoma kinase (ALK; also known as ALK tyrosine kinase receptor) inhibitors (ALKi) are effective in treating lung cancer patients with chromosomal rearrangement of ALK. However, continuous treatment with ALKis invariably leads to acquired resistance in cancer cells. In this study, we propose an efficient strategy to suppress ALKi resistance through a meta-analysis of transcriptome data from various cell models of acquired resistance to ALKis. We systematically identified gene signatures that consistently showed altered expression during the development of resistance and conducted computational drug screening using these signatures. We identified emetine as a promising candidate compound to inhibit the growth of ALKi-resistant cells. We demonstrated that emetine exhibited effectiveness in inhibiting the growth of ALKi-resistant cells, and further interpreted its impact on the resistant signatures through drug-induced RNA-sequencing data. Our transcriptome-guided systematic approach paves the way for efficient drug discovery to overcome acquired resistance to cancer therapy.
Collapse
Affiliation(s)
- Sang‐Min Park
- College of PharmacyChungnam National UniversityDaejeonKorea
| | - Keeok Haam
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
| | - Haejeong Heo
- Personalized Genomic Medicine Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Department of Functional GenomicsUniversity of Science and Technology (UST)DaejeonKorea
| | - Doyeong Kim
- College of PharmacyChungnam National UniversityDaejeonKorea
| | - Min‐Ju Kim
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug DevelopmentPusan National UniversityBusanKorea
| | - Hyo‐Jung Jung
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
| | - Seongwon Cha
- Korean Medicine (KM) Data DivisionKorea Institute of Oriental MedicineDaejeonKorea
| | - Mirang Kim
- Aging Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Personalized Genomic Medicine Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonKorea
- Department of Functional GenomicsUniversity of Science and Technology (UST)DaejeonKorea
| | - Haeseung Lee
- Department of Pharmacy, College of Pharmacy and Research Institute for Drug DevelopmentPusan National UniversityBusanKorea
| |
Collapse
|
32
|
Sun T, Singh S, Wang HC, Lee J, Dolatshad H, Cheong PL, Higgs DR, Boultwood J, Pellagatti A. Identification of new candidate drugs in myelodysplastic syndromes with splicing factor mutations by transcriptional profiling and connectivity map analysis. Br J Haematol 2025; 206:1086-1091. [PMID: 39988885 PMCID: PMC11985364 DOI: 10.1111/bjh.20026] [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: 11/26/2024] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
We sought to identify new candidate drugs for repurposing to myelodysplastic syndromes (MDS). Connectivity map analysis was performed on gene expression signatures generated from bone marrow CD34+ cells of splicing factor mutant MDS patients. Celastrol and Withaferin A (WA), two top-ranking compounds identified, markedly inhibited proliferation, arrested the cell cycle and induced apoptosis in leukaemia cells. These compounds also inhibited the viability of primary bone marrow MDS cells. We showed that Celastrol and WA inhibit interleukin-1 receptor-associated kinase 4-mediated nuclear factor kappa-light-chain-enhancer of activated B cells signalling activation in splicing factor mutant MDS and leukaemia cells. Celastrol and WA may represent novel candidate drugs for the treatment of MDS.
Collapse
Affiliation(s)
- Tianyu Sun
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Shalini Singh
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Hayson Chenyu Wang
- Shanghai Ninth People's HospitalShanghai Jiaotong University School of MedicineShanghaiChina
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
| | - Juseong Lee
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Hamid Dolatshad
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Pak Leng Cheong
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Douglas R. Higgs
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Jacqueline Boultwood
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Andrea Pellagatti
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
33
|
Carroll E, Scaber J, Huber KVM, Brennan PE, Thompson AG, Turner MR, Talbot K. Drug repurposing in amyotrophic lateral sclerosis (ALS). Expert Opin Drug Discov 2025; 20:447-464. [PMID: 40029669 PMCID: PMC11974926 DOI: 10.1080/17460441.2025.2474661] [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: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Identifying treatments that can alter the natural history of amyotrophic lateral sclerosis (ALS) is challenging. For years, drug discovery in ALS has relied upon traditional approaches with limited success. Drug repurposing, where clinically approved drugs are reevaluated for other indications, offers an alternative strategy that overcomes some of the challenges associated with de novo drug discovery. AREAS COVERED In this review, the authors discuss the challenge of drug discovery in ALS and examine the potential of drug repurposing for the identification of new effective treatments. The authors consider a range of approaches, from screening in experimental models to computational approaches, and outline some general principles for preclinical and clinical research to help bridge the translational gap. Literature was reviewed from original publications, press releases and clinical trials. EXPERT OPINION Despite remaining challenges, drug repurposing offers the opportunity to improve therapeutic options for ALS patients. Nevertheless, stringent preclinical research will be necessary to identify the most promising compounds together with innovative experimental medicine studies to bridge the translational gap. The authors further highlight the importance of combining expertise across academia, industry and wider stakeholders, which will be key in the successful delivery of repurposed therapies to the clinic.
Collapse
Affiliation(s)
- Emily Carroll
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jakub Scaber
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Kilian V. M. Huber
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Paul E. Brennan
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| |
Collapse
|
34
|
Yang Y, Wang L, Yu L, Chang C, Zhang H, Hu L, Liu J, Zhang Y, Han H, Zhang H, Zhou Y, Wang J. Monocytes Expressing IL-36G Play a Crucial Role in Atopic Dermatitis. J Cell Mol Med 2025; 29:e70503. [PMID: 40159643 PMCID: PMC11955416 DOI: 10.1111/jcmm.70503] [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: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
Atopic dermatitis (ad) is a chronic inflammatory skin disease, with recent studies indicating that immune cells, such as monocytes and inflammatory cytokines, play a crucial role. By retrieving datasets from public databases and analysing immune cell infiltration in lesional skin using CIBERSORT, we found that monocytes and M2 macrophages were significantly upregulated in atopic dermatitis. Differentially expressed gene (DEG) functional enrichment analysis revealed that cytokine-cytokine receptor interaction was the most significantly enriched pathway. Further analysis of cytokines and their receptors, along with their correlation with infiltrating immune cells, identified IL36G-expressing monocytes as a key target in atopic dermatitis. We compared immune cell infiltration and cytokine-related targets in similar inflammatory skin diseases, such as psoriasis and urticaria, to evaluate similarities and differences among these three skin conditions. The analysis revealed that IL36G-expressing monocytes were also highly expressed in psoriasis but did not play a pivotal role in urticaria. Finally, we used molecular docking to predict and validate drugs targeting IL36G. Our study highlights IL36G-expressing monocytes as a common key target in atopic dermatitis and psoriasis, offering novel insights and therapeutic strategies for these related diseases.
Collapse
Affiliation(s)
- Yitao Yang
- School of MedicineShanghai UniversityShanghaiChina
| | - Lei Wang
- Hubei Shizhen LaboratoryHubei University of Chinese MedicineWuhanChina
| | - Longmei Yu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Chenxi Chang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Honglei Zhang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Linhan Hu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Juntong Liu
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Yihang Zhang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Hui Han
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Haiyun Zhang
- School of MedicineShanghai UniversityShanghaiChina
| | - Yumei Zhou
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| | - Ji Wang
- National Institute of TCM Constitution and Preventive MedicineBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
35
|
Wan Z, Sun X, Li Y, Chu T, Hao X, Cao Y, Zhang P. Applications of Artificial Intelligence in Drug Repurposing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411325. [PMID: 40047357 PMCID: PMC11984889 DOI: 10.1002/advs.202411325] [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: 09/15/2024] [Revised: 12/12/2024] [Indexed: 04/12/2025]
Abstract
Drug repurposing identifies new therapeutic uses for the existing drugs originally developed for different indications, aiming at capitalizing on the established safety and efficacy profiles of known drugs. Thus, it is beneficial to bypass of early stages of drug development, and to reduction of the time and cost associated with bringing new therapies to market. Traditional experimental methods are often time-consuming and expensive, making artificial intelligence (AI) a promising alternative due to its lower cost, computational advantages, and ability to uncover hidden patterns. This review focuses on the availability of AI algorithms in drug development, and their positive and specific roles in revealing repurposing of the existing drugs, especially being integrated with virtual screening. It is shown that the existing AI algorithms excel at analyzing large-scale datasets, identifying the complicated patterns of drug responses from these datasets, and making predictions for potential drug repurposing. Building on these insights, challenges remain in developing efficient AI algorithms and future research, including integrating drug-related data across databases for better repurposing, enhancing AI computational efficiency, and advancing personalized medicine.
Collapse
Affiliation(s)
- Zhaoman Wan
- State Key Laboratory of Common Mechanism Research for Major DiseasesSuzhou Institute of Systems MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhouJiangsu215123China
| | - Xinran Sun
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193China
| | - Yi Li
- Hunan Agriculture University College of Plant ProtectionChangshaHunan410128China
| | - Tianyao Chu
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| | - Xueyu Hao
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| | - Yang Cao
- College of Life SciencesSichuan UniversityChengduSichuan610041China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| |
Collapse
|
36
|
Bao N, Liu J, Wang H, Xing L, Xie Z, Liu C, Jin S, Jia J, Zhang M, Fan J. Drug Repurposing and Screening for Multiple Sclerosis Targeting Microglia and Macrophages. Mol Neurobiol 2025; 62:4724-4742. [PMID: 39485630 DOI: 10.1007/s12035-024-04602-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Microglia/macrophages (MG/Mφ) play a central role in the pathogenesis of multiple sclerosis (MS). However, the intricacies of the immunomodulatory microenvironment in MS, particularly the heterogeneity and regulatory mechanisms of MG/Mφ subpopulations, remain elusive. The commonly used treatment options for MS have several drawbacks, such as significant side effects and uncertain efficacy. The exploration of developing new drugs targeting MG/Mφ for the treatment of MS remains to be investigated. We identified three distinct subpopulations of MG/Mφ, among which MG/Mφ_3 significantly increased as the experimental autoimmune encephalomyelitis (EAE) progressed. Ifenprodil and RO-25-6981 demonstrated notable inhibition of inflammatory factor expression, accompanied by reduced cytotoxicity. The interaction modes of these compounds with the common binding pocket in the GluN1b-GluN2B amino terminal domain heterodimer were elucidated. Virtual docking, based on the N-methyl-D-aspartate (NMDA) receptor, showed that homo-skeleton compounds of ifenprodil potentially exhibit low binding free energy with the receptor, including eliprodil and volinanserin. In vitro cell models corroborated the effective inhibition of inflammatory factor expression and minimal cytotoxicity of eliprodil and volinanserin. CoMFA (standard error of estimate = 0.378, R2 = 0.928, F values = 241.255, Prob. of R2 = 0) and topomer CoMFA (q2 = 0.553, q2 stderr = 0.77, intercept = - 1.48, r2 = 0.908, r2 stderr = 0.35) were established based on the inhibitors of NMDA receptor. The contour maps of CoMFA and topomer CoMFA models give structural information to improve the inhibitory function. This study underscores the involvement of MG/Mφ in inflammatory pathways during MS progression and offers promising compound candidates for MS therapy targeting MG/Mφ.
Collapse
Affiliation(s)
- Nandi Bao
- Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Jing Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Heran Wang
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Lei Xing
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Zhonghui Xie
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chuanbin Liu
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Shaowei Jin
- National Supercomputing Shenzhen Center, Shenzhen, 518052, China
| | - Jianjun Jia
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China.
| | - Minghua Zhang
- Medical Supplies Center of PLA General Hospital, Beijing, 100853, China.
| | - Jiao Fan
- Institute of Geriatrics, National Clinical Research Center of Geriatrics Disease, the Second Medical Center of PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
37
|
Biswal S, Sahoo SK, Biswal BK. Shikonin a potent phytotherapeutic: a comprehensive review on metabolic reprogramming to overcome drug resistance in cancer. Mol Biol Rep 2025; 52:347. [PMID: 40156720 DOI: 10.1007/s11033-025-10459-6] [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: 11/28/2024] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Drug resistance remains a major challenge in cancer therapy, often leading to treatment failure. Metabolic reprogramming, a hallmark of cancer, plays a pivotal role in drug resistance. Phytocompounds, particularly shikonin, a naphthoquinone derived from Lithospermum erythrorhizon, have garnered significant interest as potential alternatives for cancer prevention and treatment. This review focuses on the anticancer properties of shikonin, particularly its ability to modulate metabolic reprogramming and overcome drug resistance. This review, based on extensive searches in databases like PubMed, Web of Science, Google Scholar, and Scopus, highlights shikonin's potential as a therapeutic agent. Shikonin exhibits a wide range of anticancer activities, including induction of apoptosis, autophagy, necroptosis, inhibition of angiogenesis, invasion, and migration, as well as disruption of the cell cycle and promotion of DNA damage. It targets altered cancer cell metabolism to inhibit proliferation and reverse drug resistance, making it a promising candidate for therapeutic development. Preliminary clinical trials suggest that shikonin can enhance the efficacy of established chemotherapeutic agents, immunotherapies, and radiation through additive and synergistic interactions. Despite its promise, further research is needed to elucidate the precise mechanisms underlying shikonin's metabolic reprogramming effects in cancer. A comprehensive understanding could pave the way for its integration into standard oncological treatments. With its capacity to act on multiple cancer pathways and enhance conventional treatments, shikonin stands out as a viable candidate for combating drug-resistant cancers and advancing clinical oncology.
Collapse
Affiliation(s)
- Stuti Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | | | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
38
|
Zhang L, Li H, Shi L, Geng J, Zhang H, Chen H, Zhao P, Xiao Y, Lu J, Li Z, Pu H, Hou C, Li C, Gao C, Song X, Bao Z, Zhai B, Guo B, Yang B, Lu X, Yu Q. Mechanism and Efficacy of Etanercept in Treating Autoimmune-like Manifestations of Coronavirus Disease 2019 in elderly individuals. Immunobiology 2025; 230:152898. [PMID: 40168796 DOI: 10.1016/j.imbio.2025.152898] [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: 12/05/2024] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
During the COVID-19 pandemic, extensive research focused on universal treatments, but few studies addressed treatment regimens for elderly patients. This study aimed to evaluate the effects of etanercept, a TNF inhibitor, in elderly individuals with COVID-19 through observational analysis of compassionate use cases. The results showed that after one month of etanercept treatment, clinical indicators such as C-reactive protein, D-dimer, and fibrinogen normalised, whereas the control group receiving conventional treatment did not fully recover. Single-cell sequencing was performed on seven patients treated with etanercept and two uninfected individuals. Based on our data and in conjunction with external data, a comprehensive characterization map involving 400,000 cells was created. Transcriptomic analysis revealed autoimmune-like manifestations in elderly patients, highlighting the importance of immunotherapy. Plasma cells, platelets, and B cells were the most treatment-sensitive cells. Analysis of five drug types, including antiviral, etanercept, glucocorticoids, tocilizumab, and others, showed that tocilizumab was associated with an increased thrombosis risk in elderly patients. Meanwhile, etanercept alleviated autoimmune-like manifestations by inhibiting platelet factor 4 and suppressing TNF-α. Molecular docking showed etanercept's strong affinity (-15.0 kcal/mol) for the spike protein of the SARS-CoV-2 Omicron variant, suggesting it may protect immune-compromised patients. Our findings support etanercept as a potential treatment for elderly COVID-19 patients with autoimmune-like manifestations.
Collapse
Affiliation(s)
- Lizhong Zhang
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China
| | - Hongyi Li
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Lei Shi
- Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100853, China
| | - Jie Geng
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China
| | - Haojun Zhang
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China
| | - Haoran Chen
- School of Management, Shanxi Medical University, Taiyuan, 030000, China
| | - Peng Zhao
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China
| | - Yang Xiao
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Jinqi Lu
- Department of Computer Science, Whiting School of Engineering, Johns Hopkins University, 3400 N Charles St, Baltimore, MD 21218, USA
| | - Zhilun Li
- School of basic medicine and clinical pharmacy, China Pharmaceutical University, Nanjing 211100, China
| | - Hongbin Pu
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Chuandong Hou
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Chenghui Li
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Chumeng Gao
- Fuxing Road Outpatient Department, Jingnan Medical District, PLA General Hospital, Beijing 100842, China
| | - Xia Song
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China
| | - Zhuocheng Bao
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China
| | - Bing Zhai
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China
| | - Bo Guo
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China
| | - Bo Yang
- Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China
| | - Xuechun Lu
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China; Department of Hematology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Disease, Beijing 100853, China; Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People's Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China.
| | - Qi Yu
- Basic Medicine College, Shanxi Medical University, Taiyuan 030000, China; School of Management, Shanxi Medical University, Taiyuan, 030000, China; Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
39
|
Zhong X, Wei Q, Tiwari A, Wang Q, Tan Y, Chen R, Yan Y, Cox NJ, Li B. A Genetics-guided Integrative Framework for Drug Repurposing: Identifying Anti-hypertensive Drug Telmisartan for Type 2 Diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.22.25324223. [PMID: 40166562 PMCID: PMC11957187 DOI: 10.1101/2025.03.22.25324223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Drug development is a long and costly process, and repurposing existing drugs for use toward a different disease or condition may serve as a cost-effective alternative. As drug targets with genetic support have a doubled success rate, genetics-informed drug repurposing holds promise in translating genetic findings into therapeutics. In this study, we developed a Genetics Informed Network-based Drug Repurposing via in silico Perturbation (GIN-DRIP) framework and applied the framework to repurpose drugs for type-2 diabetes (T2D). In GIN-DRIP for T2D, it integrates multi-level omics data to translate T2D GWAS signals into a genetics-informed network that simultaneously encodes gene importance scores and a directional effect (up/down) of risk genes for T2D; it then bases on the GIN to perform signature matching with drug perturbation experiments to identify drugs that can counteract the effect of T2D risk alleles. With this approach, we identified 3 high-confidence FDA-approved candidate drugs for T2D, and validated telmisartan, an anti-hypertensive drug, in our EHR data with over 3 million patients. We found that telmisartan users were associated with a reduced incidence of T2D compared to users of other anti-hypertensive drugs and non-users, supporting the therapeutic potential of telmisartan for T2D. Our framework can be applied to other diseases for translating GWAS findings to aid drug repurposing for complex diseases.
Collapse
Affiliation(s)
- Xue Zhong
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Qiang Wei
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Anshul Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Quan Wang
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Yuting Tan
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Rui Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| | - Yan Yan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Nancy J Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Bingshan Li
- Department of Molecular Physiology and Biophysics, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN
| |
Collapse
|
40
|
Zhang Y, Qin L, Liu J. Bioinformatics and machine learning approaches to explore key biomarkers in muscle aging linked to adipogenesis. BMC Musculoskelet Disord 2025; 26:285. [PMID: 40121419 PMCID: PMC11929359 DOI: 10.1186/s12891-025-08528-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Adipogenesis is intricately linked to the onset and progression of muscle aging; however, the relevant biomarkers remain unclear. This study sought to identify key genes associated with adipogenesis in the context of muscle aging. Firstly, gene expression profiles from biopsies of the vastus lateralis muscle in both young and elderly population were retrieved from the GEO database. After intersecting with the results of differential gene analysis, weighted gene co-expression network analysis, and sets of adipogenesis-related genes, 29 adipogenesis-related differential expressed genes (ARDEGs) were selected. Connectivity Map (cMAP) analysis identified tamsulosin, fraxidin, and alaproclate as key target compounds. In further, using three machine learning algorithms and the friends analysis, four hub ARDEGs, ESRRA, RXRG, GADD45A, and CEBPB were identified and verified in vivo aged mice muscles. Immune infiltration analysis showed a strong link between several immune cells and hub ARDEGs. In all, these findings suggested that ESRRA, RXRG, GADD45A, and CEBPB could serve as adipogenesis related biomarkers in muscle aging.
Collapse
Affiliation(s)
- Yumin Zhang
- Division of Geriatric Endocrinology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| | - Li Qin
- Division of Geriatric Endocrinology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Juan Liu
- Division of Geriatric Endocrinology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.
| |
Collapse
|
41
|
Tanoli Z, Fernández-Torras A, Özcan UO, Kushnir A, Nader KM, Gadiya Y, Fiorenza L, Ianevski A, Vähä-Koskela M, Miihkinen M, Seemab U, Leinonen H, Seashore-Ludlow B, Tampere M, Kalman A, Ballante F, Benfenati E, Saunders G, Potdar S, Gómez García I, García-Serna R, Talarico C, Beccari AR, Schaal W, Polo A, Costantini S, Cabri E, Jacobs M, Saarela J, Budillon A, Spjuth O, Östling P, Xhaard H, Quintana J, Mestres J, Gribbon P, Ussi AE, Lo DC, de Kort M, Wennerberg K, Fratelli M, Carreras-Puigvert J, Aittokallio T. Computational drug repurposing: approaches, evaluation of in silico resources and case studies. Nat Rev Drug Discov 2025:10.1038/s41573-025-01164-x. [PMID: 40102635 DOI: 10.1038/s41573-025-01164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Repurposing of existing drugs for new indications has attracted substantial attention owing to its potential to accelerate drug development and reduce costs. Hundreds of computational resources such as databases and predictive platforms have been developed that can be applied for drug repurposing, making it challenging to select the right resource for a specific drug repurposing project. With the aim of helping to address this challenge, here we overview computational approaches to drug repurposing based on a comprehensive survey of available in silico resources using a purpose-built drug repurposing ontology that classifies the resources into hierarchical categories and provides application-specific information. We also present an expert evaluation of selected resources and three drug repurposing case studies implemented within the Horizon Europe REMEDi4ALL project to demonstrate the practical use of the resources. This comprehensive Review with expert evaluations and case studies provides guidelines and recommendations on the best use of various in silico resources for drug repurposing and establishes a basis for a sustainable and extendable drug repurposing web catalogue.
Collapse
Affiliation(s)
- Ziaurrehman Tanoli
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland.
| | | | - Umut Onur Özcan
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aleksandr Kushnir
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kristen Michelle Nader
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Yojana Gadiya
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
- Bonn-Aachen International Center for Information Technology (B-IT), University of Bonn, Bonn, Germany
| | - Laura Fiorenza
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milan, Italy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Markus Vähä-Koskela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mitro Miihkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Umair Seemab
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Henri Leinonen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Brinton Seashore-Ludlow
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Marianna Tampere
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Adelinn Kalman
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Flavio Ballante
- Chemical Biology Consortium Sweden (CBCS), SciLifeLab, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Emilio Benfenati
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Gary Saunders
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | | | | | - Wesley Schaal
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Andrea Polo
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Susan Costantini
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Enrico Cabri
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marc Jacobs
- Fraunhofer-Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Jani Saarela
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Alfredo Budillon
- Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Päivi Östling
- Science for Life Laboratory (SciLifeLab), Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Henri Xhaard
- Drug Discovery and Chemical Biology (DDCB) Consortium, Biocenter Finland, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Jordi Quintana
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Mestres
- Chemotargets SL, Parc Científic de Barcelona, Barcelona, Catalonia, Spain
- Institut de Quimica Computacional i Catalisi, Facultat de Ciencies, Universitat de Girona, Girona, Catalonia, Spain
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
| | - Anton E Ussi
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Donald C Lo
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Martin de Kort
- European Infrastructure for Translational Medicine (EATRIS ERIC), Amsterdam, The Netherlands
| | - Krister Wennerberg
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | | | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
42
|
Zhou W, Ruan H, Zhu L, Chen S, Yang M. Unveiling a Novel Glioblastoma Deep Molecular Profiling: Insight into the Cancer Cell Differentiation-Related Mechanisms. ACS OMEGA 2025; 10:10230-10250. [PMID: 40124014 PMCID: PMC11923693 DOI: 10.1021/acsomega.4c09586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/27/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND the sophisticated cellular heterogeneity of cell populations in glioblastoma (GBM) has been a key factor influencing tumor progression and response to therapy. The lack of more precise stratification based on cellular differentiation status poses a great challenge to therapeutic strategies. MATERIALS AND METHODS harnessing the bulk multiomics and single-nucleus RNA sequencing data available from the National Center for Biotechnology Information (NCBI) and The Cancer Genome Atlas (TCGA) Program repositories, we developed a novel and accurate GBM risk classification using an ensemble consensus clustering approach based on the junction of prognosis and trajectory analysis. Comprehensive cluster labeling and multiomics data characterization were also performed. RESULTS a novel GBM stratification model was constructed using 45 malignant cell fate genes: (a) energy metabolism-enhanced-type GBM; (b) invasion-enhanced-type GBM; (c) invasion-attenuated-type GBM; and (d) glycolysis-dominant energy metabolism-enhanced-type GBM. The biological plausibility of the model was verified through a range of comprehensive analyses of multiomics data, showing that cases with invasion-attenuated-type were the best prognosis and energy metabolism-enhanced-type the poorest. CONCLUSIONS the study has uncovered GBM complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying tumorigenesis. This precise stratification system provided implications for further studies of individual therapies.
Collapse
Affiliation(s)
- Weili Zhou
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Hongtao Ruan
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Lihua Zhu
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Shunqiang Chen
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Muyi Yang
- Department of Radiology, Henan Provincial People’s Hospital & the
People’s Hospital of Zhengzhou University, Zhengzhou, Henan 450003, China
| |
Collapse
|
43
|
Meng J, Hu D, Wang B, Zhu Y, Lu C, Deng Y, Cai H, Wang B, He Y, Qian W. Fine mapping and candidate gene analysis of the major QTL qSW-A03 for seed weight in Brassica napus. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2025; 138:76. [PMID: 40097747 DOI: 10.1007/s00122-025-04866-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Seed weight is a determining factor for improving rapeseed productivity. In the present study, a high-density genetic map was constructed via genome resequencing in an RIL population derived from a cross of two rapeseed varieties, ZS11 and DL704, with great differences in thousand-seed weight (TSW). A total of 1,306 bins involving 1,261,526 markers were used to construct the bin map. On the basis of the genetic map, QTL mapping for seed weight was performed. In total, 15 QTLs associated with TSW were detected. A major and stable QTL, qSW-A03, was mapped to a 2.8 cM interval on chromosome A03. Fine mapping delimited the qSW-A03 locus into a 59-kb region, and 11 genes within this region were predicted. By employing a combination of gene variation, gene expression difference and gene coexpression network analysis of seed weight, BnaDUF1666 was identified as a promising candidate gene. This study provides useful information for the genetic dissection of seed weight and promotes the molecular breeding of high-yield rapeseed.
Collapse
Affiliation(s)
- Jiangyu Meng
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Dingxue Hu
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Bin Wang
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Yuelin Zhu
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Chunyan Lu
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Yan Deng
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Huiying Cai
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China
| | - Baohua Wang
- School of Life Sciences, Nantong University, Nantong, 226019, Jiangsu, China
| | - Yajun He
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China.
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China.
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China.
| | - Wei Qian
- Academy of Agricultural Sciences, Southwest University, Chongqing, 400715, China.
- Integrative Science Center of Germplasm Creation in Western China (Chongqing) Science City, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China.
- Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China.
| |
Collapse
|
44
|
Li X, Zhang X, Liu T, Zhang G, Chen D, Lin S. Identification of immune characteristic biomarkers and therapeutic targets in cuproptosis for rheumatoid arthritis by integrated bioinformatics analysis and single-cell RNA sequencing analysis. Front Med (Lausanne) 2025; 12:1520400. [PMID: 40166070 PMCID: PMC11955502 DOI: 10.3389/fmed.2025.1520400] [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: 11/01/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Rheumatoid arthritis (RA) is a chronic autoimmune disorder intricately liked with inflammation. Cuproptosis, an emerging type of cell death, has been implicated in the initiation and development of RA. However, the exact alterations in the expression and biological function of cuproptosis-related genes (CRGs) in RA remain poorly understood. Therefore, our study aims to elucidate the potential association between CRGs and RA, with the goal of identifying novel biomarkers for the treatment and prognosis of RA. Methods In this study, we identified ten differentially expressed cuproptosis-related genes (DE-CRGs) between patients with RA and controls. Through comprehensive functional enrichment and protein-protein interaction (PPI) network analysis, we explored the functional roles of the DE-CRGs. Additionally, we investigated the correlation between DE-CRGs and immune infiltration, immune factors, diagnostic efficacy, and potential therapeutic drugs. Results Leveraging single-cell RNA sequencing data, we conducted a detailed analysis to elucidate alterations in various cell clusters associated with RA. Our study unveiled a significant association between DE-CRGs and diverse biological functions, as well as potential drug candidates. Discussion These findings provide crucial insights into the involvement of DE-CRGs in the pathogenesis of RA and shed light on potential therapeutic strategies.
Collapse
Affiliation(s)
- Xianbin Li
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou, China
- Jiujiang Key Laboratory of Digital Technology, Jiujiang, China
| | - Xueli Zhang
- Department of Medical Technology, Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Tao Liu
- School of Computer and Big Data Science, Jiujiang University, Jiujiang, China
| | - Guodao Zhang
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou, China
| | - Dan Chen
- Department of Rheumatology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Suxian Lin
- Department of Rheumatology, Wenzhou People’s Hospital, Wenzhou, China
| |
Collapse
|
45
|
Gu Z, Ye F, Luo H, Li X, Gong Y, Mao S, Jia X, Han X, Han B, Fu Y, Cheng X, Li J, Shao Z, Wen P, Hu X, Zhuang Z. Metformin sensitizes triple-negative breast cancer to histone deacetylase inhibitors by targeting FGFR4. J Biomed Sci 2025; 32:36. [PMID: 40091020 PMCID: PMC11912690 DOI: 10.1186/s12929-025-01129-7] [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: 02/07/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by high malignancy, strong invasiveness, and a propensity for distant metastasis, leading to poor prognosis and relatively limited treatment options. Metformin, as a first-line oral hypoglycemic agent, has garnered widespread research interest in recent years due to its potential in cancer prevention and treatment. However, its efficacy varies significantly across different tumor types. Histone deacetylase inhibitors (HDACi), such as SAHA, have demonstrated antitumor activity, but TNBC responds poorly to HDACi monotherapy, possibly due to feedback activation of the JAK-STAT pathway. Exploring the synergistic potential and underlying mechanisms of combining metformin with HDACi in TNBC treatment is crucial. METHODS We predicted the synergistic effects of metformin and SAHA in TNBC using multiple computational methods (CMap, DTsyn, and DrugComb). We also developed a cancer-specific compound mimic library (CDTSL) and applied a three-step strategy to identify genes fitting the "metformin sensitization" model. Subsequently, we evaluated the synergistic effects of metformin and SAHA in TNBC cell lines through cell proliferation, colony formation, and apoptosis assays. Furthermore, we investigated the molecular mechanisms of the combined treatment using techniques such as transcriptome sequencing, chromatin immunoprecipitation (ChIP), Western blotting, and measurement of extracellular acidification rate (ECAR). Additionally, we assessed the in vivo antitumor effects of the combined therapy in a nude mouse subcutaneous xenograft model. RESULTS CMap, DTsyn, and DrugComb all predicted the synergistic effects of SAHA and metformin in TNBC. The screening results revealed that HDAC10 played a key role in metformin sensitization. We found that the combination of metformin and SAHA exhibited synergistic antitumor effects (combination index CI < 0.9) in TNBC cell lines. Mechanistically, metformin inhibited histone acetylation on FGFR4, thereby blocking the feedback activation of FGFR4 downstream pathways induced by SAHA. Furthermore, metformin interfered with the glycolysis process induced by SAHA, altering the metabolic reprogramming of tumor cells. In in vivo experiments, the combined treatment of metformin and SAHA significantly inhibited the growth of subcutaneous tumors in nude mice. CONCLUSIONS Metformin enhances the sensitivity of TNBC to HDAC inhibitors by blocking the FGFR4 pathway and interfering with metabolic reprogramming. When used in combination with SAHA, metformin exhibits synergistic antitumor effects. Our study provides a theoretical basis for the combined application of HDAC inhibitors and metformin, potentially offering a new strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Zhangyuan Gu
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gao-Ke Road, Shanghai, 201204, China
| | - Fugui Ye
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hong Luo
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoguang Li
- Shanghai Henlius Biotech Inc., Shanghai, 200233, China
| | - Yue Gong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Xiaoqing Jia
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gao-Ke Road, Shanghai, 201204, China
| | - Xiangchen Han
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Boyue Han
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yun Fu
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gao-Ke Road, Shanghai, 201204, China
| | - Xiaolin Cheng
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gao-Ke Road, Shanghai, 201204, China
| | - Jiejing Li
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gao-Ke Road, Shanghai, 201204, China
| | - Zhiming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Precision Cancer Medical Center, Affiliated to Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 201315, China.
| | - Peizhen Wen
- Department of General Surgery, School of Medicine, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, Xiamen University, No. 2000 Xiang'an East Road, Xiamen, 361005, Fujian, China.
- Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Organ Transplantation Institute of Xiamen University, Xiamen University, Xiamen, 361005, Fujian, China.
| | - Xin Hu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Precision Cancer Medical Center, Affiliated to Fudan University Shanghai Cancer Center, No.688 Hong-Qu Road, Shanghai, 201315, China.
| | - Zhigang Zhuang
- Department of Breast Surgery, Shanghai Key Laboratory of Maternal-Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, No. 2699 West Gao-Ke Road, Shanghai, 201204, China.
| |
Collapse
|
46
|
Zhang Y, Guo Z, Lai R, Zou X, Ma L, Cai T, Huang J, Huang W, Zou B, Zhou J, Li J. Comprehensive Analysis Based on Genes Associated With Cuproptosis, Ferroptosis, and Pyroptosis for the Prediction of Diagnosis and Therapies in Coronary Artery Disease. Cardiovasc Ther 2025; 2025:9106621. [PMID: 40124544 PMCID: PMC11929595 DOI: 10.1155/cdr/9106621] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 11/06/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Coronary artery disease (CAD) is a complex condition influenced by genetic factors, lifestyle, and other risk factors that contribute to increased mortality. This study is aimed at evaluating the diagnostic potential of genes associated with cuproptosis, ferroptosis, and pyroptosis (CFP) using network modularization and machine learning methods. CAD-related datasets GSE42148, GSE20680, and GSE20681 were sourced from the GEO database, and genes related to CFP genes were gathered from MsigDB and FerrDb datasets and literature. To identify diagnostic genes linked to these pathways, weighted gene coexpression network analysis (WGCNA) was used to isolate CAD-related modules. The diagnostic accuracy of key genes in these modules was then assessed using LASSO, SVM, and random forest models. Immunity and drug sensitivity correlation analyses were subsequently performed to investigate possible underlying mechanisms. The function of a potential gene, STK17B, was analyzed through western blot and transwell assays. Two CAD-related modules with strong correlations were identified and validated. The SVM model outperformed LASSO and random forest models, demonstrating superior discriminative power (AUC = 0.997 in the blue module and AUC = 1.000 in the turquoise module), with nine key genes identified: CTDSP2, DHRS7, NLRP1, MARCKS, PELI1, RILPL2, JUNB, STK17B, and SLC40A1. Knockdown of STK17B inhibited cell migration and invasion in human umbilical vein endothelial cells. In summary, our findings suggest that CFP genes hold potential as diagnostic biomarkers and therapeutic targets, with STK17B playing a role in CAD progression.
Collapse
Affiliation(s)
- Yongyi Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Zhehan Guo
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Renkui Lai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xu Zou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Liuling Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Tianjin Cai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jingyi Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Wenxiang Huang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Bingcheng Zou
- Schoole of Life Science, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinming Zhou
- Schoole of Life Science, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinxin Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
47
|
Alijagic A, Seilitz FS, Bredberg A, Hakonen A, Larsson M, Selin E, Sjöberg V, Kotlyar O, Scherbak N, Repsilber D, Kärrman A, Wang T, Särndahl E, Engwall M. Deciphering the phenotypic, inflammatory, and endocrine disrupting impacts of e-waste plastic-associated chemicals. ENVIRONMENTAL RESEARCH 2025; 269:120929. [PMID: 39862959 DOI: 10.1016/j.envres.2025.120929] [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: 11/13/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
As the volume of plastic waste from electrical and electronic equipment (WEEE) continues to rise, a significant portion is disposed of in the environment, with only a small fraction being recycled. Both disposal and recycling pose unknown health risks that require immediate attention. Existing knowledge of WEEE plastic toxicity is limited and mostly relies on epidemiological data and association studies, with few insights into the underlying toxicity mechanisms. Therefore, this study aimed to perform comprehensive chemical screening and mechanistic toxicological assessment of WEEE plastic-associated chemicals. Chemical analysis, utilizing suspect screening based on high-resolution mass spectrometry, along with quantitative target chemical analysis, unveiled numerous hazardous compounds including polyaromatic compounds, organophosphate flame retardants, phthalates, benzotriazoles, etc. Toxicity endpoints included perturbation of morphological phenotypes using the Cell Painting assay, inflammatory response, oxidative stress, and endocrine disruption. Results demonstrated that WEEE plastic chemicals altered the phenotypes of the cytoskeleton, endoplasmic reticulum, and mitochondria in a dose-dependent manner. In addition, WEEE chemicals induced inflammatory responses in resting macrophages and altered inflammatory responses in lipopolysaccharide-primed macrophages. Furthermore, WEEE chemicals activated the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, indicating oxidative stress, and the aryl hydrocarbon receptor (AhR). Endocrine disruption was also observed through the activation of estrogenic receptor-α (ER-α) and the induction of anti-androgenic activity. The findings show that WEEE plastic-associated chemicals exert effects in multiple subcellular sites, via different receptors and mechanisms. Thus, an integrated approach employing both chemical and toxicological methods is essential for comprehensive assessment of the toxicity mechanisms and cumulative chemical burden of WEEE plastic-associated chemicals.
Collapse
Affiliation(s)
- Andi Alijagic
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden; Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, SE-701 82, Sweden.
| | | | - Anna Bredberg
- RISE, Research Institutes of Sweden, Gothenburg, SE-412 58, Sweden
| | - Aron Hakonen
- Sensor Visions AB, Hisings Backa, SE-455 22, Sweden
| | - Maria Larsson
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden
| | - Erica Selin
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden
| | - Viktor Sjöberg
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden
| | - Oleksandr Kotlyar
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden; Centre for Applied Autonomous Sensor Systems (AASS), Robot Navigation & Perception Lab (RNP), Örebro University, SE-701 82, Örebro, Sweden
| | - Nikolai Scherbak
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden
| | - Dirk Repsilber
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, SE-701 82, Sweden
| | - Anna Kärrman
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden
| | - Thanh Wang
- Department of Physics, Chemistry and Biology (IFM), Linköping University, SE-583 30, Linköping, Sweden; Department of Thematic Studies, Environmental Change, Linköping University, SE-58183, Linköping, Sweden
| | - Eva Särndahl
- Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, SE-701 82, Sweden; School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, SE-701 82, Sweden
| | - Magnus Engwall
- Man-Technology-Environment Research Center (MTM), Örebro University, Örebro, SE-701 82, Sweden
| |
Collapse
|
48
|
Wang F, Wang J, Chen T, Wang S, Meng X, Shen Y, Xu X. Systematic Identification of Mitochondrial Signatures in Alzheimer's Disease and Inflammatory Bowel Disease. Mol Neurobiol 2025:10.1007/s12035-025-04826-4. [PMID: 40085351 DOI: 10.1007/s12035-025-04826-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Mitochondrial dysfunction is increasingly recognized as a shared feature of Alzheimer's disease (AD) and inflammatory bowel disease (IBD), linked through overlapping pathways of hypoxia and immune dysregulation. Our study integrated transcriptomic and genetic analyses to uncover mitochondria-related mechanisms underlying these diseases. By analyzing multiple AD and IBD datasets through differential expression gene (DEG) analyses, biological pathway enrichment, and co-expression module construction, we identified hypoxia-induced mitochondrial dysfunction as a central risk factor for both conditions. Key findings revealed several mitochondrial-related genes shared between AD and IBD, including BCL6, PFKFB3, NDUFS3, and COX5B, which serve as critical regulators bridging mitochondrial and immune pathways. Drug enrichment analyses using Drug Signatures Database (DsigDB) and the Connectivity Map (cMAP) identified promising therapeutic candidates, including decitabine, DMOG, and estradiol, targeting shared regulators such as BCL6, PFKFB3, MAFF, and TGFBI. These drugs demonstrated potential to modulate mitochondrial autophagy and oxidative phosphorylation (OXPHOS), pathways enriched in the constructed interaction network with BCL6 and PFKFB3 as central nodes. Mendelian randomization (MR) analysis further identified MAP1LC3A as significantly associated with increased risk for both AD and IBD, while NME1 emerged as strongly protective, suggesting their roles as therapeutic targets. Our findings underscore hypoxia-induced mitochondrial dysfunction as a unifying mechanism in AD and IBD, mediated by hypoxia-inducible factor-1α (HIF-1α). By identifying key mitochondria-associated genes and pathways, this study highlights innovative therapeutic targets and contributes to a deeper understanding of the gut-brain interplay in neurodegeneration and chronic inflammation. These insights pave the way for precision medicine strategies targeting mitochondrial dysfunction in AD and IBD.
Collapse
Affiliation(s)
- Fei Wang
- School of Basic Medical Science, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jiaqi Wang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Tong Chen
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shuaibin Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, Anhui, China
| | - XiangYu Meng
- School of Basic Medical Sciences, Medical School, Hubei Minzu University, Enshi, 445000, Hubei, China
| | - Yin Shen
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Xuan Xu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
49
|
Krishna N, Ramalakshmi NA, Krishnamurthy RG. Comprehensive Bioinformatics Analysis Reveals Molecular Signatures and Potential Caloric Restriction Mimetics with Neuroprotective Effects: Validation in an In Vitro Stroke Model. J Mol Neurosci 2025; 75:32. [PMID: 40080242 DOI: 10.1007/s12031-025-02328-5] [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: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Caloric restriction (CR) is a dietary intervention that reduces calorie intake without inducing malnutrition, demonstrating lifespan-extending effects in preclinical studies and some human trials, along with potential benefits in ameliorating age-related ailments. Caloric restriction mimetics (CRMs) are compounds mimicking CR effects, offering a potential therapeutic avenue for age-related diseases. This study explores the potential protective effects of CR on the brain neocortex (GSE11291) and the identification of CRMs using integrative bioinformatics and systems biology approaches. Our findings indicate that long-term CR activates cellular pathways improving mitochondrial function, enhancing antioxidant capacity, and reducing inflammation, potentially providing neuroprotection. The key signaling pathways enriched in our study include PPAR, mTOR, FoxO, AMPK, and Notch signaling pathways, which are crucial regulators of metabolism, cellular stress response, neuroprotection, and longevity. We identify key signaling molecules and molecular mechanisms associated with CR, including transcription factors, kinase regulators, and microRNAs linked to differentially expressed genes. Furthermore, potential CRMs such as rapamycin, replicating CR-related health benefits, are identified. Additionally, machine learning models were developed to classify small molecules based on their CNS activity and anti-inflammatory properties. As a proof of concept, we have demonstrated the ischemic neuroprotective effects of two top-ranked candidate reference molecules (CRMs) using the oxygen-glucose deprivation (OGD) model, an established in vitro stroke model. However, further investigations are essential to fully elucidate the therapeutic potential of these CRMs. In summary, our study suggests that long-term CR entails protective mechanisms preserving and safeguarding neuronal function, potentially impacting the treatment of age-related neurological diseases. Moreover, our findings contribute to the identification of potential genes and regulatory molecules involved in CR, along with potential CRMs, providing a promising foundation for future research in the field of neurological disorder treatment.
Collapse
Affiliation(s)
- Navami Krishna
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Calicut, Kerala, India, 673601
| | | | | |
Collapse
|
50
|
Asiaee A, Abrams ZB, Pua HH, Coombes KR. Transcriptome Complexity Disentangled: A Regulatory Molecules Approach. Int J Mol Sci 2025; 26:2510. [PMID: 40141153 PMCID: PMC11942001 DOI: 10.3390/ijms26062510] [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: 01/05/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Transcription factors (TFs) and microRNAs (miRNAs) are fundamental regulators of gene expression, cell state, and biological processes. This study investigated whether a small subset of TFs and miRNAs could accurately predict genome-wide gene expression. We analyzed 8895 samples across 31 cancer types from The Cancer Genome Atlas and identified 28 miRNA and 28 TF clusters using unsupervised learning. Medoids of these clusters could differentiate tissues of origin with 92.8% accuracy, demonstrating their biological relevance. We developed Tissue-Agnostic and Tissue-Aware models to predict 20,000 gene expressions using the 56 selected medoid miRNAs and TFs. The Tissue-Aware model attained an R2 of 0.70 by incorporating tissue-specific information. Despite measuring only 1/400th of the transcriptome, the prediction accuracy was comparable to that achieved by the 1000 landmark genes. This suggests the transcriptome has an intrinsically low-dimensional structure that can be captured by a few regulatory molecules. Our approach could enable cheaper transcriptome assays and analysis of low-quality samples. It also provides insights into genes that are heavily regulated by miRNAs/TFs versus alternative mechanisms. However, model transportability was impacted by dataset discrepancies, especially in miRNA distribution. Overall, this study demonstrates the potential of a biology-guided approach for robust transcriptome representation.
Collapse
Affiliation(s)
- Amir Asiaee
- Department of Biostatistics, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, USA
| | - Zachary B. Abrams
- Institute for Informatics, Washington University, 4444 Forest Park Avenue, St. Louis, MO 63108, USA;
| | - Heather H. Pua
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 Medical Center Drive, Nashville, TN 37240, USA;
| | - Kevin R. Coombes
- Department of Population Health Science, Medical College of Georgia, 1120 15th Street, Augusta, GA 30912, USA;
| |
Collapse
|