1
|
Fujita Y, De Velasco MA, Hayashi H, Nakagawa K, Nishio K. Exploration of genes related to the development of cancer of unknown primary. Oncol Rep 2025; 53:72. [PMID: 40314076 PMCID: PMC12062861 DOI: 10.3892/or.2025.8905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 05/03/2025] Open
Abstract
The biological basis of the development of cancer of unknown primary (CUP) remains largely unknown, with no evidence of whether a common biological basis exists at present. Our previous multicenter clinical study predicted the primary site of CUP for site‑specific therapy. Concomitantly with the study, a microarray analysis of tumor mRNA samples obtained from 60 participants of the study with CUP was performed, and a gene expression profile specific to CUP was constructed. Several of the genes identified as being upregulated/downregulated in CUP could potentially be clinically useful common biomarkers of CUP. In the present study, to identify genes that may be more closely related to the development of CUP (characterized by its metastatic potential) among the upregulated genes, cell‑based small interfering RNA screening was performed in vitro, and two genes, protein kinase DNA‑activated catalytic subunit (PRKDC) and proteasome subunit β type‑4 (PSMB4), were identified to be possibly involved in the metastatic ability of CUP, since knockdown of these genes resulted in reduced migration of A549 cells. These genes were further knocked down in A549 cells using short hairpin RNAs (shRNAs) and the cells were implanted into the footpad of mice. Marked suppression of the metastatic ability of implanted cells from the footpad to the popliteal lymph node (LN) was observed in cells transfected with the shRNAs for PRKDC and PSMB4. In addition, bortezomib, a proteasome inhibitor, markedly reduced the ability of cells implanted into the footpad to metastasize to the LNs, as well as cell growth at the metastatic site, compared with vehicle or NU7447 (inhibitor of PRKDC). These findings indicated that proteasomal function activation augmented the metastatic ability of malignant CUP cells.
Collapse
Affiliation(s)
- Yoshihiko Fujita
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Marco A. De Velasco
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
2
|
Guo S, Zhang Q, Liu YJ, Hu YY, Liu C, Shen H, Liu J. Hypoxia-induced RHCG as a key regulator in psoriasis and its modulation by secukinumab. APL Bioeng 2025; 9:026115. [PMID: 40351602 PMCID: PMC12065634 DOI: 10.1063/5.0250742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
The interaction between keratinocytes (KCs) and immune cells is essential in the pathogenesis of psoriasis. Understanding this crosstalk is crucial for developing effective treatment strategies. Recent studies indicate that Rh family C-type glycoprotein (RHCG) enhances cell proliferation and alters cell differentiation; however, its exact pathogenic mechanisms in psoriasis remain unclear. We employed bioinformatics approaches, including spatial transcriptomics analysis, single-cell transcriptomics analysis, and bulk data analysis, to elucidate the biological functions of RHCG. These predictions were validated through ex vivo experiments and analysis of clinical specimens. In psoriatic skin, RHCG protein levels were significantly upregulated, with an expanded expression area. Notably, RHCG expression was induced under hypoxic conditions. Furthermore, the upregulation of RHCG enhanced the expression of KC markers S100 Calcium Binding Protein A family (S100A) and Keratin 17 (KRT17), while decreasing Keratin 1 (KRT1) expression. Additionally, RHCG overexpression increased the secretion of C-X-C motif chemokine ligand 14 (CXCL14) from KCs, which subsequently activated dendritic cells. Importantly, treatment with secukinumab effectively ameliorated psoriasis by downregulating RHCG expression and inhibiting associated signaling pathways, whereas glucocorticoid and methotrexate treatments resulted in elevated RHCG expression. These findings indicate that RHCG plays a significant role in hypoxia-induced cellular crosstalk and suggest that RHCG-associated signaling may contribute to the superior efficacy of biological agents compared to conventional hormonal and immunosuppressive therapies.
Collapse
Affiliation(s)
- Shun Guo
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, People's Republic of China
| | - Qian Zhang
- Department of Dermatology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600 Jiangsu, People's Republic of China
| | | | | | - Cong Liu
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, People's Republic of China
| | - Hui Shen
- Department of Dermatology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600 Jiangsu, People's Republic of China
| | - Jia Liu
- Department of Dermatology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, People's Republic of China
| |
Collapse
|
3
|
Imran M, Alshrari AS, Hafiz MN, Jawad MM, Khan A, Alanazi FJ, Asdaq SMB. Exploring therapeutic paradigm focusing on genes, proteins, and pathways to combat leprosy and tuberculosis: A network medicine and drug repurposing approach. J Infect Public Health 2025; 18:102763. [PMID: 40153981 DOI: 10.1016/j.jiph.2025.102763] [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: 02/27/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Leprosy and tuberculosis caused by Mycobacterium leprae and Mycobacterium tuberculosis, respectively, are chronic infections with significant public health implications. While leprosy affects the skin and peripheral nerves and tuberculosis primarily targets the lungs, both diseases involve systemic immune responses. This study integrates transcriptomic analysis cheminformatics and molecular dynamics simulations to identify molecular mechanisms and potential therapeutic targets. METHODS Transcriptomic datasets were analyzed to identify dysregulated genes and pathways. Pathway enrichment tissue-specific and bulk RNA-seq expression analyses provided biological context. System biology networks revealed regulatory hub genes and molecular docking studies evaluated CHEMBL compounds as potential therapeutics. Molecular dynamics (MD) simulations assessed the stability of top ligand-protein complexes through RMSD RMSF and MM-GBSA free energy calculations. RESULTS Gene expression analysis identified 13 core dysregulated genes, including HSP90AA1 MAPK8IP3 and ZMPSTE24. Tissue-specific expression localized pivotal genes to lung tissues and immune cells with HSP90AA1 highly expressed in alveolar macrophages and epithelial cells. HSP90AA1 gene emerged as a central hub gene with 96 interactions involved in stress response pathways. Docking studies identified CHEMBL3653862 and CHEMBL3653884 with strong binding affinities (-10.16 to -12.69 kcal/mol) interacting with Asp93 and Tyr139. MD simulations confirmed binding stability with RMSD fluctuations within 2.1-3.5 Å and MM-GBSA energy values supporting ligand-protein stability. CONCLUSION This study identifies HSP90AA1 as a potential drug target in leprosy and tuberculosis. Findings support host-directed therapy approaches and highlight the importance of computational modeling in accelerating drug discovery. The study provides a foundation for future experimental validation, including in vitro and in vivo testing to advance drug repurposing strategies for these chronic infections.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia; Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia.
| | - Ahmed S Alshrari
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Mariah N Hafiz
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Mohammed M Jawad
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia
| | - Fadiyah Jadid Alanazi
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia; Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia
| | | |
Collapse
|
4
|
Lin F, Hu S, Chen J, Li H, Li M, Li R, Xu M, Luo M. MiR-125b suppresses bladder Cancer cell growth and triggers apoptosis by regulating IL-6/IL-6R/STAT3 axis in vitro and in vivo. Cytokine 2025; 190:156926. [PMID: 40120148 DOI: 10.1016/j.cyto.2025.156926] [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: 12/27/2024] [Revised: 03/02/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Bladder cancer (BLCA) is an aggressive malignancy characterized by limited therapeutic options and a poor prognosis. Research has indicated that abnormally expressed miRNAs play a significant role in the pathogenesis of BLCA, although the specific mechanisms remain unclear. MiR-125b plays a tumor suppressor role in a variety of cancers and affects the biological processes of cancer cells such as proliferation, invasion, migration and apoptosis by regulating different signaling pathways. Elucidation of the molecular mechanisms underlying miR-125b may provide clinical therapeutic strategies for bladder cancer. Here, miR-125b was downregulated whereas its targets IL-6R and STAT3 were upregulated in BLCA, as evidenced by bioinformatics analysis. Kaplan-Meier analysis confirmed that miR-125b serves as an independent prognostic factor linked to overall survival (OS) in patients with bladder cancer. Furthermore, overexpression of miR-125b significantly inhibited BLCA cell proliferation, migration, and invasion, while promoting apoptosis, as evidenced by an increased Bax/Bcl-2 ratio and activated cleaved caspase-3. Further investigations demonstrated that miR-125b directly targets and downregulates both IL-6R and STAT3. In a xenograft model, miR-125b overexpression effectively inhibited tumor growth in bladder cancer by blocking IL-6/IL-6R and STAT3 signaling pathways. Collectively, these findings broaden our understanding of the mechanism by which miR-125b acting as a BLCA suppressor in apoptotic regulation by targeting the IL-6/IL-6R/STAT3 signaling pathway, providing novel insights regarding the design of novel miRNA based therapeutic strategies against BLCA.
Collapse
Affiliation(s)
- Fang Lin
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Shaorun Hu
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Jinxiang Chen
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Haiyang Li
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Mengting Li
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Rong Li
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Min Xu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China.
| | - Mao Luo
- Basic Medicine Research Innovation Center for cardiometabolic diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Municipal Key Laboratory of Thrombosis and Vascular Biology, Luzhou, Sichuan, China; Department of Pharmacy, The Second People's Hospital of Yibin, Yibin, Sichuan, China..
| |
Collapse
|
5
|
Cui Q, Fu S, Yu D, Li M, Li Y. Impact of Non-SMC Condensin I Complex Subunit D2 Upregulation on Oral Squamous Cell Carcinoma Prognosis. Int Dent J 2025; 75:1818-1827. [PMID: 40245749 PMCID: PMC12022477 DOI: 10.1016/j.identj.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/23/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE To explore the influence of non-SMC condensin I complex subunit D2 (NCAPD2) on the prognosis of oral squamous cell carcinoma (OSCC) and the correlation between NCAPD2 and OSCC. METHODS In this study, NCAPD2 gene expression profiles of OSCC and normal tissues were collected from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The real-time quantitative polymerase chain reaction (RT-qPCR) was employed to preliminarily validate OSCC cell strains and normal epithelial cell strains. Besides EdU, cell scratch, and transwell assays were performed to assess the proliferation, migration, and invasion of OSCC cell strains with the silence of NCAPD2. Moreover, immunohistochemistry (IHC) staining was utilised to measure the expression of NCAPD2 and tumour-related markers in 74 OSCC specimens. Finally, the Kaplan-Meier analysis was performed to evaluate the influence of NCAPD2 in the prognosis of OSCC. RESULTS The expression of NCAPD2 in OSCC tissues was higher than that in normal tissues. Inhibiting NCAPD2 can reduce the proliferation and migration of OSCC cell lines and inhibit the invasion of these cells. The IHC staining results indicated that the high expression of NCAPD2 in OSCC tissues was positively correlated with T stages, Ki67 expression, and affected sites. The Kaplan-Meier analysis results validated that the up-regulated expression of NCAPD2 was significantly correlated with the poor overall survival (OS) of OSCC patients. CONCLUSION NCAPD2 is a potential molecular marker for the poor prognosis of OSCC, and it is expected to become a target for the treatment of this carcinoma.
Collapse
Affiliation(s)
- Qingying Cui
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shuai Fu
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Diping Yu
- Department of Pathology, Hospital of Pu'er, Kunming University of Science and Technology, Kunming, China
| | - Ming Li
- Department of Oral and Maxillofacial Surgery, Kunming Medical University School and Hospital of Stomatology, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Yong Li
- College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
6
|
Bhattacharya A, Stutvoet TS, Perla M, Loipfinger S, Jalving M, Reyners AKL, Vermeer PD, Drapkin R, de Bruyn M, de Vries EGE, de Jong S, Fehrmann RSN. Transcriptional pattern enriched for synaptic signaling is associated with shorter survival of patients with high-grade serous ovarian cancer. eLife 2025; 13:RP101369. [PMID: 40359002 DOI: 10.7554/elife.101369] [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] [Indexed: 05/15/2025] Open
Abstract
Bulk transcriptomic analyses of high-grade serous ovarian cancer (HGSOC) so far have not uncovered potential drug targets, possibly because subtle, disease-relevant transcriptional patterns are overshadowed by dominant, non-relevant ones. Our aim was to uncover disease-outcome-related patterns in HGSOC transcriptomes that may reveal novel drug targets. Using consensus-independent component analysis, we dissected 678 HGSOC transcriptomes of systemic therapy naïve patients-sourced from public repositories-into statistically independent transcriptional components (TCs). To enhance c-ICA's robustness, we added 447 transcriptomes from non-serous histotypes, low-grade serous, and non-cancerous ovarian tissues. Cox regression and survival tree analysis were performed to determine the association between TC activity and overall survival (OS). Finally, we determined the activity of the OS-associated TCs in 11 publicly available spatially resolved ovarian cancer transcriptomes. We identified 374 TCs, capturing prominent and subtle transcriptional patterns linked to specific biological processes. Six TCs, age, and tumor stage stratified patients with HGSOC receiving platinum-based chemotherapy into ten distinct OS groups. Three TCs were linked to copy-number alterations affecting expression levels of genes involved in replication, apoptosis, proliferation, immune activity, and replication stress. Notably, the TC identifying patients with the shortest OS captured a novel transcriptional pattern linked to synaptic signaling, which was active in tumor regions within all spatially resolved transcriptomes. The association between a synaptic signaling-related TC and OS supports the emerging role of neurons and their axons as cancer hallmark-inducing constituents of the tumor microenvironment. These constituents might offer a novel drug target for patients with HGSOC.
Collapse
Affiliation(s)
- Arkajyoti Bhattacharya
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Thijs S Stutvoet
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mirela Perla
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Stefan Loipfinger
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anna K L Reyners
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Paola D Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, United States
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center and Basser Center for BRCA, University of Pennsylvania, Perelman School of Medicine, Philadelphia, United States
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Li Y, Yu Y, Hu S, Li S. Identification of programmed cell death-related genes and construction of a prognostic model in oral squamous cell carcinoma using single-cell and transcriptome analysis. Discov Oncol 2025; 16:713. [PMID: 40346375 PMCID: PMC12064537 DOI: 10.1007/s12672-025-02520-4] [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: 01/13/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is characterized by poor prognosis and high mortality. Understanding programmed cell death-related genes could provide valuable insights into disease progression and treatment strategies. METHODS RNA-sequencing data from 341 OSCC tumor tissues and 31 healthy samples were analyzed from TCGA database, with validation using 76 samples from GSE41613. Single-cell RNA sequencing data was obtained from GSE172577 (6 OSCC samples). Differentially expressed genes (DEGs) were identified and intersected with 1,254 programmed cell death-related genes. A protein-protein interaction network was constructed, and key modules were identified. Univariate Cox, LASSO, and multivariate Cox regression analyses were performed to build a prognostic model. Model performance was evaluated using Kaplan-Meier analysis, ROC curves, and nomogram validation. RESULTS The study identified 200 candidate genes from the intersection of DEGs and programmed cell death-related genes, which were further refined to 57 hub genes through PPI network analysis. A prognostic signature consisting of five genes (MET, GSDMB, KIT, PRKAG3, and CDKN2A) was established and validated. The model demonstrated good predictive performance in both training and validation cohorts (AUC > 0.6 for 1-, 2-, and 3-year survival). Single-cell analysis revealed that prognostic genes were predominantly expressed in stromal and epithelial cells. Cell communication analysis indicated strong interactions between stromal and epithelial cells. CONCLUSIONS This study developed and validated a novel five-gene prognostic signature for OSCC based on programmed cell death-related genes. The model shows promising clinical application potential for risk stratification and personalized treatment of OSCC patients.
Collapse
Affiliation(s)
- Yongheng Li
- Department of Stomatology, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Harbin, 150001, Heilongjiang Province, China.
| | - Yang Yu
- Department of Stomatology, Qunli Branch, The First Affiliated Hospital of Harbin Medical University, 2075 Qunli Seventh Avenue, Harbin, 150001, Heilongjiang Province, China
| | - Shaonan Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Simin Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| |
Collapse
|
8
|
Jiang Z, Ruan X, Zhou X, Li S, Wang C, Huang L, He Z, Zhang Y, Wen C. Phlorizin attenuates lupus nephritis via upregulating PI3K/Akt pathway-mediated Treg differentiation. Int Immunopharmacol 2025; 154:114607. [PMID: 40186900 DOI: 10.1016/j.intimp.2025.114607] [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: 08/26/2024] [Revised: 03/03/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Lupus nephritis (LN) leads to widespread kidney damage and nephron loss, establishing it as a major contributor to acute and chronic kidney injury, which can progress to end-stage renal disease. Phlorizin (PHZ), a major pharmacologically active constituent derived from Lithocarpus polystachyus Rehd., has been shown to exhibit significant immunomodulatory and anti-inflammatory properties. Growing evidence indicates that PHZ may exert a protective influence on kidney function. However, the therapeutic effect and mechanism of PHZ in treating LN need to be elucidated. METHODS The PHZ-associated targets were identified through tools such as PharmMapper, SwissTargetPrediction, SuperPred and Targetnet. Simultaneously, LN-associated target spots were retrieved fromOMIM, DisGeNET, GeneCards, and GEO databases. Additionally, Venny 2.1.0 was employed to analyze the overlap between drug targets and disease targets. Following this, the DAVID software was employed to perform enrichment analyses for GO terms and KEGG pathways on the shared drug-disease target sites. Following this, the construction of protein-protein interaction (PPI) networks for these intersecting targets was carried out using the STRING database and Cytoscape software, aiming to pinpoint critical targets. Ultimately, molecular docking alongside dynamic simulations was used to evaluate the binding affinity between PHZ and the critical genes. Based on these findings, PHZ or Dexamethasone (DXSM) was administered to female MRL/lpr mice, which are predisposed to lupus. The therapeutic effects of PHZ on LN were evaluated by assessing renal function and the degree of kidney inflammation. Concurrently, flow cytometry was employed to measure the percentage of CD4+ T cell subsets. Additionally, relevant signaling pathways were examined through western blot analysis. Furthermore, CD4+CD25+Foxp3+ regulatory T (Treg) cells were induced in vitro. Flow cytometry and immunoblotting were performed to confirm the role and mechanism of PHZ in Treg cell differentiation. RESULTS The PHZ compound specifically targeted 161 genes associated with LN. PPI analysis revealed that among all the target genes, Akt1, ALB, MMP9, HSP90AA1, and NF-κB1 exhibited the highest centrality. KEGG pathway analysis suggested that the phosphatidylinositol 3 kinase/protein kinase B (PI3K/AKT) signaling pathway could play a crucial role in the treatment of LN. Molecular docking revealed that PHZ exhibits a strong affinity for binding with AKT1. Experimental studies, both in vitro and in vivo, showed that PHZ might alleviate LN by promoting Treg differentiation via activation of the PI3K/AKT signaling pathway. CONCLUSIONS Integrating network pharmacology, bioinformatics, and experimental validation, our study systematically deciphers the therapeutic efficacy and molecular mechanisms of PHZ against LN. Network pharmacology analysis and bioinformatics suggested PI3K/AKT signaling as the pivotal pathway to treat LN, while subsequent in vivo and in vitro experiments confirmed that PHZ exerts its therapeutic effects through activating the PI3K/AKT signaling pathway, ultimately driving FOXP3-dependent regulatory T cell differentiation.
Collapse
Affiliation(s)
- Zhangsheng Jiang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinyi Ruan
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xingchen Zhou
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Suling Li
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chenxi Wang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lin Huang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhixing He
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yun Zhang
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Chengping Wen
- Innovation Center for Medical Basic Research of Autoimmune Diseases, China National Ministry of Education, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
9
|
Sharma A, Tayal S, Bhatnagar S. Analysis of stress response in multiple bacterial pathogens using a network biology approach. Sci Rep 2025; 15:15342. [PMID: 40316612 PMCID: PMC12048639 DOI: 10.1038/s41598-025-91269-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 02/19/2025] [Indexed: 05/04/2025] Open
Abstract
Stress response in bacterial pathogens promotes adaptation, virulence and antibiotic resistance. In this study, a network approach is applied to identify the common central mediators of stress response in five emerging opportunistic pathogens; Enterococcus faecium Aus0004, Staphylococcus aureus subsp. aureus USA300, Klebsiella pneumoniae MGH 78,578, Pseudomonas aeruginosa PAO1, and Mycobacterium tuberculosis H37Rv. A Protein-protein interaction network (PPIN) was constructed for each stressor using Cytoscape3.7.1 from the differentially expressed genes obtained from Gene expression omnibus datasets. A merged PPIN was constructed for each bacterium. Hub-bottlenecks in each network were the central stress response proteins and common pathways enriched in stress response were identified using KOBAS3.0. 31 hub-bottlenecks were common to each individual stress response, merged networks in all five pathogens and an independent cross stress (CS) response dataset of Escherichia coli. The 31 central nodes are in the RpoS mediated general stress regulon and also regulated by other stress response systems. Analysis of the 20 common metabolic pathways modulating stress response in all five bacteria showed that carbon metabolism pathway had the highest crosstalk with other pathways like amino acid biosynthesis and purine metabolism pathways. The central proteins identified can serve as targets for novel wide-spectrum antibiotics to overcome multidrug resistance.
Collapse
Affiliation(s)
- Anjali Sharma
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, 110078, India
| | - Sonali Tayal
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, 110078, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, 110078, India.
| |
Collapse
|
10
|
Zhao C, Lee YT, Melehy A, Kim M, Yang JZ, Zhang C, Kim J, Zhang RY, Lee J, Kim H, Ju Y, Tsai YJ, Zhou XJ, Han SHB, Sadeghi S, Finn RS, Saab S, Lu DS, Chiang J, Park JH, Brennan TV, Wisel SA, Alsudaney M, Kuo A, Ayoub WS, Kim H, Trivedi HD, Wang Y, Vipani A, Kim IK, Todo T, Steggerda JA, Voidonikolas G, Kosari K, Nissen NN, Saouaf R, Singal AG, Sim MS, Elashoff DA, You S, Agopian VG, Yang JD, Tseng HR, Zhu Y. Extracellular vesicle digital scoring assay for assessment of treatment responses in hepatocellular carcinoma patients. J Exp Clin Cancer Res 2025; 44:136. [PMID: 40307890 PMCID: PMC12044846 DOI: 10.1186/s13046-025-03379-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND There are no validated biomarkers for assessing hepatocellular carcinoma (HCC) treatment response (TR). Extracellular vesicles (EVs) are promising circulating biomarkers that may detect minimal residual disease in patients with treated HCC. METHODS We developed the HCC EV TR Score using HCC EV Digital Scoring Assay involving click chemistry-mediated enrichment of HCC EVs, followed by absolute quantification of HCC EV-specific genes by RT-digital PCR. Six HCC EV-specific genes were selected and validated through i) a comprehensive data analysis pipeline with an unprecedentedly large collection of liver transcriptome datasets (n = 9,160), ii) RNAscope validation on HCC tissues (n = 6), and iii) a pilot study on early- or intermediate-stage HCC and liver cirrhosis patients (n = 70). The performance of HCC EV TR Score was assessed in a phase-2 retrospective case-control study (n = 100). RESULTS HCC EV TR Scores, calculated from pre- and post-treatment plasma samples in the phase-2 case-control study, accurately differentiated post-treatment viable from nonviable HCC in the training (area under the ROC curve [AUROC] of 0.90, n = 49) and validation set (AUROC of 0.88, n = 51). At an optimal cutoff of 0.76 identified in the training set, HCC EV TR Score had high accuracy in detecting viable tumors (sensitivity: 76.5%, specificity: 88.2%) and found residual disease not initially observed on MRI in six patients with a median lead time of 63 days. CONCLUSIONS This EV-based digital scoring approach shows great promise to augment cross-sectional imaging for the assessment of HCC treatment response.
Collapse
Grants
- U01 CA230705 NCI NIH HHS
- K08 CA259534 NCI NIH HHS
- R21 CA280444 NCI NIH HHS
- R01 CA255727 NCI NIH HHS
- R01CA277530, R01CA255727, R01CA253651, R01CA253651-04S1, R21CA280444, R01CA246304, U01EB026421, K08CA259534, R44CA288163, U01CA271887, and U01CA230705 NCI NIH HHS
- U01 EB026421 NIBIB NIH HHS
- U01 CA271887 NCI NIH HHS
- R44 CA288163 NCI NIH HHS
- R01 CA277530 NCI NIH HHS
- R01 CA253651 NCI NIH HHS
- R01 CA246304 NCI NIH HHS
- National Cancer Institute
Collapse
Affiliation(s)
- Chen Zhao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
| | - Yi-Te Lee
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Andrew Melehy
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Minhyung Kim
- Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jacqueline Ziqian Yang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Ceng Zhang
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Jina Kim
- Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Ryan Y Zhang
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Junseok Lee
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Hyoyong Kim
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Yong Ju
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Yuan-Jen Tsai
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, 110301, Taiwan
| | - Xianghong Jasmine Zhou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Steven-Huy B Han
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angles (UCLA), Los Angeles, CA, 90095, USA
| | - Saeed Sadeghi
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angles (UCLA), Los Angeles, CA, 90095, USA
| | - Richard S Finn
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angles (UCLA), Los Angeles, CA, 90095, USA
| | - Sammy Saab
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angles (UCLA), Los Angeles, CA, 90095, USA
| | - David S Lu
- Department of Interventional Radiology, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Jason Chiang
- Department of Interventional Radiology, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Jae-Ho Park
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Todd V Brennan
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Steven A Wisel
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Manaf Alsudaney
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Alexander Kuo
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Walid S Ayoub
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Hyunseok Kim
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Hirsh D Trivedi
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Yun Wang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Aarshi Vipani
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Irene K Kim
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Tsuyoshi Todo
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Justin A Steggerda
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Georgios Voidonikolas
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Kambiz Kosari
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Nicholas N Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Rola Saouaf
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Amit G Singal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Myung Shin Sim
- Department of Medicine Statistics Core, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - David A Elashoff
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angles (UCLA), Los Angeles, CA, 90095, USA
| | - Sungyong You
- Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA.
| | - Vatche G Agopian
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
- Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, 90048, USA.
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
| | - Yazhen Zhu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
- Department of Molecular and Medical Pharmacology, California Nanosystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
| |
Collapse
|
11
|
Yabaji SM, Zhernovkov V, Araveti PB, Lata S, Rukhlenko OS, Abdullatif SA, Vanvalkenburg A, Alekseev Y, Ma Q, Dayama G, Lau NC, Johnson WE, Bishai WR, Crossland NA, Campbell JD, Kholodenko BN, Gimelbrant AA, Kobzik L, Kramnik I. Lipid Peroxidation and Type I Interferon Coupling Fuels Pathogenic Macrophage Activation Causing Tuberculosis Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.05.583602. [PMID: 38496444 PMCID: PMC10942339 DOI: 10.1101/2024.03.05.583602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
A quarter of human population is infected with Mycobacterium tuberculosis, but less than 10% of those infected develop pulmonary TB. We developed a genetically defined sst1-susceptible mouse model that uniquely reproduces a defining feature of human TB: the development of necrotic lung granulomas and determined that the sst1-susceptible phenotype was driven by the aberrant macrophage activation. This study demonstrates that the aberrant response of the sst1-susceptible macrophages to prolonged stimulation with TNF is primarily driven by conflicting Myc and antioxidant response pathways leading to a coordinated failure 1) to properly sequester intracellular iron and 2) to activate ferroptosis inhibitor enzymes. Consequently, iron-mediated lipid peroxidation fueled Ifn-beta superinduction and sustained the Type I Interferon (IFN-I) pathway hyperactivity that locked the sst1-susceptible macrophages in a state of unresolving stress and compromised their resistance to Mtb. The accumulation of the aberrantly activated, stressed, macrophages within granuloma microenvironment led to the local failure of anti-tuberculosis immunity and tissue necrosis. The upregulation of Myc pathway in peripheral blood cells of human TB patients was significantly associated with poor outcomes of TB treatment. Thus, Myc dysregulation in activated macrophages results in an aberrant macrophage activation and represents a novel target for host-directed TB therapies.
Collapse
|
12
|
Wang Y, Zheng L, Liu J, Zhang M, Kan Y, Wang W, Yang J. Prognostic role and tumor-suppressive effects of CADM family members and the potential molecular mechanisms of CADM1 in neuroblastoma. Discov Oncol 2025; 16:648. [PMID: 40310517 PMCID: PMC12045911 DOI: 10.1007/s12672-025-02350-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The exact role of cell adhesion molecule (CADM) family members in neuroblastoma is still being explored. Here we uncovered the survival association and the possible mechanisms of CADMs in neuroblastoma through comprehensive bioinformatic analyses. Then the results of CADM1 were verified in neuroblastoma cell lines. METHODS CADMs expression was examined by cBioPortal and TARGET databases and verified in several GEO datasets. Kaplan-Meier plot, log-rank test, the ROC curve, and Cox regression analysis were utilized to assess the prognostic value of CADMs in neuroblastoma. Through functional enrichment analysis and interaction network construction, hub genes were screened to explore the molecular mechanism of CADMs in neuroblastoma. We tested the abilities of cell growth and migration in neuroblastoma cells when CADM1 was silenced and overexpressed respectively. We then used western blot to verify the phosphorylation levels of AKT/GSK-3β pathways. RESULTS The expression of CADM1-4 was significantly down-regulated in neuroblastoma patients with unfavorable prognostic factors. Moreover, CADM1 and CADM3 increased the accuracy of classical clinical indicators for predicting survival rate. The top 10 KEGG pathways for CADMs and their co-expression genes were mainly enriched in the mitotic cell cycle and the process of chromosomal duplication. Furthermore, our study showed that CADM1 inhibited neuroblastoma cells proliferation, migration and the phosphorylation of GSK-3β. CONCLUSIONS Decreased expression of CADM1 and CADM3 was significantly associated with poor outcomes in neuroblastoma. CADM1 may suppress neuroblastoma cell proliferation and migration through regulating the phosphorylation of GSK-3β.
Collapse
Affiliation(s)
- Yu Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Lingling Zheng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jun Liu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Mingyu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Kan
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
13
|
Wan C, Qu Y, Ye Z, Zhang T, Ma H, Chen M, Hou W, Ji Z. Comparative analysis of gene regulation in single cells using Compass. CELL REPORTS METHODS 2025:101035. [PMID: 40345198 DOI: 10.1016/j.crmeth.2025.101035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 05/11/2025]
Abstract
Single-cell multi-omics is a transformative technology that measures both gene expression and chromatin accessibility in individual cells. However, most studies concentrate on a single tissue and are unable to determine whether a gene is regulated by a cis-regulatory element (CRE) in just one tissue or across multiple tissues. We developed Compass for comparative analysis of gene regulation across a large number of human and mouse tissues. Compass consists of a database, CompassDB, and an open-source R software package, CompassR. CompassDB contains processed single-cell multi-omics data of more than 2.8 million cells from hundreds of cell types. Building upon CompassDB, CompassR enables visualization and comparison of gene regulation across multiple tissues. We demonstrated that CompassR can identify CRE-gene linkages specific to a tissue type and their associated transcription factors in real examples.
Collapse
Affiliation(s)
- Changxin Wan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA; Program of Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Yilong Qu
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Zhiyou Ye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Tianbei Zhang
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Huifang Ma
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Ming Chen
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Wenpin Hou
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York City, NY, USA
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA; Program of Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
14
|
Anjum F, Alsharif A, Bakhuraysah M, Shafie A, Hassan MI, Mohammad T. Discovering Novel Biomarkers and Potential Therapeutic Targets of Amyotrophic Lateral Sclerosis Through Integrated Machine Learning and Gene Expression Profiling. J Mol Neurosci 2025; 75:61. [PMID: 40304918 DOI: 10.1007/s12031-025-02340-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that has multiple factors that make its molecular pathogenesis difficult to understand and its diagnosis and treatment during the early stages difficult to determine. Discovering novel biomarkers in ALS for diagnostic and therapeutic potential has become important. Consequently, bioinformatics and machine learning algorithms are useful for identifying differentially expressed genes (DEGs) and potential biomarkers, as well as understanding the molecular mechanisms and intricacies of diseases such as ALS. To achieve the aim of the present study, six datasets obtained from the Gene Expression Omnibus (GEO) were utilized and analyzed using an integrative bioinformatics and machine learning approach. Log transformation was done during data preprocessing, RMA normalization was performed, and the batch effect was corrected. Differential expression analysis identified 206 DEGs that were significantly associated with different biological processes, including muscle function, energy metabolism, and mitochondrial membrane activity. Functional enrichment analysis highlighted pathways, including those related to prion disease, Parkinson's disease, and ATP synthesis via chemiosmotic coupling. We employed a multi-step machine learning framework incorporating random forest, LASSO regression, and SVM-RFE to identify robust biomarkers. This approach identified three key genes, CHRNA1, DLG5, and PLA2G4C, which could be explored as promising biomarkers for ALS after further validation. The internal validation, including principal component analysis (PCA) and ROC-AUC analysis, demonstrated strong diagnostic potential of these hub genes, achieving an AUC of 0.96. This work highlights the utility of bioinformatics and machine learning in identifying key genes as biomarkers for diagnostic and therapeutic potential in ALS.
Collapse
Affiliation(s)
- Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
- King Salman Center for Disability Research, Riyadh, 11614, Saudi Arabia
| | - Abdulaziz Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
- King Salman Center for Disability Research, Riyadh, 11614, Saudi Arabia
| | - Maha Bakhuraysah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
- King Salman Center for Disability Research, Riyadh, 11614, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
15
|
Dou H, Wu R, Wang H, Wang X, Su Y. CCR5 + T cells as a potential biomarker for primary Sjögren's disease based on bioinformatics analysis. Clin Rheumatol 2025:10.1007/s10067-025-07460-6. [PMID: 40293619 DOI: 10.1007/s10067-025-07460-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 04/13/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE To identify and verify potential biomarkers for primary Sjögren's disease (pSjD) using bioinformatics analysis and explore the molecular immune mechanisms of biomarkers.male-to-female ratio of 1:9 METHODS: The pSjD datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differential expression analysis, weighted gene co-expression network analysis (WGCNA) and functional analysis were conducted. PPI network analysis was performed and the hub genes were screened by Cytoscape software. The diagnostic value was assessed by receiver operating characteristic (ROC) analysis. To explore biomarker-immune cell relations, we used CIBERSORT for cell-type identification, combined with scRNA-seq data. Lastly, we validated the expression of the biomarker in human samples. RESULTS A total of 96 overlapping genes, including 1 downregulated and 95 upregulated genes, were obtained. Based on the enrichment analysis, these overlapping genes were mapped to terms related to the functions and regulation of the immune system. CCR5 was identified as a critical biomarker and demonstrated high diagnostic accuracy for pSjD. From CIBERSORT analysis, CCR5 was significantly associated with diverse immune cells. Further scRNA-seq analysis indicated that CCR5 was specifically upregulated in T cells of pSjD salivary gland tissues, which was confirmed in pSjD patients. CONCLUSION Our findings show the role of CCR5 in pSjD, mediated by immune mechanisms. CCR5 is localized in T cells of pSjD salivary glands. Elevated CCR5 expression may be a key biomarker, and increased CCR5 + T cells could aid future diagnosis, prognosis, and treatment of pSjD.
Collapse
Affiliation(s)
- Huixin Dou
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruiqing Wu
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xiaoyan Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Yingying Su
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
16
|
Zhang Z, Zhao T, Meng W, Chen J, He C, Sun X, Huang H. DNA methylation-driven genes in hepatocellular carcinoma patients: insights into immune infiltration and prognostic implications. Front Med (Lausanne) 2025; 12:1520380. [PMID: 40357287 PMCID: PMC12066630 DOI: 10.3389/fmed.2025.1520380] [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: 10/31/2024] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) poses a significant global burden as a highly prevalent and life-threatening malignant tumor that endangers human life and wellbeing. The purpose of this study was to examine how DNA methylation-driven genes impact the prognosis of HCC patients. Methods Differentially expressed genes from The Cancer Genome Atlas, GSE76427, GSE25097 and GSE14520 datasets were collected to perform differential expression analysis between HCC patients and controls. Weighted gene coexpression network analysis (WGCNA) was subsequently performed to create coexpression modules for the DEGs. Then, ssGSEA was employed to investigate the infiltration of immune cells in HCC. Enrichment analysis and methylation were carried out for the module genes. We utilized Kaplan-Meier survival analysis to assess patient prognosis. Results Eight coexpression modules were identified via WGCNA for 1927 upregulated and 1,231 downregulated DEGs, after which the hub genes of the modules were identified. Module 5 had high immune infiltration, and the hub gene SCAMP3 was positively associated with Tcm. Module 3 exhibited a low level of immune infiltration, and the expression of the hub gene HCLS1 was negatively correlated with T cells and dendritic cells. Furthermore, we obtained five hub genes (BOP1, BUB1B, NOTCH3, SCAMP3, and SNRPD2) as methylation-driven genes. BOP1 and BUB1B were found to be correlated with unfavorable overall survival in patients with HCC. Conclusion HCLS1 and SCAMP3 are associated with immunity, whereas BOP1 and BUB1B are modified by methylation and may serve as prognostic markers for HCC.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Hepatobiliary Surger, Guangxi Medical University Affliated Wuming Hospital, Nanning, China
| | - Tongling Zhao
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
| | - Weida Meng
- Department of Hepatobiliary Surger, Guangxi Medical University Affliated Wuming Hospital, Nanning, China
| | - Jiahao Chen
- Department of Hepatobiliary Surger, Guangxi Medical University Affliated Wuming Hospital, Nanning, China
| | - Chengyi He
- Department of Hepatobiliary Surger, Guangxi Medical University Affliated Wuming Hospital, Nanning, China
| | - Xing Sun
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hai Huang
- Department of Hepatobiliary Surger, Guangxi Medical University Affliated Wuming Hospital, Nanning, China
| |
Collapse
|
17
|
Fu Y, Zhang N, Cheng J, Qin X, Zhou X, Du X, Wang Y, Wang J, Zhang D. Identification of novel biomarkers and prognostic model for neuroblastoma using Mendelian randomization and transcriptomic analysis. Discov Oncol 2025; 16:587. [PMID: 40261562 PMCID: PMC12014998 DOI: 10.1007/s12672-025-02414-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the most common extracranial malignant tumor in children, presenting significant challenges in prognosis and treatment stratification. This study aims to identify novel biomarkers for NB and develop a prognostic model using comprehensive analytical methods, including Mendelian randomization (MR) analysis. METHODS Utilizing bioinformatics and Mendelian randomization methods, we explored biomarkers associated with neuroblastoma at the mRNA level. We used chip expression data from the GEO database to screen for differentially expressed genes (DEGs) and conducted two-sample MR analysis using expression quantitative trait loci (eQTL) and neuroblastoma data from the IEU database to identify co-expressed genes through colocalization. A relevant prognostic model was constructed using lasso regression based on the co-expressed genes. Furthermore, we confirmed the correlation between high-risk and low-risk groups with the tumor microenvironment and immune cell infiltration. Subsequently, we evaluated the relationship between risk scores and sensitivity to immunotherapy and anti-tumor drugs. RESULTS Differential analysis identified 485 downregulated and 349 upregulated genes that play important roles in NB. MR analysis identified 4 significant co-expressed genes associated with NB: CAV2, CTSK, LXN, and NDRG2. GO and KEGG enrichment analyses revealed that these genes are involved in crucial biological processes and pathways. A prognostic model based on these four genes was constructed, and its independence as a prognostic factor was confirmed. NB patients were divided into two different risk score groups, with survival analysis indicating that the high-risk group had poorer overall survival, lower immune infiltration, and poorer immune therapy response. In contrast, the low-risk group showed potential efficacy in immunotherapy and higher sensitivity to anti-tumor drugs. CONCLUSION Our findings provide new insights into the molecular basis of NB, identifying four novel biomarkers and developing a risk scoring model based on four co-expressed genes. This model has the potential to become an effective tool for predicting prognosis and guiding treatment in NB patients.
Collapse
Affiliation(s)
- Yongcheng Fu
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Nan Zhang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jian Cheng
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiaohan Qin
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiaoran Du
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuanyuan Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jingyue Wang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Da Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
18
|
Feng B, Guo HY, Ning Y, Zhao YY, Wang X, Cui R. LPCAT3 regulates the immune infiltration and prognosis of ccRCC patients by mediating ferroptosis and endoplasmic reticulum stress. Discov Oncol 2025; 16:574. [PMID: 40253575 PMCID: PMC12009263 DOI: 10.1007/s12672-025-02283-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) accounts for 70% of renal cell carcinoma (RCC) cases. Although surgery remains the mainstay treatment, renal injury and high metastasis rates after nephrectomy dramatically reduce patient quality of life. Drugs that stimulate the immune system by targeting checkpoint pathways improve overall survival in patients with RCC. Here, we investigated the applicability of lysophosphatidylcholine acyltransferase 3 (LPCAT3) as a target for immunotherapy. METHODS In the present study, high LPCAT3 expression in ccRCC was identified using The Cancer Genome Atlas (TCGA) data and validated in two external cohorts from the Gene Expression Omnibus (GEO) database. qRT-PCR was performed to identify the mRNA level of LPCAT3 in tumors and adjacent normal tissues. And immunohistochemistry was used to evaluate the protein level of LPCAT3 between two groups of samples. Furthermore, gene set enrichment analysis was performed to explore the biological processes and pathways related to LPCAT3 expression. Key gene expression and correlation analyses were performed to determine the crosstalk among LPCAT3 expression, ferroptosis, and endoplasmic reticulum stress (ERS). Subsequently, CIBERSORT was used to analyze the immune infiltration status of patients with high and low LPCAT3 expression. RESULTS TCGA and GEO data revealed that LPCAT3 expression in ccRCC tumor tissues was higher than that in adjacent normal tissues; moreover, patients with high LPCAT3 expression had better survival outcomes. qRT-PCR and immunohistochemistry verified the high LPCAT3 expression in tumor tissue. Pathways related to ferroptosis and ERS were upregulated in patients with high LPCAT3 expression. Univariate and multivariate regression analyses revealed that low LPCAT3 levels represent an independent risk factor for ccRCC. LPCAT3 expression was positively correlated with M2 macrophage infiltration levels but negatively correlated with the memory B cell, CD8+ T cell, follicular helper T cell, regulatory T cell, activated natural killer cell, and activated memory CD4+ T cell infiltration levels. CONCLUSIONS LPCAT3was identified as a ccRCC biomarker and may regulate immune infiltration and prognosis in ccRCC by mediating ferroptosis and ERS. Thus, it has potential for exploitation as a prognostic and immune therapeutic target for patients with ccRCC.
Collapse
Affiliation(s)
- Bei Feng
- Department of Nephrology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hai-Ying Guo
- Department of Nephrology, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Ning
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu-Ying Zhao
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiang Wang
- Department of Nephrology, The First People's Hospital in Jinzhou, Dalian, China
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Cui
- Department of Nephrology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
19
|
Yang SQ, Ge YJ, Shen CY. Disclosing antifungal activity of Huangqin decoction upon Trichophyton mentagrophytes and exploring its potential inhibitory mechanisms through transcriptome sequencing and qRT-PCR. Sci Rep 2025; 15:13321. [PMID: 40246952 PMCID: PMC12006297 DOI: 10.1038/s41598-025-97689-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
Trichophyton mentagrophytes (T. mentagrophytes) is a prevalent pathogen that causes human and animal dermatophytosis. The clinical treatment of the infections is challenging due to the prolonged treatment duration, limited efficacy, antifungal resistance and side effects of existing drugs. Modern research has reported that the classic Traditional Chinese medicine (TCM) prescription Huangqin decoction (HQD) along with its principal ingredients could exhibit antifungal properties. Given the valued advantages of TCM such as broad-spectrum antifungal activity, low incidence of drug resistance and low toxicity, this study investigated the antifungal activity of HQD against T. mentagrophytes and explored the potential inhibitory mechanism, aimed to provide new clues for the treatment of dermatophytosis. By detecting minimal inhibitory concentration (MIC) using the broth microdilution method, the results showed that HQD could significantly inhibit the growth of T. mentagrophytes, with a minimal inhibitory concentration (MIC) of 3.13 mg/mL. The transcriptome sequencing and quantitative real-time PCR (qRT-PCR) technology were combined to shed light on the complicated adaptive responses of T. mentagrophytes upon HQD. The results demonstrated that at MIC, compared with the control group, a total of 730 differentially expressed genes (DEGs) were detected in T. mentagrophytes after HQD exposure (FDR adjusted p-value < 0.05), of which 547 were up-regulated and 183 were down-regulated. These DEGs were abundant in "single-organism metabolic process", "catalytic activity" and "oxidoreductase activity", and were significantly enriched in seven signaling pathways including glutathione metabolism, DNA replication, glyoxylate and dicarboxylate metabolism, taurine and hypotaurine metabolism, carotenoid biosynthesis, ubiquitin-mediated proteolysis, and cyanoamino acid metabolism. The results of transcriptome profiling were verified using qRT-PCR for a subset of 10 DEGs. The overall evidence indicated that HQD had a significant anti-dermatophyte activity and the adaptive responses of T. mentagrophytes upon HQD might be related to targeting glutathione S-transferase (GST) gene that could conjugate with toxic xenobiotics to defense oxidative stress, the inhibition of DNA replication pathway by downgrading the DNA replication licensing factors MCM3, MCM5 and ribonuclease H1 (RNaseH1) genes, and the repressed expression of phosphatidylserine decarboxylase (PSD) gene related to phospholipid synthesis which was indispensable for hyphal morphology, hyphal differentiation and cell wall integrity. Our study showed a new theoretical basis for the effective control of T. mentagrophytes infection and the effect of HQD on fungi, which are expected to offer aids for discovering new antifungal agents upon dermatophytosis.
Collapse
Affiliation(s)
- Su-Qing Yang
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, P.R. China
| | - You-Jin Ge
- Nanchang People's Hospital (The Third Hospital of Nanchang), Nanchang, 330009, P.R. China
| | - Cheng-Ying Shen
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, P.R. China.
| |
Collapse
|
20
|
Ye W, Zhang X, Tang Z, Hu Y, Zheng Y, Yuan Y. Comprehensive analysis of glycometabolism-related genes reveals PLOD2 as a prognostic biomarker and therapeutic target in gastric cancer. BMC Gastroenterol 2025; 25:256. [PMID: 40229676 PMCID: PMC11998276 DOI: 10.1186/s12876-025-03878-z] [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: 01/21/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide, with limited therapeutic options and a poor prognosis, particularly in advanced stages. Glycometabolism, a hallmark of cancer, plays a critical role in tumor progression, immune evasion, and response to therapy. However, the specific roles of glycometabolism-related genes and their prognostic and therapeutic implications in GC remain inadequately understood. METHODS Transcriptomic and clinical data from GC patients were retrieved from TCGA and GEO databases. Glycometabolism-related genes were identified and analyzed using machine learning algorithms to construct a prognostic model. Functional assays, immune profiling, and pathway enrichment analyses were performed to explore the roles of these genes in tumor progression, immune-modulatory effects, and drug resistance. PLOD2, the gene with the highest prognostic significance, was further investigated to uncover its underlying regulatory mechanisms, roles in immune modulation, and contribution to therapeutic resistance. RESULTS A glycometabolism-related prognostic model consisting of four genes (PLOD2, CHSY3, SLC2A3 and SLC5A1) was developed and validated, effectively stratifying GC patients into high- and low-risk subgroups with distinct survival outcomes. Among these, PLOD2 emerged as the most significant gene, exhibiting strong associations with tumor progression and poor survival. Functional analyses revealed that PLOD2 promotes glycolysis and tumor progression through activation of the PI3K/AKT/mTOR pathway. Immune profiling revealed that PLOD2 overexpression is associated with an immunosuppressive tumor microenvironment, characterized by increased M2 macrophage infiltration and reduced immune activity. Moreover, treatment with rapamycin, an mTOR inhibitor, significantly suppressed PLOD2-mediated proliferation and anchorage-independent growth in GC cells, highlighting the central role of the PI3K/AKT/mTOR pathway in PLOD2-driven oncogenic behaviors. CONCLUSIONS This study identifies PLOD2 as a key prognostic biomarker and therapeutic target in gastric cancer. As a central component in a glycometabolism-related model, PLOD2 promotes glycolysis, tumor progression, and immune evasion via the PI3K/AKT/mTOR pathway. The model effectively stratifies patient risk, offering both prognostic utility and therapeutic insight. Targeting PLOD2-mediated pathways may represent a promising strategy for precision therapy and improved clinical outcomes in gastric cancer.
Collapse
Affiliation(s)
- Wanchun Ye
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Xiaolei Zhang
- Department of Clinical Laboratory, Jinan Fourth People's Hospital, Jinan, China
| | - Zhongjie Tang
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yufeng Hu
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yuanliang Zheng
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China
| | - Yuping Yuan
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, The Second Affiliated Hospital of Shanghai University, Wenzhou, China.
| |
Collapse
|
21
|
Bao J, Wen J, Chang C, Mu S, Chen J, Shivakumar M, Cui Y, Erus G, Yang Z, Yang S, Wen Z, Zhao Y, Kim D, Duong-Tran D, Saykin AJ, Zhao B, Davatzikos C, Long Q, Shen L. A genetically informed brain atlas for enhancing brain imaging genomics. Nat Commun 2025; 16:3524. [PMID: 40229250 PMCID: PMC11997130 DOI: 10.1038/s41467-025-57636-6] [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: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
Brain imaging genomics has manifested considerable potential in illuminating the genetic determinants of human brain structure and function. This has propelled us to develop the GIANT (Genetically Informed brAiN aTlas) that accounts for genetic and neuroanatomical variations simultaneously. Integrating voxel-wise heritability and spatial proximity, GIANT clusters brain voxels into genetically informed regions, while retaining fundamental anatomical knowledge. Compared to conventional (non-genetics) brain atlases, GIANT exhibits smaller intra-region variations and larger inter-region variations in terms of voxel-wise heritability. As a result, GIANT yields increased regional SNP heritability, enhanced polygenicity, and its polygenic risk score explains more brain volumetric variation than traditional neuroanatomical brain atlases. We provide extensive validation to GIANT and demonstrate its neuroanatomical validity, confirming its generalizability across populations with diverse genetic ancestries and various brain conditions. Furthermore, we present a comprehensive genetic architecture of the GIANT regions, covering their functional annotation at the molecular levels, their associations with other complex traits/diseases, and the genetic and phenotypic correlations among GIANT-defined imaging endophenotypes. In summary, GIANT constitutes a brain atlas that captures the complexity of genetic and neuroanatomical heterogeneity, thereby enhancing the discovery power and applicability of imaging genomics investigations in biomedical science.
Collapse
Affiliation(s)
- Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA
- New York Genome Center (NYGC), New York, NY, USA
| | - Changgee Chang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shizhuo Mu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jiong Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuhan Cui
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhijian Yang
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zixuan Wen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Yize Zhao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Duy Duong-Tran
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Mathematics, United States Naval Academy, Annapolis, MD, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for AI and Data Science for Integrated Diagnostics (AI2D), Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Yue C, Chen B, Pan F, Wang Z, Yu H, Liu G, Li W, Wang R, Tang Y. TCnet: A Novel Strategy to Predict Target Combination of Alzheimer's Disease via Network-Based Methods. J Chem Inf Model 2025; 65:3866-3878. [PMID: 40172120 DOI: 10.1021/acs.jcim.5c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder with an unclear pathogenesis; the traditional ″single gene-single target-single drug″ strategy is insufficient for effective treatment. This study explores a novel strategy for the multitarget therapy of AD by integrating multiomics data and employing network analysis. Different from conventional single-target methods, TCnet adopts a mechanism-driven strategy, utilizing multiomics data to decompose disease mechanisms, construct potential target combinations, and prioritize the optimal combinations using a scoring function. TCnet not only advances our understanding of disease mechanisms but also facilitates large-scale drug screening. This approach was further employed to screen active compounds from Huang-Lian-Jie-Du-Tang (HLJDT), identifying quercetin as a candidate targeting GSK3β and ADAM17. Subsequent in vitro experiments confirmed the neuroprotective and anti-inflammatory effects of quercetin. Overall, TCnet offers a promising approach for predicting target combinations and provides new insights and directions for drug discovery in AD.
Collapse
Affiliation(s)
- Chengyuan Yue
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Baiyu Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Fei Pan
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ze Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Rui Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
23
|
Wu C, Xie X, Yang X, Du M, Lin H, Huang J. Applications of gene pair methods in clinical research: advancing precision medicine. MOLECULAR BIOMEDICINE 2025; 6:22. [PMID: 40202606 PMCID: PMC11982013 DOI: 10.1186/s43556-025-00263-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: 09/06/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025] Open
Abstract
The rapid evolution of high-throughput sequencing technologies has revolutionized biomedical research, producing vast amounts of gene expression data that hold immense potential for biological discovery and clinical applications. Effectively mining these large-scale, high-dimensional data is crucial for facilitating disease detection, subtype differentiation, and understanding the molecular mechanisms underlying disease progression. However, the conventional paradigm of single-gene profiling, measuring absolute expression levels of individual genes, faces critical limitations in clinical implementation. These include vulnerability to batch effects and platform-dependent normalization requirements. In contrast, emerging approaches analyzing relative expression relationships between gene pairs demonstrate unique advantages. By focusing on binary comparisons of two genes' expression magnitudes, these methods inherently normalize experimental variations while capturing biologically stable interaction patterns. In this review, we systematically evaluate gene pair-based analytical frameworks. We classify eleven computational approaches into two fundamental categories: expression value-based methods quantifying differential expression patterns, and rank-based methods exploiting transcriptional ordering relationships. To bridge methodological development with practical implementation, we establish a reproducible analytical pipeline incorporating feature selection, classifier construction, and model evaluation modules using real-world benchmark datasets from pulmonary tuberculosis studies. These findings position gene pair analysis as a transformative paradigm for mining high-dimensional omics data, with direct implications for precision biomarker discovery and mechanistic studies of disease progression.
Collapse
Affiliation(s)
- Changchun Wu
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xueqin Xie
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xin Yang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Mengze Du
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, 611844, China
| | - Hao Lin
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Jian Huang
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| |
Collapse
|
24
|
Asim MN, Ibrahim MA, Zaib A, Dengel A. DNA sequence analysis landscape: a comprehensive review of DNA sequence analysis task types, databases, datasets, word embedding methods, and language models. Front Med (Lausanne) 2025; 12:1503229. [PMID: 40265190 PMCID: PMC12011883 DOI: 10.3389/fmed.2025.1503229] [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: 09/28/2024] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Deoxyribonucleic acid (DNA) serves as fundamental genetic blueprint that governs development, functioning, growth, and reproduction of all living organisms. DNA can be altered through germline and somatic mutations. Germline mutations underlie hereditary conditions, while somatic mutations can be induced by various factors including environmental influences, chemicals, lifestyle choices, and errors in DNA replication and repair mechanisms which can lead to cancer. DNA sequence analysis plays a pivotal role in uncovering the intricate information embedded within an organism's genetic blueprint and understanding the factors that can modify it. This analysis helps in early detection of genetic diseases and the design of targeted therapies. Traditional wet-lab experimental DNA sequence analysis through traditional wet-lab experimental methods is costly, time-consuming, and prone to errors. To accelerate large-scale DNA sequence analysis, researchers are developing AI applications that complement wet-lab experimental methods. These AI approaches can help generate hypotheses, prioritize experiments, and interpret results by identifying patterns in large genomic datasets. Effective integration of AI methods with experimental validation requires scientists to understand both fields. Considering the need of a comprehensive literature that bridges the gap between both fields, contributions of this paper are manifold: It presents diverse range of DNA sequence analysis tasks and AI methodologies. It equips AI researchers with essential biological knowledge of 44 distinct DNA sequence analysis tasks and aligns these tasks with 3 distinct AI-paradigms, namely, classification, regression, and clustering. It streamlines the integration of AI into DNA sequence analysis tasks by consolidating information of 36 diverse biological databases that can be used to develop benchmark datasets for 44 different DNA sequence analysis tasks. To ensure performance comparisons between new and existing AI predictors, it provides insights into 140 benchmark datasets related to 44 distinct DNA sequence analysis tasks. It presents word embeddings and language models applications across 44 distinct DNA sequence analysis tasks. It streamlines the development of new predictors by providing a comprehensive survey of 39 word embeddings and 67 language models based predictive pipeline performance values as well as top performing traditional sequence encoding-based predictors and their performances across 44 DNA sequence analysis tasks.
Collapse
Affiliation(s)
- Muhammad Nabeel Asim
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, Germany
- Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany
| | - Muhammad Ali Ibrahim
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, Germany
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Arooj Zaib
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Andreas Dengel
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, Germany
- Intelligentx GmbH (intelligentx.com), Kaiserslautern, Germany
- Department of Computer Science, Technical University of Kaiserslautern, Kaiserslautern, Germany
| |
Collapse
|
25
|
Mani S, Lalani SR, Pammi M. Genomics and multiomics in the age of precision medicine. Pediatr Res 2025:10.1038/s41390-025-04021-0. [PMID: 40185865 DOI: 10.1038/s41390-025-04021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Precision medicine is a transformative healthcare model that utilizes an understanding of a person's genome, environment, lifestyle, and interplay to deliver customized healthcare. Precision medicine has the potential to improve the health and productivity of the population, enhance patient trust and satisfaction in healthcare, and accrue health cost-benefits both at an individual and population level. Through faster and cost-effective genomics data, next-generation sequencing has provided us the impetus to understand the nuances of complex interactions between genes, diet, and lifestyle that are heterogeneous across the population. The emergence of multiomics technologies, including transcriptomics, proteomics, epigenomics, metabolomics, and microbiomics, has enhanced the knowledge necessary for maximizing the applicability of genomics data for better health outcomes. Integrative multiomics, the combination of multiple 'omics' data layered over each other, including the interconnections and interactions between them, helps us understand human health and disease better than any of them separately. Integration of these multiomics data is possible today with the phenomenal advancements in bioinformatics, data sciences, and artificial intelligence. Our review presents a broad perspective on the utility and feasibility of a genomics-first approach layered with other omics data, offering a practical model for adopting an integrated multiomics approach in pediatric health care and research. IMPACT: Precision medicine provides a paradigm shift from a conventional, reactive disease control approach to proactive disease prevention and health preservation. Phenomenal advancements in bioinformatics, data sciences, and artificial intelligence have made integrative multiomics feasible and help us understand human health and disease better than any of them separately. The genotype-first approach or reverse phenotyping has the potential to overcome the limitations of the phenotype-first approach by identifying new genotype-phenotype associations, enhancing the subclassification of diseases by widening the phenotypic spectrum of genetic variants, and understanding functional mechanisms of genetic variations.
Collapse
Affiliation(s)
- Srinivasan Mani
- Department of Pediatrics, University at Buffalo, Buffalo, NY, USA.
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Mohan Pammi
- Division of Neonatology, Department of Pediatrics, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
26
|
Sun Y, Tang Y, Qi Q, Pang J, Chen Y, Wang H, Liang J, Tang W. 101 Machine Learning Algorithms for Mining Esophageal Squamous Cell Carcinoma Neoantigen Prognostic Models in Single-Cell Data. Int J Mol Sci 2025; 26:3373. [PMID: 40244296 PMCID: PMC11989522 DOI: 10.3390/ijms26073373] [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: 01/24/2025] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignant tumors in the digestive tract, characterized by a high recurrence rate and inadequate immunotherapy options. We analyzed mutation data of ESCC from public databases and employed 10 machine learning algorithms to generate 101 algorithm combinations. Based on the optimal range determined by the concordance index, we randomly selected one combination from the best-performing algorithms to construct a prognostic model consisting of five genes (DLX5, MAGEA4, PMEPA1, RCN1, and TIMP1). By validating the correlation between the prognostic model and antigen-presenting cells (APCs), we revealed the antigen-presentation efficacy of the model. Through the analysis of immune infiltration in ESCC, we uncovered the mechanisms of immune evasion associated with the disease. In addition, we examined the potential impact of the five prognostic genes on ESCC progression. Based on these insights, we identified anti-tumor small-molecule compounds targeting these prognostic genes. This study primarily simulates the tumor microenvironment (TME) and antigen presentation processes in ESCC patients, predicting the role of the neoantigen-based prognostic model in ESCC patients and their potential responses to immunotherapy. These results suggest a potential approach for identifying therapeutic targets in ESCC, which may contribute to the development of more effective treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, No. 727, Jingming South Road, Kunming 650500, China; (Y.S.); (Y.T.); (Q.Q.); (J.P.); (Y.C.); (H.W.); (J.L.)
| |
Collapse
|
27
|
Parikh SJ, Terron HM, Burgard LA, Maranan DS, Butler DD, Wiseman A, LaFerla FM, Lane S, Leissring MA. Targeted Control of Gene Expression Using CRISPR-Associated Endoribonucleases. Cells 2025; 14:543. [PMID: 40214496 PMCID: PMC11988398 DOI: 10.3390/cells14070543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/29/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
CRISPR-associated endoribonucleases (Cas RNases) cleave single-stranded RNA in a highly sequence-specific manner by recognizing and binding to short RNA sequences known as direct repeats (DRs). Here, we investigate the potential of exploiting Cas RNases for the regulation of target genes with one or more DRs introduced into the 3' untranslated region, an approach we refer to as DREDGE (direct repeat-enabled downregulation of gene expression). The DNase-dead version of Cas12a (dCas12a) was identified as the most efficient among five different Cas RNases tested and was subsequently evaluated in doxycycline-regulatable systems targeting either stably expressed fluorescent proteins or an endogenous gene. DREDGE performed superbly in stable cell lines, resulting in up to 90% downregulation with rapid onset, notably in a fully reversible and highly selective manner. Successful control of an endogenous gene with DREDGE was demonstrated in two formats, including one wherein both the DR and the transgene driving expression of dCas12a were introduced in one step by CRISPR-Cas. Our results establish DREDGE as an effective method for regulating gene expression in a targeted, highly selective, and fully reversible manner, with several advantages over existing technologies.
Collapse
Affiliation(s)
- Sagar J. Parikh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Heather M. Terron
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Luke A. Burgard
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Derek S. Maranan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Dylan D. Butler
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Abigail Wiseman
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Frank M. LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Shelley Lane
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| | - Malcolm A. Leissring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697, USA
| |
Collapse
|
28
|
Yang X, Ming Y, Zhou Z, Zhou X, Rao C. Identification of key immune genes of drug-induced liver injury induced by tolvaptan based on bioinformatics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04084-0. [PMID: 40178603 DOI: 10.1007/s00210-025-04084-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Drug-induced liver injury (DILI) poses critical challenges in preclinical drug development and is a primary reason for candidate drug attrition. The incidence of DILI has risen in recent years. While immune-related genes (IRGs) are crucial in immune infiltration, their expression and regulatory mechanisms in tolvaptan-induced DILI remain largely uncharacterized. RNA sequencing data related to DILI and associated clinical data were sourced from the Gene Expression Omnibus (GEO), and IRGs were obtained from the ImmPort database. Differentially expressed genes (DEGs) from DILI and IRGs were intersected to identify differentially expressed immune-related genes (DEIRGs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were used to elucidate the biological functions of DEIRGs. In addition, a protein-protein interaction (PPI) network of DEIRGs was constructed. Immunocytes and immune regulation analyses were conducted using the CIBERSORT tool. Receiver operating characteristic (ROC) curves were constructed to assess the diagnostic accuracy of individual DEIRGs. Networks of transcription factor and microRNA co-regulation were constructed using the NetworkAnalyst database. The expression of DEIRGs in DILI samples was quantified with RT-qPCR. From GSE99878, 204 DEGs were identified, with 23 matching IRGs exhibiting significant expression differences in 17 DEIRGs. The ROC curve analysis suggested satisfactory diagnostic values for six DEIRGs. The potential gene regulatory network comprised 214 microRNAs, 257 transcription factors, and 23 DEIRGs. Finally, RT-qPCR confirmed the expression levels of nine DEIRGs, aligning with public database results. The study revealed numerous immune-related biomarkers, verifying expression in five pivotal genes (ICAM1, CXCL10, IGF1, CX3CL1, and EGFR) and highlighting four genes with notable diagnostic potential (TNFAIP3, BDNF, NR1D2, and PPARA). Additionally, it explored the roles of key biomarkers in inflammatory responses, relevant signaling pathways, and interaction networks, offering new insights into DILI diagnosis, mechanistic understanding, and treatment strategies.
Collapse
Affiliation(s)
- Xiyun Yang
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 611137, China
| | - Yuxuan Ming
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 611137, China
| | - Zhihui Zhou
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 611137, China
| | - Xinyi Zhou
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 611137, China
| | - Chaolong Rao
- School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu Sichuan, 611137, China.
- State Key Laboratory of Traditional Chinese Medicine Resources in Southwest China, Chengdu Sichuan, 611137, China.
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Wenjiang District, Chengdu City, 611137, Sichuan, China.
| |
Collapse
|
29
|
Xie W, Luo Z, Xiao J, Zhang X, Zhang C, Yang P, Li L. Identification of biomarkers related to propionate metabolism in schizophrenia. Front Psychiatry 2025; 16:1504699. [PMID: 40242178 PMCID: PMC12000038 DOI: 10.3389/fpsyt.2025.1504699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
Purpose Schizophrenia (SCZ) is a severe mental disorder with complex etiology. Research shows propionate metabolism is crucial for neurological function and health. This suggests abnormalities in propionate metabolism may link to SCZ. Therefore, identifying biomarkers associated with propionate metabolism might be beneficial for the diagnosis and treatment of SCZ patients. Methods SCZ datasets and propionate metabolism-related genes (PMRGs) from public databases were obtained. DE-PMRGs were identified through differential and correlation analysis of PMRGs. Machine learning was used to screen for key genes and validate expression levels, aiming to identify potential biomarkers. Gene Set Enrichment Analysis (GSEA) and immune infiltration analysis were performed on the biomarkers. An upstream regulatory network was constructed, and potential drugs targeting these biomarkers were explored. Finally, real-time fluorescence quantitative PCR (qPCR) was used to verify biomarker expression levels. Result A total of 11 DE-PMRGs were identified, and machine learning technology was employed to further screen for 5 key genes. Among these, LY96 and TMEM123 emerged as potential biomarkers through expression verification. A diagnostic model was developed, achieving an area under the curve (AUC) greater than 0.7, which indicates strong diagnostic performance. Additionally, nomograms based on these biomarkers demonstrated promising predictive capabilities in assessing the risk of SCZ. To explore gene functions and regulatory mechanisms at a deeper level, a competitive endogenous RNA (ceRNA) regulatory network was constructed, including 2 biomarkers, 72 microRNAs, and 202 long non-coding RNAs. In addition, a regulatory network containing 2 biomarkers and 104 transcription factors (TFs) was also established to investigate the transcription factors interacting with the biomarkers. Potential biomarker-targeted drugs were identified by exploring the DrugBank database; notably, LY96 exhibited higher binding affinities for four drugs, with docking scores consistently below-5 kcal/mol. The qPCR results indicated that the expression levels of LY96 and TMEM123 in the whole blood of SCZ patients were significantly higher than those in the healthy control group, which was consistent with the results in the GSE38484 and GSE27383 datasets. Conclusion This study identified disease diagnostic biomarkers associated with propionate metabolism in SCZ, specifically LY96 and TMEM123. These findings offer novel perspectives for the diagnosis and management of SCZ.
Collapse
Affiliation(s)
| | | | | | | | | | - Ping Yang
- School of Clinical Medicine, Hunan Brain Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Liang Li
- School of Clinical Medicine, Hunan Brain Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
30
|
Zhang N, Tian X, Liu F, Jin X, Zhang J, Hao L, Jiang S, Liu Q. Reversal of sorafenib resistance in hepatocellular carcinoma by curcumol: insights from network pharmacology, molecular docking, and experimental validation. Front Pharmacol 2025; 16:1514997. [PMID: 40242448 PMCID: PMC12000033 DOI: 10.3389/fphar.2025.1514997] [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: 10/22/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Background Curcumol, a bioactive sesquiterpenoid extracted from traditional Chinese medicine (TCM), has demonstrated potential in overcoming tumor drug resistance. However, its mechanisms in reversing drug resistance, particularly in hepatocellular carcinoma (HCC) resistant to sorafenib, are not yet fully elucidated. This study aims to explore the molecular mechanisms by which curcumol reverses sorafenib resistance in HCC using a combination of network pharmacology, molecular docking, and in vivo and in vitro experiments. Methods We identified curcumol targets and genes associated with sorafenib-resistant HCC, resulting in a set of overlapping targets. These intersection targets underwent enrichment analysis using DAVID, and a protein-protein interaction (PPI) network was constructed via the STRING database and Cytoscape. Molecular docking confirmed the binding of curcumol to core targets. In vitro assays, including CCK-8, colony formation assay, apoptosis detection, wound healing, and Transwell assays, evaluated curcumol's effects on sorafenib-resistant HCC cells. Western blotting assessed the impact on PI3K/AKT and JAK/STAT3 signaling pathways. Additionally, a sorafenib-resistant HCC xenograft mouse model was established to observe the in vivo efficacy of curcumol combined with sorafenib. Results We identified 117 potential targets for curcumol in reversing sorafenib resistance in HCC. Among them, five core targets-ALB, STAT3, HSP90AA1, HSP90AB1, and SRC-showed strong binding affinity with curcumol. KEGG pathway analysis of the intersecting genes highlighted significant involvement of the PI3K/AKT, JAK/STAT3, Ras, Rap1, HIF-1, FoxO, and mTOR signaling pathways. In vitro experiments revealed that curcumol significantly enhanced the sensitivity of sorafenib-resistant HCC cells to sorafenib, inhibiting cell proliferation, invasion, and migration while promoting apoptosis. In vivo studies further confirmed that curcumol combined with sorafenib effectively inhibited tumor growth in sorafenib-resistant HCC models. Conclusion This study provides compelling evidence that curcumol can reverse sorafenib resistance in HCC by modulating multiple signaling pathways, including PI3K/AKT and JAK/STAT3 pathways. Our findings suggest that curcumol holds promise as a novel therapeutic agent for overcoming drug resistance in HCC, offering a new avenue for clinical intervention.
Collapse
Affiliation(s)
- Ni Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, Shandong, China
| | - Fen Liu
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaohan Jin
- Jining No. 1 People’s Hospital, Shandong First Medical University, Jining, China
- Center for Post-Doctoral Studies, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaqi Zhang
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, Shandong, China
| | - Lingli Hao
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, Shandong, China
| | - Shulong Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, Shandong, China
| | - Qingbin Liu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
31
|
Xiong Y, Li J, Jin W, Sheng X, Peng H, Wang Z, Jia C, Zhuo L, Zhang Y, Huang J, Zhai M, Lyu B, Sun J, Zhou M. PCMR: a comprehensive precancerous molecular resource. Sci Data 2025; 12:551. [PMID: 40169679 PMCID: PMC11961594 DOI: 10.1038/s41597-025-04899-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/25/2025] [Indexed: 04/03/2025] Open
Abstract
Early detection and intervention of precancerous lesions are crucial in reducing cancer morbidity and mortality. Comprehensive analysis of genomic, transcriptomic, proteomic and epigenomic alterations can provide insights into the early stages of carcinogenesis. However, the lacke of an integrated, well-curated data resource of molecular signatures limits our understanding of precancerous processes. Here, we introduce a comprehensive PreCancerous Molecular Resource (PCMR), which compiles 25,828 molecular profiles of precancerous samples paired with normal or malignant counterparts. These profiles cover precancerous lesions of 35 cancer types across 20 organs and tissues, derived from tissue samples, liquid biopsies, cell lines and organoids, with data from transcriptomics, proteomics and epigenomics. PCMR includes 62,566 precancer-gene associations derived from differential analysis and text-mining using the ChatGPT large language model. We examined PCMR dataset reliability and significance by the authoritative precancerous molecular signature, along with its biological and clinical relevance. Overall, PCMR will serve as a valuable resource for advancing precancer research and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Yichun Xiong
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Jiaqi Li
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Wang Jin
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Xiaoran Sheng
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Hui Peng
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Zhiyi Wang
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Caifeng Jia
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Lili Zhuo
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Yibo Zhang
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Jingzhe Huang
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Modi Zhai
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Beibei Lyu
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China
| | - Jie Sun
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China.
| | - Meng Zhou
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, P. R. China.
| |
Collapse
|
32
|
Danaeifar M, Najafi A. Artificial Intelligence and Computational Biology in Gene Therapy: A Review. Biochem Genet 2025; 63:960-983. [PMID: 38635012 DOI: 10.1007/s10528-024-10799-1] [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: 08/16/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
One of the trending fields in almost all areas of science and technology is artificial intelligence. Computational biology and artificial intelligence can help gene therapy in many steps including: gene identification, gene editing, vector design, development of new macromolecules and modeling of gene delivery. There are various tools used by computational biology and artificial intelligence in this field, such as genomics, transcriptomic and proteomics data analysis, machine learning algorithms and molecular interaction studies. These tools can introduce new gene targets, novel vectors, optimized experiment conditions, predict the outcomes and suggest the best solutions to avoid undesired immune responses following gene therapy treatment.
Collapse
Affiliation(s)
- Mohsen Danaeifar
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Science, P.O. Box 19395-5487, Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Science, P.O. Box 19395-5487, Tehran, Iran.
| |
Collapse
|
33
|
Dong K, Ye Z, Hu F, Shan C, Wen D, Cao J. An evolutionary dynamics analysis of the plant DEK gene family reveals the role of BnaA02g08940D in drought tolerance. Int J Biol Macromol 2025; 298:140053. [PMID: 39828179 DOI: 10.1016/j.ijbiomac.2025.140053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
DEK is a chromatin protein that interacts with DNA to influence chromatin formation, thereby affecting plant growth, development, and stress response. This study investigates the molecular evolution of the DEK family in plants, with a particular focus on the Brassica species. A total of 127 DEK genes were identified in 34 plants and classified into seven groups based on the phylogenetic analysis. The distribution of motifs and gene structure is similar within each group, indicating a high degree of conservation. The results of the collinearity analysis indicated that the DEK protein has undergone a certain degree of evolutionary conservation. The expansion of the DEK family is primarily attributable to whole-genome duplication (WGD) or segmental duplication events. The DEK protein has undergone purification during its evolutionary history, and several positively selected sites have been identified. Moreover, the examination of cis-acting elements and expression patterns revealed that the BnDEKs play a significant role in plant growth and stress response. The protein-protein interaction network identified several noteworthy proteins that interact with DEK. These analyses enhance our comprehension of the DEK gene family and establish the foundation for additional validation of its function. Further research demonstrated that the overexpression of one DEK family member, BnaA02g08940D, enhanced the transgenic Arabidopsis tolerance to drought and osmosis. This indicates that the DEK family may respond when plants are subjected to drought stress, thereby strengthening the plant's resilience.
Collapse
Affiliation(s)
- Kui Dong
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Ziyi Ye
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Fei Hu
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Chaofan Shan
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Dongyu Wen
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Jun Cao
- School of Life Sciences, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
34
|
Hussain A, Mohammad T, Khan S, Alajmi MF, Yadav DK, Hassan MI. Seven Hub Genes Associated with Huntington's Disease and Diagnostic and Therapeutic Potentials Identified by Computational Biology. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025; 29:154-163. [PMID: 40059764 DOI: 10.1089/omi.2025.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Huntington's disease (HD) is characterized by progressive motor dysfunction and cognitive decline. Early diagnosis and new therapeutic targets are essential for effective interventions. We performed integrative analyses of mRNA profiles from three microarrays and one RNA-seq dataset from the Gene Expression Omnibus database. The datasets included were GSE8762, GSE24250, GSE45516, and GSE64810. Data pre-processing included background correction, normalization, log2 transformation, probe-to-gene symbol mapping, and differential expression analysis. We identified 80 differentially expressed genes (DEGs) based on a significance threshold (p < 0.05) and absolute log fold change (logFC) >0.65. Additionally, we conducted Gene Ontology (GO) and pathway analyses of the identified genes. Protein-protein interactions among DEGs revealed a network from which seven hub genes (VIM, COL1A1, COL3A1, COL1A2, DCN, CXCR2, and S100A9) were identified using the cytoHubba plugin in Cytoscape software. Two top DEGs, IGHG1 (up-regulated) and PITX1 (up-regulated), also hold potential as therapeutic targets. Insofar as biological contextualization of the findings is concerned, the top enriched GO terms were skeletal system development, blood vessel development, and vasculature development. Molecular function terms highlighted signaling receptor binding, extracellular matrix structural constituent, and platelet-derived growth factor binding. Notably, the significant KEGG pathways included amoebiasis, the AGE-RAGE signaling pathway in diabetic complications, and the relaxin signaling pathway. In conclusion, the present computational biology integrative analyses of multiple datasets discovered new DEGs and seven hub genes, shedding light on molecular mechanisms of HD. These findings call for translational clinical omics research and may potentially lead to future precision medicine interventions and novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Shumayila Khan
- International Health Division, Indian Council of Medical Research, New Delhi, India
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Gachon University, Yeonsu-gu Incheon, Republic of Korea
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
35
|
Clarke DJ, Evangelista JE, Xie Z, Marino GB, Byrd AI, Maurya MR, Srinivasan S, Yu K, Petrosyan V, Roth ME, Milinkov M, King CH, Vora JK, Keeney J, Nemarich C, Khan W, Lachmann A, Ahmed N, Agris A, Pan J, Ramachandran S, Fahy E, Esquivel E, Mihajlovic A, Jevtic B, Milinovic V, Kim S, McNeely P, Wang T, Wenger E, Brown MA, Sickler A, Zhu Y, Jenkins SL, Blood PD, Taylor DM, Resnick AC, Mazumder R, Milosavljevic A, Subramaniam S, Ma’ayan A. Playbook workflow builder: Interactive construction of bioinformatics workflows. PLoS Comput Biol 2025; 21:e1012901. [PMID: 40179105 PMCID: PMC11967941 DOI: 10.1371/journal.pcbi.1012901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
The Playbook Workflow Builder (PWB) is a web-based platform to dynamically construct and execute bioinformatics workflows by utilizing a growing network of input datasets, semantically annotated API endpoints, and data visualization tools contributed by an ecosystem of collaborators. Via a user-friendly user interface, workflows can be constructed from contributed building-blocks without technical expertise. The output of each step of the workflow is added into reports containing textual descriptions, figures, tables, and references. To construct workflows, users can click on cards that represent each step in a workflow, or construct workflows via a chat interface that is assisted by a large language model (LLM). Completed workflows are compatible with Common Workflow Language (CWL) and can be published as research publications, slideshows, and posters. To demonstrate how the PWB generates meaningful hypotheses that draw knowledge from across multiple resources, we present several use cases. For example, one of these use cases prioritizes drug targets for individual cancer patients using data from the NIH Common Fund programs GTEx, LINCS, Metabolomics, GlyGen, and ExRNA. The workflows created with PWB can be repurposed to tackle similar use cases using different inputs. The PWB platform is available from: https://playbook-workflow-builder.cloud/.
Collapse
Affiliation(s)
- Daniel J.B. Clarke
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - John Erol Evangelista
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Zhuorui Xie
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Giacomo B. Marino
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Anna I. Byrd
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Mano R. Maurya
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Sumana Srinivasan
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Keyang Yu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Varduhi Petrosyan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Matthew E. Roth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | | | - Charles Hadley King
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Jeet Kiran Vora
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Jonathon Keeney
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Christopher Nemarich
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - William Khan
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Alexander Lachmann
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Nasheath Ahmed
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Alexandra Agris
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Juncheng Pan
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Srinivasan Ramachandran
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Eoin Fahy
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Emmanuel Esquivel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | | | - Bosko Jevtic
- Persida Inc., Brooklyn, New York, United States of America
| | - Vuk Milinovic
- Persida Inc., Brooklyn, New York, United States of America
| | - Sean Kim
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Patrick McNeely
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Tianyi Wang
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Eric Wenger
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Miguel A. Brown
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Alexander Sickler
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Yuankun Zhu
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Sherry L. Jenkins
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Philip D. Blood
- Pittsburgh Supercomputing Center, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Deanne M. Taylor
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Adam C. Resnick
- Department of Biomedical and Health Informatics; Department of Pediatrics, The Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Data Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Raja Mazumder
- Department of Biochemistry and Molecular Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC, United States of America
| | - Aleksandar Milosavljevic
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Shankar Subramaniam
- Department of Bioengineering, University of California San Diego, La Jolla, California, United States of America
| | - Avi Ma’ayan
- Department of Pharmacological Sciences, Windreich Department of Artificial Intelligence and Human Health, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
36
|
Hameed AR, Fakhri Ali S, N Almanaa T, Aljasir MA, Alruwetei AM, Sanami S, Ayaz H, Ali I, Ahmad F, Ahmad S. Exploring the hub genes and potential drugs involved in Fanconi anemia using microarray datasets and bioinformatics analysis. J Biomol Struct Dyn 2025; 43:3297-3310. [PMID: 38149868 DOI: 10.1080/07391102.2023.2297008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023]
Abstract
Fanconi anemia (FA) is a genetic disorder that occurs when certain genes responsible for repairing DNA replication and promoting homologous recombination fail to function properly. This leads to severe clinical symptoms and a wide range of cancer-related characteristics. Recent treatment approaches for FA involve hematopoietic stem cell transplantation (HSCT), which helps restore the population of stem cells. A survival study using p-values indicated that specific hub genes play a significant role in diagnosing and predicting the disease. To find potential medications that interact with the identified hub genes, researchers inferred drugs. Among hub genes, TP53 was found to be particularly promising through computational analysis. Further investigation focused on two drugs, Topiramate and Tocofersolan predicted based on drug bank database analysis. Molecular docking strategies were employed to assess the best binding pose of these drugs with TP53. Topiramate showed a binding affinity of -6.5 kcal/mol, while Tocofersolan showed -8.5 kcal/mol against the active residues within the binding pocket. Molecular dynamics (MD) simulations were conducted to observe the stability of each drug's interaction with the TP53 protein over time. Both drugs exhibited stable confirmation with only slight changes in the loop region of the TP53 protein during the simulation intervals. Results also shows that there was a high fluctuation observed during apo-sate simulation time intervals as compared to complex system. Hence, it is suggested that the exploration of structure-based drug design holds promising results to specific target. This could potentially lead to a breakthrough in future experimental approaches for FA treatment.
Collapse
Affiliation(s)
- Alaa R Hameed
- Department of Medical Laboratory Techniques, School of Life Sciences, Dijlah University College, Baghdad, Iraq
| | - Sama Fakhri Ali
- Department of Anesthesia Techniques, School of Life Sciences, Dijlah University College, Baghdad, Iraq
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Abdullah Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abdulmohsen M Alruwetei
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim, Saudi Arabia
| | - Samira Sanami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hassan Ayaz
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Pakistan
| | - Ijaz Ali
- Center for Applied Mathematics and Bioinformatics (CAMB), Gulf University for Science and Technology, West Mishref, Kuwait
| | - Faisal Ahmad
- Foundation University Medical College, Foundation University Islamabad, Islamabad, Pakistan
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Department of Natural Sciences, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
37
|
Chen Y, Zou J. Simple and effective embedding model for single-cell biology built from ChatGPT. Nat Biomed Eng 2025; 9:483-493. [PMID: 39643729 DOI: 10.1038/s41551-024-01284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 10/16/2024] [Indexed: 12/09/2024]
Abstract
Large-scale gene-expression data are being leveraged to pretrain models that implicitly learn gene and cellular functions. However, such models require extensive data curation and training. Here we explore a much simpler alternative: leveraging ChatGPT embeddings of genes based on the literature. We used GPT-3.5 to generate gene embeddings from text descriptions of individual genes and to then generate single-cell embeddings by averaging the gene embeddings weighted by each gene's expression level. We also created a sentence embedding for each cell by using only the gene names ordered by their expression level. On many downstream tasks used to evaluate pretrained single-cell embedding models-particularly, tasks of gene-property and cell-type classifications-our model, which we named GenePT, achieved comparable or better performance than models pretrained from gene-expression profiles of millions of cells. GenePT shows that large-language-model embeddings of the literature provide a simple and effective path to encoding single-cell biological knowledge.
Collapse
Affiliation(s)
- Yiqun Chen
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
- Department of Computer Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
38
|
Sanfilippo C, Castrogiovanni P, Imbesi R, Vecchio M, Vinciguerra M, Blennow K, Zetterberg H, Di Rosa M. Sex-specific modulation of FOLR1 and its cycle enzyme genes in Alzheimer's disease brain regions. Metab Brain Dis 2025; 40:163. [PMID: 40153031 DOI: 10.1007/s11011-025-01578-x] [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: 08/11/2024] [Accepted: 03/13/2025] [Indexed: 03/30/2025]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive cognitive and functional decline. Its incidence increases significantly with age and is more prevalent in women than men. We investigated the folate receptor alpha (FOLR1) gene expression levels in the central nervous system (CNS) of AD and non-demented healthy control (NDHC) subjects. Our cohort included 3,946 samples: 2,391 NDHC and 1,555 AD patients, stratified by brain region, age, and sex. Interestingly, a significant increase in FOLR1 expression was observed only in females with AD compared to NDHC females. Furthermore, we found that FOLR1 expression was differentially increased in the prefrontal cortex (PFC) and diencephalon (DIE) only in AD females. Moreover, in females, genes involved in the folic acid (FA) cycle that drives DNA synthesis were significantly modulated. In contrast, in males, downregulation of TYMS effectively blocks the completion of the cycle, thereby preventing downstream DNA synthesis. Tissue Transcriptome Deconvolution (TTD) analysis revealed astrocytes and endothelial cells associated with FOLR1 expression in both AD males and females. Gene Ontology analysis supported these findings, showing enrichment in processes aligned with these cell types. Positive correlations between brain FOLR1 expression and markers for astrocytes (glial fibrillary acidic protein) and endothelial cells (CD31) provided further validation. Our findings suggest a potential role for sex-dependent FOLR1 expression and its association with specific brain regions and cellular processes in AD.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- Neurologic Unit, AOU "Policlinico-San Marco", Department of Medical, Surgical Sciences and Advanced Technologies, GF, Ingrassia, University of Catania, Via Santa Sofia n.78, Catania, Sicily, 95100, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Michele Vecchio
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute, Medical University Varna, Varna, Bulgaria
- Liverpool Centre for Cardiovascular Science, Faculty of Health, Liverpool John Moores University, Liverpool, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer'S Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
39
|
Luo T, Guo W, Ji W, Du W, Lv Y, Feng Z. Monocyte CCL2 signaling possibly contributes to increased asthma susceptibility in type 2 diabetes. Sci Rep 2025; 15:10768. [PMID: 40155667 PMCID: PMC11953320 DOI: 10.1038/s41598-025-95039-1] [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: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
In recent years, the respiratory system has been increasingly recognized as a key target organ in diabetes. Although observational studies have established significant clinical associations between type 2 diabetes (T2D), antidiabetic medication use, and asthma, the causal relationships and underlying molecular mechanisms remain unclear. This study employed a bidirectional two-sample Mendelian randomization (MR) approach combined with bioinformatics analysis to explore the causal relationships between T2D and asthma subtypes and complications, with a focus on immune-regulatory mechanisms. The MR analysis utilized inverse-variance weighted (IVW) and meta-analysis methods to evaluate overall effects, with sensitivity analyses confirming the robustness of the findings. Bioinformatics analysis focused on differential gene expression and pathway enrichment to identify potential molecular networks. The MR analysis showed that T2D has a significant positive causal effect on asthma (P < 0.05), with severe autoimmune T2D showing strong associations with specific asthma subtypes (eosinophilic and mixed asthma) and complications (e.g., acute respiratory infections and pneumonia) (P < 0.05). Bioinformatics analysis identified the monocyte-CCL2 signaling axis as a key mechanism linking T2D and asthma, where hyperglycemia-induced monocyte activation may promote asthma development. These findings reveal shared inflammatory pathways and deepen our understanding of the molecular mechanisms linking these two chronic diseases.
Collapse
Affiliation(s)
- Tian Luo
- Department of Respiratory and Critical Care Medicine, The People's Hospital of Sishui, Jining, 273200, Shandong, China
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Weihong Guo
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Wentao Ji
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - WeiWei Du
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Yanhua Lv
- Department of Respiratory and Critical Care Medicine, Shunde Hospital of Southern Medical University, Shunde, 528300, Guangdong, China.
| | - Zhijun Feng
- Postdoctoral Innovation Practice Base, Jiangmen Central Hospital, Southern Medical University, Jiangmen, 529030, Guangdong, China.
| |
Collapse
|
40
|
Abid J, Al-Rawi MBA, Mahmood A, Li A, Jiang T. Identification and functional characterization of key biomarkers in diffuse large B-cell lymphoma: emphasis on STYX as a prognostic marker and therapeutic target. Hereditas 2025; 162:45. [PMID: 40128844 PMCID: PMC11931869 DOI: 10.1186/s41065-025-00411-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: 11/23/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBC) is the most common subtype of non-Hodgkin lymphoma, characterized by its aggressive nature and poor prognosis in advanced stages. Despite advances in treatment, the molecular mechanisms driving DLBC progression remain incompletely understood, necessitating the identification of novel biomarkers for diagnosis and prognosis. In this study, we analyzed two publicly available datasets (GSE32018 and GSE56315) from the Gene Expression Omnibus database (GEO) to identify overlapping differentially expressed genes (DEGs). Later on, a comprehensive in silico and in vitro methodology was adopted to decipher the role of identify DEGs in DLBC. DEGs analysis of GSE32018 and GSE56315 datasets identified five overlapping gene: SP3, CSNK1A1, STYX, SIRT5, and MGEA5. Expression validation using the GEPIA2 database confirmed the upregulation of SP3, CSNK1A1, STYX, and SIRT5, and the downregulation of MGEA5 in DLBC tissues compared to normal controls. Furthermore, mutational analysis revealed that CSNK1A1 was the only gene among these DEGs to exhibit mutations, with a 2.7% mutation frequency in DLBC patients. Methylation analysis highlighted a negative correlation between DEGs methylation levels and mRNA expression, while survival analysis identified high STYX expression as significantly associated with poorer overall survival in DLBC patients. Functional assays demonstrated that STYX knockdown in U2932 cells led to reduced cell proliferation, colony formation, and enhanced wound healing, indicating STYX's pivotal role in DLBC cell survival and migration. Additionally, gene enrichment analysis revealed the involvement of these DEGs in key biological processes, including intracellular trafficking and myeloid progenitor cell differentiation. These findings emphasize the potential of SP3, CSNK1A1, STYX, SIRT5, and MGEA5 as biomarkers and therapeutic targets in DLBC, particularly highlighting STYX as a promising prognostic marker and potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Junaid Abid
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300222, China
| | - Mahmood Basil A Al-Rawi
- Department of Optometry, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Ahmad Mahmood
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - An Li
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Urumqi, 830054, China.
| | - Tiemin Jiang
- Department of Hepatobiliary & Hydatid Diseases, Digestive & Vascular Surgery Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Urumqi, 830054, China.
| |
Collapse
|
41
|
Roberson EDO. The small genomics lab experience optimizing data cold storage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643355. [PMID: 40166252 PMCID: PMC11956953 DOI: 10.1101/2025.03.18.643355] [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
Translational research is often a collaborative enterprise that involves basic science researchers, clinicians, and experts in genomics and bioinformatics. While there are central university and industry cores to support data generation, long-term storage often falls to the individual investigators. We frequently fulfill the role of long-term FASTQ file storage for our collaborators. To reduce our cold storage space, we tested the space savings for gzip and zstandard algorithms on an old set of FASTQ files. We found that zstandard had a better overall compression ratio than the best gzip algorithm, amounting to more than 20% space savings overall compared to gzip. It may be worth transitioning to zstandard compression for small, collaborative genomics labs to minimize cold storage costs.
Collapse
Affiliation(s)
- Elisha D. O. Roberson
- Washington University in St. Louis, Departments of Medicine & Genetics, Division of Rheumatology, St. Louis, MO 63110
| |
Collapse
|
42
|
Loganathan T, George Priya Doss C. Computational molecular insights into ibrutinib as a potent inhibitor of HER2-L755S mutant in breast cancer: gene expression studies, virtual screening, docking, and molecular dynamics analysis. Front Mol Biosci 2025; 12:1510896. [PMID: 40177517 PMCID: PMC11962039 DOI: 10.3389/fmolb.2025.1510896] [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: 10/15/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Background The proposed study integrates several advanced computational techniques to unravel the molecular mechanisms underlying breast cancer progression and drug resistance. Methods We investigated HER2-L755S mutation through a multi-step approach, including gene expression analysis, molecular docking, and molecular dynamics simulations. Results and Discussion By conducting a network-based analysis of gene expression data from breast cancer samples, key hub genes such as MYC, EGFR, CDKN2A, ERBB2, CDK1, E2F1, TOP2A, MDM2, TGFB1, and FOXM1 were identified, all of which are critical in tumor growth and metastasis. The study mainly focuses on the ERBB2 gene, which encodes the HER2 protein, and its common mutation HER2-L755S, associated with breast cancer and resistance to the drug lapatinib. The HER2-L755S mutation contributes to both tumorigenesis and therapeutic failure. To address this, alternative therapeutic strategies were investigated using combinatorial computational approaches. The stability and flexibility of the HER2-L755S mutation were evaluated through comparative molecular dynamics simulations over 1000 ns using Gromacs in the unbound (Apo) state. Virtual screening with Schrodinger Glide identified ibrutinib as a promising alternative to lapatinib for targeting the HER2-L755S mutant. Detailed docking and molecular dynamics simulations in the bound (Holo) state demonstrated that the HER2-L755S-ibrutinib complex exhibited higher binding affinity and lower binding energy, indicating more stable interactions compared to other complexes. MM-PBSA analysis revealed that the HER2-L755S-ibrutinib complex had more negative binding energy than the HER2-L755S-afatinib, HER2-L755S-lapatinib, and HER2-L755S-neratinib complexes, suggesting that ibrutinib forms the most stable complex with favorable binding interactions. Conclusion These results provide in-depth atomic-level insights into the binding mechanisms of these inhibitors, highlighting ibrutinib as a potentially effective inhibitor for the clinical treatment of breast cancer.
Collapse
Affiliation(s)
| | - C. George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
43
|
Tian BX, Yu ZX, Qiu X, Chen LP, Zhuang YL, Chen Q, Gu YH, Hou MJ, Gu YF. Development and validation of a 14-CpG DNA methylation signature and drug targets for prognostic prediction in breast cancer. Front Med (Lausanne) 2025; 12:1548726. [PMID: 40177272 PMCID: PMC11961922 DOI: 10.3389/fmed.2025.1548726] [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: 12/20/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Background Breast cancer (BC) is the most prevalent cancer among women and a leading cause of cancer-related deaths worldwide. Emerging evidence suggests that DNA methylation, a well-studied epigenetic modification, regulates various cellular processes critical for cancer development and progression and holds promise as a biomarker for cancer diagnosis and prognosis, potentially enhancing the efficacy of precision therapies. Methods We developed a robust prognostic model for BC based on DNA methylation and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). We analyzed the association of the model with clinicopathological features, survival outcomes, and chemotherapy drug sensitivity. Results A set of 216 differentially methylated CpGs was identified by intersecting three datasets (TCGA, GSE22249, and GSE66695). Using univariate Cox proportional hazard and LASSO Cox regression analyses, we constructed a 14-CpG model significantly associated with progression-free interval (PFI), disease-specific survival (DSS), and overall survival (OS) in BC patients. Kaplan-Meier (KM) survival analysis, receiver operating characteristic (ROC) analysis, and nomogram validation confirmed the clinical value of the signature. The Cox analysis showed a significant association between the signature and PFI and DSS in BC patients. KM analysis effectively distinguished high-risk from low-risk patients, while ROC analysis demonstrated high sensitivity and specificity in predicting BC prognosis. A nomogram based on the signature effectively predicted 5- and 10-year PFI and DSS. Additionally, combining our model with clinical risk factors suggested that patients in the I-II & M+ subgroup could benefit from adjuvant chemotherapy regarding PFI, DSS, and OS. Gene Ontology (GO) functional enrichment and KEGG pathway analyses indicated that the top 3,000 differentially expressed genes (DEGs) were enriched in pathways related to DNA replication and repair and cell cycle regulation. Patients in the high-risk group might benefit from drugs targeting DNA replication and repair processes in tumor cells. Conclusion The 14-CpG model serves as a useful biomarker for predicting prognosis in BC patients. When combined with TNM staging, it offers a potential strategy for individualized clinical decision-making, guiding personalized therapeutic regimen selection for clinicians.
Collapse
Affiliation(s)
- Bao-xing Tian
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-xi Yu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Qiu
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-ping Chen
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-lian Zhuang
- Department of Nursing, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Chen
- Department of Nursing, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-hua Gu
- Department of Nursing, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng-jie Hou
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-fan Gu
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Yousefpour Shahrivar R, Karami F, Karami E. Differential gene expression patterns in Niemann-Pick Type C and Tay-Sachs diseases: Implications for neurodegenerative mechanisms. PLoS One 2025; 20:e0319401. [PMID: 40106490 PMCID: PMC11922228 DOI: 10.1371/journal.pone.0319401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/01/2025] [Indexed: 03/22/2025] Open
Abstract
Lysosomal storage disorders (LSDs) are a group of rare genetic conditions characterized by the impaired function of enzymes responsible for lipid digestion. Among these LSDs, Tay-Sachs disease (TSD) and Niemann-Pick type C (NPC) may share a common gene expression profile. In this study, we conducted a bioinformatics analysis to explore the gene expression profile overlap between TSD and NPC. Analyses were performed on RNA-seq datasets for both TSD and NPC from the Gene Expression Omnibus (GEO) database. Datasets were subjected to differential gene expression analysis utilizing the DESeq2 package in the R programming language. A total of 147 differentially expressed genes (DEG) were found to be shared between the TSD and NPC datasets. Enrichment analysis was then performed on the DEGs. We found that the common DEGs are predominantly associated with processes such as cell adhesion mediated by integrin, cell-substrate adhesion, and urogenital system development. Furthermore, construction of protein-protein interaction (PPI) networks using the Cytoscape software led to the identification of four hub genes: APOE, CD44, SNCA, and ITGB5. Those hub genes not only can unravel the pathogenesis of related neurologic diseases with common impaired pathways, but also may pave the way towards targeted gene therapy of LSDs.In addition, they serve as the potential biomarkers for related neurodegenerative diseases warranting further investigations.
Collapse
Affiliation(s)
- Ramin Yousefpour Shahrivar
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Fatemeh Karami
- Department of Medical Genetics, Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ebrahim Karami
- Department of Electrical and Computer Engineering, Faculty of Engineering and Applied Sciences, Memorial University of Newfoundland, St. John's, Canada
| |
Collapse
|
45
|
Brown GS, Wengler J, Fabelico AJS, Muir A, Tubbs A, Warren A, Millett AN, Yu XX, Pavlidis P, Rogic S, Piccolo SR. Using semantic search to find publicly available gene-expression datasets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643153. [PMID: 40161731 PMCID: PMC11952526 DOI: 10.1101/2025.03.13.643153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Millions of high-throughput, molecular datasets have been shared in public repositories. have been shared in public repositories. Researchers can reuse such data to validate their own findings and explore novel questions. A frequent goal is to find multiple datasets that address similar research topics and to either combine them directly or integrate inferences from them. However, a major challenge is finding relevant datasets due to the vast number of candidates, inconsistencies in their descriptions, and a lack of semantic annotations. This challenge is first among the FAIR principles for scientific data. Here we focus on dataset discovery within Gene Expression Omnibus (GEO), a repository containing 100,000s of data series. GEO supports queries based on keywords, ontology terms, and other annotations. However, reviewing these results is time-consuming and tedious, and it often misses relevant datasets. We hypothesized that language models could address this problem by summarizing dataset descriptions as numeric representations (embeddings). Assuming a researcher has previously found some relevant datasets, we evaluated the potential to find additional relevant datasets. For six human medical conditions, we used 30 models to generate embeddings for datasets that human curators had previously associated with the conditions and identified other datasets with the most similar descriptions. This approach was often, but not always, more effective than GEO's search engine. Our top-performing models were trained on general corpora, used contrastive-learning strategies, and used relatively large embeddings. Our findings suggest that language models have the potential to improve dataset discovery, perhaps in combination with existing search tools.
Collapse
Affiliation(s)
- Grace S. Brown
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | - James Wengler
- Department of Biology, Brigham Young University, Provo, Utah, USA
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
| | | | - Abigail Muir
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | - Anna Tubbs
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | - Amanda Warren
- Department of Biology, Brigham Young University, Provo, Utah, USA
| | - Alexandra N. Millett
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xinrui Xiang Yu
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul Pavlidis
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sanja Rogic
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
46
|
Liu H, Liu Y, Zhao Y, Ma Y, Chen Q, Xu H, Wang X, Guo X, Wang H, Chen Z, Zhang S, Han B. A scoping review of human genetic resources management policies and databases in high- and middle-low-income countries. BMC Med Ethics 2025; 26:37. [PMID: 40089739 PMCID: PMC11909912 DOI: 10.1186/s12910-025-01192-7] [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: 08/30/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND This review examines global human genetic resources management, focusing on genetic data policies and repositories in high- and middle-low-income countries. METHODS A comprehensive search strategy was employed across multiple databases, including official government websites and Google, to gather relevant literature on human genetic resources management policies and genetic resource databases. Documents were screened for relevance, focusing on high-income countries (United States, United Kingdom, Japan) and middle-low-income countries (China, India, Kenya). Data were extracted, coded, and analyzed to identify common themes and differences in genetic resource management practices. RESULTS High-income countries benefit from robust legal frameworks and advanced technological infrastructures. The United States enforces the Health Insurance Portability and Accountability Act and the Genetic Information Nondiscrimination Act to protect privacy and facilitate data sharing, while Japan relies on the Act on the Protection of Personal Information and ethical guidelines. Additionally, high-income countries host a variety of genetic databases and biobanks that support scientific research. In contrast, middle-low-income countries like China, India, and Kenya are still developing their frameworks. China has regulations such as the Biosecurity Law and the Regulations on the Management of Human Genetic Resources, but still requires more unified standards. India's policies focus on genetic research and data protection through the Biological Diversity Act, while Kenya seeks to improve data management through the 2019 Data Protection Act. CONCLUSION Significant disparities exist in human genetic resources management between high-income and middle-low-income countries. High-income countries have robust systems balancing privacy protection with research facilitation, supported by comprehensive and large-scale databases for scientific research. Middle-low-income countries need to enhance legal frameworks and build population-specific databases. Promoting equitable data sharing and adopting best practices from high-income countries are essential for advancing global scientific discovery and ensuring fair management of genetic resources.
Collapse
Affiliation(s)
- Hongwei Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yin Liu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yanyan Zhao
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Henan Province Engineering Research Center of Artificial Intelligence and Internet of Things Wise Medical, Zhengzhou, China
| | - Yingqi Ma
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Qiong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Huifang Xu
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiaoyang Wang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiaoli Guo
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Hong Wang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Henan Province Engineering Research Center of Artificial Intelligence and Internet of Things Wise Medical, Zhengzhou, China
| | - Shaokai Zhang
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Binbin Han
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
47
|
Shen F, Liu X, Ding F, Yu Z, Shi X, Cheng L, Zhang X, Jing C, Zhao Z, Cao H, Zhao B, Liu J. Integrative bioinformatics analysis of high-throughput sequencing and in vitro functional analysis leads to uncovering key hub genes in esophageal squamous cell carcinoma. Hereditas 2025; 162:38. [PMID: 40087784 PMCID: PMC11908063 DOI: 10.1186/s41065-025-00398-4] [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: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCA) is a type of cancer that starts in the cells lining the esophagus, the tube connecting the throat to the stomach. It is known for its aggressive nature and poor prognosis. Understanding the key factors that drive this cancer is crucial for developing better diagnostic tools and treatments. METHODS Gene expression profiles of ESCA were analyzed using Gene Expression Omnibus (GEO) datasets (GSE23400, GSE29001, GSE92396, and GSE1420) from the GEO database. Differentially expressed genes (DEGs) were identified using the limma package, and a protein-protein interaction (PPI) network was constructed using the STRING database. Hub genes were identified based on the degree method. Further validation was performed through reverse transcription quantitative PCR (RT-qPCR), mutational and copy number variation (CNV) analysis via the cBioPortal database, promoter methylation analysis using the OncoDB and GSCA databases, survival analysis, immune infiltration analysis through the GSCA database, and functional assays, including knockdown of key genes. RESULTS We identified four key hub genes, COL3A1, COL4A1, COL5A2, and CXCL8 that play significant roles in ESCA. These genes were highly expressed in ESCA tissues and cell lines, with expression levels significantly (p-value < 0.001) elevated compared to normal controls. Receiver operating characteristic (ROC) curve analysis revealed exceptional diagnostic performance for all four genes, with area under the curve (AUC) values of 1.0, indicating perfect sensitivity and specificity in distinguishing ESCA from normal controls. Mutational analysis revealed that COL3A1 was altered in 67% of ESCA samples, primarily through missense mutations, while COL5A2 exhibited alterations in 50% of the samples, including splice site and missense mutations. Additionally, gene amplification patterns were observed in all four hub genes, further validating their oncogenic potential in ESCA progression. A significant (p-value < 0.05) promoter hypomethylation was detected in these genes, suggesting a potential regulatory role in their expression. Functional assays demonstrated that knocking down COL3A1 and COL4A1 led to decreased cell proliferation, colony formation, and migration, indicating their critical roles in tumor progression. Additionally, these genes were involved in pathways related to the extracellular matrix and immune system modulation. CONCLUSION COL3A1, COL4A1, COL5A2, and CXCL8 are crucial in ESCA development and progression, particularly in remodeling the extracellular matrix, modulating the immune system, and promoting metastasis. These findings suggest that these genes could serve as potential biomarkers for diagnosing ESCA and targets for future therapies. Future research should focus on in vivo validation of these findings and clinical testing to assess the therapeutic potential of targeting these genes in ESCA treatment.
Collapse
Affiliation(s)
- Feng Shen
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Xing Liu
- Oncology Department, Ankang City Central Hospital, Ankang, 725000, China
| | - Fengjiao Ding
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Zhonglin Yu
- Department of Thoracic Surgery, Ankang Central Hospital, Ankang, 725000, China
| | - Xinyi Shi
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Lushan Cheng
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Xuewei Zhang
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Chengbao Jing
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Zilong Zhao
- Pathology Department, Ankang City Central Hospital, Ankang, 725000, China
| | - Hongyou Cao
- Oncology Department, People's Hospital of Ankang City, Ankang, 725000, China
| | - Bing Zhao
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China
| | - Jing Liu
- Clinical Laboratory, Ankang City Central Hospital, Ankang, 725000, China.
| |
Collapse
|
48
|
Durairaj P, Liu ZL. Brain Cytochrome P450: Navigating Neurological Health and Metabolic Regulation. J Xenobiot 2025; 15:44. [PMID: 40126262 PMCID: PMC11932283 DOI: 10.3390/jox15020044] [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: 01/07/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Human cytochrome P450 (CYP) enzymes in the brain represent a crucial frontier in neuroscience, with far-reaching implications for drug detoxification, cellular metabolism, and the progression of neurodegenerative diseases. The brain's complex architecture, composed of interconnected cell types and receptors, drives unique neuronal signaling pathways, modulates enzyme functions, and leads to distinct CYP gene expression and regulation patterns compared to the liver. Despite their relatively low levels of expression, brain CYPs exert significant influence on drug responses, neurotoxin susceptibility, behavior, and neurological disease risk. These enzymes are essential for maintaining brain homeostasis, mediating cholesterol turnover, and synthesizing and metabolizing neurochemicals, neurosteroids, and neurotransmitters. Moreover, they are key participants in oxidative stress responses, neuroprotection, and the regulation of inflammation. In addition to their roles in metabolizing psychotropic drugs, substances of abuse, and endogenous compounds, brain CYPs impact drug efficacy, safety, and resistance, underscoring their importance beyond traditional drug metabolism. Their involvement in critical physiological processes also links them to neuroprotection, with significant implications for the onset and progression of neurodegenerative diseases. Understanding the roles of cerebral CYP enzymes is vital for advancing neuroprotective strategies, personalizing treatments for brain disorders, and developing CNS-targeting therapeutics. This review explores the emerging roles of CYP enzymes, particularly those within the CYP1-3 and CYP46 families, highlighting their functional diversity and the pathological consequences of their dysregulation on neurological health. It also examines the potential of cerebral CYP-based biomarkers to improve the diagnosis and treatment of neurodegenerative disorders, offering new avenues for therapeutic innovation.
Collapse
Affiliation(s)
- Pradeepraj Durairaj
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Chemical and Biomedical Engineering, Florida A&M University, Tallahassee, FL 32310, USA
| | - Zixiang Leonardo Liu
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Chemical and Biomedical Engineering, Florida A&M University, Tallahassee, FL 32310, USA
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32310, USA
| |
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
|
Parmar G, Chudasama JM, Shah A, Aundhia C, Kardani S. Targeting cell cycle arrest in breast cancer by phytochemicals from Caryto urens L. fruit ethyl acetate fraction: in silico and in vitro validation. J Ayurveda Integr Med 2025; 16:101095. [PMID: 40081286 PMCID: PMC11932863 DOI: 10.1016/j.jaim.2024.101095] [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: 06/04/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Caryota urens, also known as Shivjata, has been documented in ancient Indian texts for its therapeutic benefits, addressing conditions from seminal weakness to gastric ulcers. This study aims to investigate its contemporary medicinal potential in treating breast cancer. OBJECTIVES The study focuses on exploring the therapeutic potential of Caryota urens fruit against breast cancer, specifically targeting cell cycle genes CDK1, CDC25A, and PLK1 through bioinformatics, network pharmacology, and in vitro validation. MATERIALS AND METHODS Using mass spectrometry and nuclear magnetic resonance (NMR), 60 key phytoconstituents from Caryota urens fruit were identified. Bioinformatics analysis, integrating Gene Cards and GEO databases, 15,474 breast cancer-associated genes focusing on the HR+/HER2-subtype were identified. Molecular docking and qPCR validated the interactions of key phytoconstituents, particularly Episesamin, with CDK1, CDC25A, and PLK1. In vitro studies were conducted on the MCF7 cell line, supplemented by ROC and survival analyses to evaluate diagnostic and therapeutic potential. RESULTS The bioinformatics analysis identified CDK1, CDC25A, and PLK1 as pivotal genes regulating cell cycle progression and breast cancer tumorigenesis. Network pharmacology and in vitro studies indicated that phytoconstituents, especially Episesamin, downregulated these genes in breast cancer cells. Molecular docking and qPCR confirmed these interactions, and ROC and survival analyses underscored their diagnostic and therapeutic significance. CONCLUSIONS This study suggests that Caryota urens fruit extract, particularly Episesamin, may inhibit breast cancer metastasis by downregulating CDK1, CDC25A, and PLK1, offering promising new strategies for targeting the cell cycle in breast cancer and emphasizing the value of integrating bioinformatics with experimental methods in cancer research.
Collapse
Affiliation(s)
- Ghanshyam Parmar
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India.
| | - Jay Mukesh Chudasama
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Ashish Shah
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Chintan Aundhia
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| | - Sunil Kardani
- Department of Pharmacy, Sumandeep Vidyapeeth Deemed to be University, Piparia, Waghodia, Vadodara, 391760, Gujarat, India
| |
Collapse
|