1
|
Ma Y, Boycott C, Zhang J, Gomilar R, Yang T, Stefanska B. SIRT1/DNMT3B-mediated epigenetic gene silencing in response to phytoestrogens in mammary epithelial cells. Epigenetics 2025; 20:2473770. [PMID: 40029260 PMCID: PMC11881848 DOI: 10.1080/15592294.2025.2473770] [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: 10/29/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
We performed an integrated analysis of genome-wide DNA methylation and expression datasets in normal cells and healthy animals exposed to polyphenols with estrogenic activity (i.e. phytoestrogens). We identified that phytoestrogens target genes linked to disrupted cellular homeostasis, e.g. genes limiting DNA break repair (RNF169) or promoting ribosomal biogenesis (rDNA). Existing evidence suggests that DNA methylation may be governed by sirtuin 1 (SIRT1) deacetylase via interactions with DNA methylating enzymes, specifically DNMT3B. Since SIRT1 was reported to be regulated by phytoestrogens, we test whether phytoestrogens suppress genes related to disrupted homeostasis via SIRT1/DNMT3B-mediated transcriptional silencing. Human MCF10A mammary epithelial cells were treated with phytoestrogens, pterostilbene (PTS) or genistein (GEN), followed by analysis of cell growth, DNA methylation, gene expression, and SIRT1/DNMT3B binding. SIRT1 occupancy at the selected phytoestrogen-target genes, RNF169 and rDNA, was accompanied by consistent promoter hypermethylation and gene downregulation in response to GEN, but not PTS. GEN-mediated hypermethylation and SIRT1 binding were linked to a robust DNMT3B enrichment at RNF169 and rDNA promoters. This was not observed in cells exposed to PTS, suggesting a distinct mechanism of action. Although both SIRT1 and DNMT3B bind to RNF169 and rDNA promoters upon GEN, the two proteins do not co-occupy the regions. Depletion of SIRT1 abolishes GEN-mediated decrease in rDNA expression, suggesting SIRT1-dependent epigenetic suppression of rDNA by GEN. These findings enhance our understanding of the role of SIRT1-DNMT3B interplay in epigenetic mechanisms mediating the impact of phytoestrogens on cell biology and cellular homeostasis.
Collapse
Affiliation(s)
- Yuexi Ma
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Cayla Boycott
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Jiaxi Zhang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Rekha Gomilar
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Tony Yang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Naidovski N, Chong SKT, Liu F, Riordan SM, Wehrhahn MC, Yuwono C, Zhang L. Human macrophage response to the emerging enteric pathogen Aeromonas veronii: Inflammation, apoptosis, and downregulation of histones. Virulence 2025; 16:2440554. [PMID: 39663607 DOI: 10.1080/21505594.2024.2440554] [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: 08/05/2024] [Revised: 11/11/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
This study investigated the pathogenic mechanisms of Aeromonas veronii in macrophages. THP-1 derived macrophages were used as a human macrophage model and were treated with A. veronii strain AS1 isolated from intestinal biopsies of an IBD patient, or Escherichia coli strain K-12. RNA was extracted and subjected to RNA sequencing and comparative transcriptomic analyses. Protein levels of IL-8, IL-1β, IL-18, and TNFα were measured using ELISA, and apoptosis was assessed using caspase 3/7 assays. Both A. veronii AS1 and E. coli K-12 significantly upregulated the expression of many genes involving inflammation. At the protein level, A. veronii AS1 induced significantly higher levels of IL-8, TNFα, mature IL-18 and IL-1β than E. coli K-12, and led to greater elevation of caspase 3/7 activities. Both A. veronii AS1 and E. coli K-12 upregulated the expression of CASP5, but not other caspase genes. A. veronii AS1 significantly downregulated the expression of 20 genes encoding histone proteins that E. coli K-12 did not. The more profound pathogenic effects of A. veronii in inducing inflammation and apoptosis in macrophages than E. coli K-12 are consistent with its role as a human enteric pathogen. The upregulated expression of CASP5 and increased release of IL-1β and IL-18 support the role of CASP5 in activation of non-canonical inflammasome. The downregulation of histone genes by A. veronii suggests a unique impact on host cell gene expression, which may represent a novel virulence strategy. These findings advance the understanding of pathogenic mechanisms of the emerging human enteric pathogen A. veronii.
Collapse
Affiliation(s)
- Nicholas Naidovski
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Sarah K T Chong
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Fang Liu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Stephen M Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital, University of New South Wales, Sydney, Australia
| | - Michael C Wehrhahn
- Douglass Hanly Moir Pathology, a Sonic Healthcare Practice, Macquarie Park, NSW, Australia
| | - Christopher Yuwono
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
3
|
Uusi-Mäkelä M, Harjula SKE, Junno M, Sillanpää A, Nätkin R, Niskanen MT, Saralahti AK, Nykter M, Rämet M. The inflammasome adaptor pycard is essential for immunity against Mycobacterium marinum infection in adult zebrafish. Dis Model Mech 2025; 18:dmm052061. [PMID: 39916610 PMCID: PMC11972081 DOI: 10.1242/dmm.052061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/03/2025] [Indexed: 03/25/2025] Open
Abstract
Inflammasomes regulate the host response to intracellular pathogens including mycobacteria. We have previously shown that the course of Mycobacterium marinum infection in adult zebrafish (Danio rerio) mimics the course of tuberculosis in human. To investigate the role of the inflammasome adaptor pycard in zebrafish M. marinum infection, we produced two zebrafish knockout mutant lines for the pycard gene with CRISPR/Cas9 mutagenesis. Although the zebrafish larvae lacking pycard developed normally and had unaltered resistance against M. marinum, the loss of pycard led to impaired survival and increased bacterial burden in the adult zebrafish. Based on histology, immune cell aggregates, granulomas, were larger in pycard-deficient fish than in wild-type controls. Transcriptome analysis with RNA sequencing of a zebrafish haematopoietic tissue, kidney, suggested a role for pycard in neutrophil-mediated defence, haematopoiesis and myelopoiesis during infection. Transcriptome analysis of fluorescently labelled, pycard-deficient kidney neutrophils identified genes that are associated with compromised resistance, supporting the importance of pycard for neutrophil-mediated immunity against M. marinum. Our results indicate that pycard is essential for resistance against mycobacteria in adult zebrafish.
Collapse
Affiliation(s)
- Meri Uusi-Mäkelä
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | | | - Maiju Junno
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Alina Sillanpää
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| | - Reetta Nätkin
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, FI-33521 Tampere, Finland
| | | | | | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
- Tays Cancer Center, Tampere University Hospital, FI-33521 Tampere, Finland
| | - Mika Rämet
- Faculty of Medicine and Health Technology, Tampere University, FI-33014 Tampere, Finland
| |
Collapse
|
4
|
Khalife M, Jia T, Caron P, Shreim A, Genoux A, Cristini A, Pucciarelli A, Leverve M, Lepeltier N, García-Rodríguez N, Dalonneau F, Ramachandran S, Fernandez Martinez L, Marcion G, Lemaitre N, Brambilla E, Garrido C, Hammond E, Huertas P, Gazzeri S, Sordet O, Eymin B. SRSF2 overexpression induces transcription-/replication-dependent DNA double-strand breaks and interferes with DNA repair pathways to promote lung tumor progression. NAR Cancer 2025; 7:zcaf011. [PMID: 40181846 PMCID: PMC11963763 DOI: 10.1093/narcan/zcaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 02/04/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
SRSF2 (serine/arginine-rich splicing factor 2) is a critical regulator of pre-messenger RNA splicing, which also plays noncanonical functions in transcription initiation and elongation. Although elevated levels of SRSF2 are associated with advanced stages of lung adenocarcinoma (LUAD), the mechanisms connecting SRSF2 to lung tumor progression remain unknown. We show that SRSF2 overexpression increases global transcription and replicative stress in LUAD cells, which correlates with the production of DNA damage, notably double-strand breaks (DSBs), likely resulting from conflicts between transcription and replication. Moreover, SRSF2 regulates DNA repair pathways by promoting homologous recombination and inhibiting nonhomologous end joining. Mechanistically, SRSF2 interacts with and enhances MRE11 (meiotic recombination 11) recruitment to chromatin, while downregulating 53BP1 messenger RNA and protein levels. Both events are likely contributing to SRSF2-mediated DNA repair process rerouting. Lastly, we show that SRSF2 and MRE11 expression is commonly elevated in LUAD and predicts poor outcome of patients. Altogether, our results identify a mechanism by which SRSF2 overexpression promotes lung cancer progression through a fine control of both DSB production and repair. Finally, we show that SRSF2 knockdown impairs late repair of ionizing radiation-induced DSBs, suggesting a more global function of SRSF2 in DSB repair by homologous recombination.
Collapse
Affiliation(s)
- Manal Khalife
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Tao Jia
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Pierre Caron
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Amani Shreim
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Aurelie Genoux
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Agnese Cristini
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Amelie Pucciarelli
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Marie Leverve
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Nina Lepeltier
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Néstor García-Rodríguez
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Fabien Dalonneau
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Shaliny Ramachandran
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Lara Fernandez Martinez
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Guillaume Marcion
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Nicolas Lemaitre
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Elisabeth Brambilla
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Carmen Garrido
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Ester M Hammond
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Pablo Huertas
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Sylvie Gazzeri
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Olivier Sordet
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Beatrice Eymin
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| |
Collapse
|
5
|
Wang Y, Wang C, Gong Y, Li Q, Liu M, Sun H. GIT2 negatively regulates the NF-κB pathway directly or indirectly by regulating TRAF3 expression to promote osteogenic differentiation of BMSCs. Tissue Cell 2025; 94:102790. [PMID: 39954559 DOI: 10.1016/j.tice.2025.102790] [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: 10/05/2024] [Revised: 01/23/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND AIMS Osteoporosis (OP) is a common disease of aging, which is closely related to the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). DNA damage, as a senescence-associated secretory phenotype (SASP), plays an important role in aging diseases including OP. GIT2 has been identified as a DNA repair gene and alleviates aging-related phenotypes. However, the relationship between GIT2 and osteogenic differentiation of BMSCs remains unclear. METHODS Here, we used bioinformatics analysis to identify the gene GIT2, which is closely related to aging, OP and DNA damage, and its downstream targets. Then, H2O2 -induced BMSCs senescence model and ovariectomy-induced mice OP model was established in vitro and in vivo, respectively. Micro-CT, H&E staining, toluidine blue staining, and calcein double labeling were used to analyze bone mass, osteogenic differentiation phenotype, and bone formation rate. Comet assay, Elisa and immunofluorescence were used to analyze senescence-related phenotypes. Western blotting was used to detect the protein levels of GIT2/TRAF3/NF-κB axis and osteogenesis-related markers. RESULTS Our results showed that GTI2 and TRAF3 were positively correlated with OP-related markers. On the one hand, GIT2 could inhibit the activation of both canonical and non-canonical NF-κB signaling pathways by positively regulating TRAF3. On the other hand, GIT2 could directly bind to P65, a component of the classical NF-κB signaling pathway, and P52, a component of the non-classical NF-κB signaling pathway, to inhibit their activation, improve DNA damage repair, alleviate cell senescence, and further promote osteogenic differentiation of BMSCs. CONCLUSIONS In summary, the present study demonstrates that GIT2 plays a crucial regulatory role in promoting osteogenic differentiation of BMSCs, which provides new ideas for the prevention and treatment of OP and other aging-related diseases.
Collapse
Affiliation(s)
- Yanna Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, Lvshunkou District 116044, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, Lvshunkou District 116044, China
| | - Ying Gong
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Dalian, Xigang District 116011, China
| | - Qingchen Li
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Dalian, Xigang District 116011, China
| | - Mozhen Liu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, No. 222, Zhongshan Road, Dalian, Xigang District 116011, China.
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, Lvshunkou District 116044, China.
| |
Collapse
|
6
|
Srivastava P, Rai A, Kumar M. Network analysis of differentially expressed genes involved in oral submucous fibrosis and oral squamous cell carcinoma: a comparative approach. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 139:583-593. [PMID: 39779388 DOI: 10.1016/j.oooo.2024.12.016] [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: 09/08/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE Oral submucous fibrosis (OSMF) is categorized as an oral potentially malignant disorder (OPMD) with an increased risk of occurrence of oral squamous cell carcinoma (OSCC). In this study, we aimed to identify the hub genes associated with OSMF and OSCC. STUDY DESIGN Using RStudio, a set of differentially expressed genes (DEGs) were identified in (A) OSMF, (B) OSCC, and (C) comparative OSMF-OSCC category, obtained from Gene Expression Omnibus (GEO). The Protein to Protein Interaction (PPI) Network, hub genes, and functional annotation were determined using Search Tool for the Retrieval of Interacting Genes (STRING), Cytoscape, and SR-Plot, Database for Annotation, Visualization and Integrated Discovery (DAVID). RESULTS A total of 2081, 2320, and 3295 DEGs were obtained from the OSMF, OSCC, and comparative categories, respectively. Hub gene and gene enrichment analysis revealed that the genes in (A) MYH6, TTN, TNNT3, MYL1, TPM2, ACTN3, NEB, MYL2, TNNT1, and TPM1; (B) CD4, SELL, CD28, CD27, PRF1, CD80, GZMB, CD40LG, ITGAX, and IL4; and (C) CD4, CD8A, CTLA4, CD28, GZMB, IL79, CD69, CD40LG, IFNG, and CD80 categories, were associated with muscle contraction, cell proliferation, and malignant transformation. CONCLUSIONS Hub genes and functional enrichment analysis revealed the diagnostic genes and the genes responsible for the malignant transformation in OSMF, OSCC, and the comparative category. A panel of identified genes will be of clinical significance in targeted therapy in future studies. (Oral Surg Oral Med Oral Pathol Oral Radiol YEAR;VOL:page range).
Collapse
Affiliation(s)
- Prerna Srivastava
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India
| | - Arpita Rai
- Dental Institute, Rajendra Institute of Medical Sciences, Ranchi, Jharkhand, India
| | - Manish Kumar
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, India.
| |
Collapse
|
7
|
Zheng Q, Wu Y, Zhang X, Zhang Y, Zhu Z, Luan B, Zang P, Sun D. Analysis and validation of hub genes for atherosclerosis and AIDS and immune infiltration characteristics based on bioinformatics and machine learning. Sci Rep 2025; 15:12316. [PMID: 40210656 DOI: 10.1038/s41598-025-96907-6] [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: 08/19/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Atherosclerosis is the major cause of cardiovascular diseases worldwide, and AIDS linked with chronic inflammation and immune activation, increases atherosclerosis risk. The application of bioinformatics and machine learning to identify hub genes for atherosclerosis and AIDS has yet to be reported. Thus, this study aims to identify the hub genes for atherosclerosis and AIDS. Gene expression profiles were downloaded from the Gene Expression Omnibus database. The Robust Multichip Average was performed for data preprocessing, and the limma package was used for screening differentially expressed genes. Enrichment analysis employed GO and KEGG, protein-protein interaction network was constructed. Hub genes were filtered using topological and machine learning algorithms and validated in external cohorts. Then immune infiltration and correlation analysis of hub genes were constructed. Nomogram, receiver operating curve, and single-sample gene set enrichment analysis were applied to evaluate hub genes. This study identified 48 intersecting genes. Enrichment analyses indicated that these genes are significantly enriched in viral response, inflammatory response, and cytokine signaling pathways. CCR5 and OAS1 were identified as common hub genes in atherosclerosis and AIDS for the first time, highlighting their roles in antiviral immunity, inflammation and immune infiltration. These findings contributed to understanding the shared pathogenesis of Atherosclerosis and AIDS and provided possible potential therapeutic targets for immunomodulatory therapy.
Collapse
Affiliation(s)
- Qirui Zheng
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Yupeng Wu
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Pan-Vascular Management Center, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Xiaojiao Zhang
- Department of Cardiology, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Yuzhu Zhang
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Zaihan Zhu
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Bo Luan
- Department of Cardiology, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China
| | - Peizhuo Zang
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China.
- Pan-Vascular Management Center, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
- Liaoning Provincial Key Laboratory of Neurointerventional Therapy and Biomaterials Research and Development, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
| | - Dandan Sun
- Department of Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110067, China.
- Shenyang Clinical Medical Research Center for Ultrasound, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
- Liaoning Provincial Key Laboratory of Neurointerventional Therapy and Biomaterials Research and Development, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, 110067, China.
| |
Collapse
|
8
|
Marquez J, Viviano S, Beckman E, Thies J, Friedland-Little J, Lam CT, Deniz E, Shelkowitz E. Polyamine metabolism is dysregulated in COXFA4-related mitochondrial disease. HGG ADVANCES 2025; 6:100418. [PMID: 39967265 PMCID: PMC11946867 DOI: 10.1016/j.xhgg.2025.100418] [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: 12/06/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Most of the chemical energy that organisms rely on to support cellular function is generated through oxidative phosphorylation, a metabolic pathway in which electron donors NADH and FADH are oxidized through a series of successive steps to generate adenosine triphosphate. These redox reactions are orchestrated by a series of five protein complexes that sit within the mitochondrial membrane. Deficiency of cytochrome c oxidase, the fourth of these complexes, is a recognized cause of mitochondrial disease. COXFA4 encodes one of the protein subunits of cytochrome c oxidase, and variants in COXFA4 have recently been reported in individuals with a range of symptoms. These symptoms can include feeding difficulties, poor growth, cardiomyopathy, Leigh or Leigh-like disease, and neurodevelopmental delay, although these symptoms vary widely between individuals. However, a mechanistic understanding of the connection between COXFA4 loss and these varied disease manifestations is lacking. Using animal modeling in Xenopus, we explored the ramifications of coxfa4 loss of function on the early developing heart. We then conducted a hypothesis naive analysis of cellular gene expression in the context of COXFA4 deletion and discovered a downstream deficiency in the ornithine decarboxylase pathway. Small-molecule-based modulation of the ornithine decarboxylase pathway in our model modified the extent of disease, including improvement of cardiac function. Our findings point to a mechanism by which COXFA4 dysfunction leads to tissue-specific disease.
Collapse
Affiliation(s)
- Jonathan Marquez
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA.
| | - Stephen Viviano
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Pediatric Genomics Discovery Program, Yale School of Medicine, New Haven, CT, USA
| | - Erika Beckman
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Jenny Thies
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Joshua Friedland-Little
- Division of Cardiology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Christina T Lam
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Engin Deniz
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Pediatric Genomics Discovery Program, Yale School of Medicine, New Haven, CT, USA
| | - Emily Shelkowitz
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA.
| |
Collapse
|
9
|
Yang B, Wang C, Chen X, Zhai H, Wu Y, Cui M, Wu J, Li W, Hua B. In silico and animal studies on the anti-cancer mechanisms of Shaoyao Decoction against colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119444. [PMID: 39929402 DOI: 10.1016/j.jep.2025.119444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE It is well known that Shaoyao Decoction (SYD), as a commonly used formula of traditional Chinese medicine (TCM), has a beneficial effect on the treatment of ulcerative colitis (UC). It is found that SYD can also prevent colitis-associated colorectal cancer (CAC). However, its potential anti-cancer mechanism is still waiting to be revealed. AIM OF THE STUDY The aim of this study is to investigate the underlying mechanisms of SYD in inhibiting CAC through silico analysis as well as animal experiment validation. MATERIALS AND METHODS The primary active compounds, potential therapeutic targets and intervening signaling pathways, which SYD might inhibit the CAC process were predicted by network pharmacology analysis combined with our previous research result of high performance liquid chromatography (HPLC). We attempted to validate the acquired hub targets from molecular docking combined with the Gene Expression Profiling Interactive Analysis (GEPIA), the Human Protein Atlas (HPA), and the cBioPortal database comprehensively. Subsequently, an animal model of CAC mice induced by azoxymethane (AOM) and dextran sulfate sodium (DSS) was constructed and treated with SYD for 14 weeks, and tumor-related physical indicators were evaluated after sacrificed. In addition, samples of colon tissues were obtained for histologic and protein level studies to verify the predicted mechanism. RESULTS We obtained 166 active ingredients of SYD and predicted 148 potential targets through network pharmacology analysis, among which quercetin, berberine, kaempferol, wogonin and naringenin were selected as core drug ingredients, and TP53, AKT1, CASP3, PTGS2 and CCND1 were identified and included into the range of core targets. GO and KEGG analyses suggested that the PI3K-Akt signaling pathway might hold a crucial role in CAC prevention and treatment by promoting apoptosis and inhibiting tumor proliferation. In the animal experiment, both SYD and SASP treatments improved the inflammatory condition and pathological damage of the colon tissues in mice. After treatments with SYD and SASP, it was found that decreases of Cyclin D1 and Survivin expression levels and increases of p53 and Cleaved caspase-3 expression levels could be mediated by decreasing the phosphorylation levels of PI3K and Akt proteins in the colon tissues of mice. CONCLUSION The results of our study provide supports that SYD effectively inhibits CAC based on modulating PI3K-Akt signaling pathway to suppress tumor proliferation process as well as to promote tumor apoptosis process.
Collapse
Affiliation(s)
- Bingwei Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chenglei Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xue Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haoyu Zhai
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ying Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Muyao Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiahe Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Weidong Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
10
|
Dasgupta S. Identification of Overlapping Genetic Signatures Between Obstructive Sleep Apnea and Lung Cancer: Moving Beyond "One Drug, One Disease" Paradigm of Pharmaceutical Innovation. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2025. [PMID: 40197140 DOI: 10.1089/omi.2025.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Traditional paradigms of pharmaceutical innovation have long relied on the "one drug, one disease" premise. However, a network mindset in unpacking disease mechanisms can be fruitful to move toward a "one drug, polydisease" paradigm of drug discovery and development. A case in point is obstructive sleep apnea (OSA) and lung cancer, which are two prevalent respiratory disorders that share common risk factors and may potentially exhibit overlapping molecular mechanisms. The putative mechanistic linkages between OSA and lung cancer remain underexplored; however, this study offers new evidence on overlapping genetic signatures between OSA and lung cancer with an in-silico approach. Bioinformatics analysis of the publicly available datasets (GSE135917 and GSE268175) identified 123 upregulated and 13 downregulated genes in OSA and 3175 upregulated and 2272 downregulated genes in lung cancer. A total of four genes (C1GALT1, TMEM106B, ZNF117, and ZNF486) were significantly upregulated with both disorders, highlighting potentially shared genetic and molecular mechanisms. Pathway and cell enrichment analysis indicated that mucin type O-glycan biosynthesis pathway and endothelial cells are strongly associated with these shared genes, lending support for their potential roles in both diseases. Moreover, hsa-miR-34a-5p, hsa-let-7g-5p, and hsa-miR-19a-3p were found to be associated with these common genes. Validation using the GEPIA2 tool confirmed the consistent expression patterns of these four genes in lung cancer. Machine learning analysis highlighted TMEM106B as the most significant biomarker candidate for distinguishing OSA and lung cancer from controls. In summary, this study supports the overarching concept that human diseases can have shared mechanistic pathways in the specific example of OSA and lung cancer. While these findings call for further research and validation, they invite rethinking the current pharmaceutical innovation paradigms to move beyond the "one drug, one disease" concept.
Collapse
Affiliation(s)
- Sanjukta Dasgupta
- Department of Biotechnology, Center for Multidisciplinary Research & Innovations, Brainware University, Barasat, India
| |
Collapse
|
11
|
Modak D, Ghosh S, Sarkar S, Roy SK, Chakraborty A, Ray A, Patel CN, Georrge JJ, Thakur S, Bhattacharjee S. Unveiling the mechanism of amelioration of adjuvant-induced rheumatoid arthritis by Drynaria quercifolia rhizome extract using network pharmacology and gene expression-based studies. Sci Rep 2025; 15:11981. [PMID: 40199969 PMCID: PMC11978801 DOI: 10.1038/s41598-025-87461-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/20/2025] [Indexed: 04/10/2025] Open
Abstract
Rhizomes of Drynaria quercifolia have long been traditionally used to manage rheumatic pain. However, there is limited research supporting this traditional practice and insufficient evidence demonstrating the molecular mechanisms of action of plant-derived bioactives in rheumatoid arthritis (RA). The current study aims to identify the effective components in Drynaria quercifolia methanol rhizome extract (DME) and their probable pharmacological mechanisms in alleviating Rheumatoid Arthritis (RA) using network-pharmacology, molecular docking, molecular-dynamics simulations, and gene expression-based validation. Gas chromatography-mass spectrometry (GC-MS) based screening identified 41 volatile phytocomponents from DME having drug-like potentiality. Network pharmacology-based screening revealed 117 therapeutic targets for RA of which 11 have been identified as core targets. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated that key target genes were mostly enriched in the inflammatory response associated with multiple signalling pathways. Molecular docking and molecular dynamics studies revealed that key target proteins like serine/threonine-protein kinase (AKT1), peroxisome proliferator-activated receptor alpha (PPARA), and peroxisome proliferator-activated receptor gamma (PPARG), exhibited strong binding affinity and stable interactions with multiple phytocomponents present in DME. For experimental verification FCA (Freund's complete adjuvant)-induced chronic arthritis model employed for further molecular investigation. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) results validated that DME significantly (p ≤ 0.05) regulate the expression of key identified target genes AKT and PPARG in experimental RA model. Moreover, this study further confirmed that DME significantly (p ≤ 0.05) downregulated pro-inflammatory mediators like COX-2, IL-6 and TNF-α at gene and protein levels and also normalized (p ≤ 0.05) different oxidative stress parameters in both the low and high dose groups of DME-treated arthritic animals. In conclusion, the network-based in silico approach indicated that the phytocomponents present in DME probably act in a synergistic way to modulate key identified targets associated with RA, which was further validated by experimental studies. Therefore, DME could be a potential alternative in immunomodulatory therapies to combat RA and related chronic inflammatory conditions.
Collapse
Affiliation(s)
- Debabrata Modak
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Subhajit Ghosh
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Sourav Sarkar
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Sudipta K Roy
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Ayan Chakraborty
- Virus Research and Diagnostic Laboratory, North Bengal Medical College and Hospital, Darjeeling, 734012, West Bengal, India
| | - Arpita Ray
- Genetics and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, 734013, West Bengal, India
| | - Chirag N Patel
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Biotechnology Research Center, Technology Innovation Institute, Abu Dhabi, United Arab Emirates
| | - John J Georrge
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Subarna Thakur
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India.
| | - Soumen Bhattacharjee
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India.
| |
Collapse
|
12
|
Wang L, Xu P, Li X, Zhang Q. Comprehensive bioinformatics analysis identified HMGB3 as a promising immunotherapy target for glioblastoma multiforme. Discov Oncol 2025; 16:478. [PMID: 40192954 PMCID: PMC11977083 DOI: 10.1007/s12672-025-02235-6] [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: 12/16/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM) presents significant therapeutic challenges due to its heterogeneous tumorigenicity, drug resistance, and immunosuppression. Although several molecular markers have been developed, there still lack of sensitive molecular for accurately detection. Studying the mechanisms underlying the development of GBM and finding relevant prognostic biomarkers remains crucial. METHODS Single-cell RNA sequencing, bulk RNA-seq, and cancer immune cycle activities of GBM were used to assess the expression of different molecular related to GBM. Bioinformatics analyses were carried to evaluate the functional of the high mobility group protein B3 (HMGB3) in GBM. RESULTS HMGB3 was highly expressed in GBM tissues and influenced the interpatient and intratumoral transcriptomic heterogeneity as well as immunosuppression in GBM. HMGB3 also contributes to a no inflamed tumor microenvironment (TME) and has an inhibitory effect on tumor-associated immune cell infiltration. Besides, HMGB3 participated GBM chemotherapeutic sensitivity and negative correlation with 140 medicines. CONCLUSION HMGB3 as a heterogeneous and immunosuppressive molecule in the GBM TME, making it a potential target for precision therapy for GBM.
Collapse
Affiliation(s)
- Libin Wang
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
- Medical Research Center, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Peizhi Xu
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
- Department of Neurosurgery, The 6th Affiliated Hospital of Shenzhen University Medical School, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China
| | - Xinglong Li
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
- Medical Research Center, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| | - Qinghua Zhang
- Department of Neurosurgery, Shenzhen Nanshan People's Hospital, Shenzhen, No. 89 Taoyuan Road, Nanshan District, Shenzhen, 518052, Guangdong, China.
| |
Collapse
|
13
|
Zhang J, Liu T, Wu H, Wei J, Qu Q. Target oxidative stress-induced disulfidptosis: novel therapeutic avenues in Parkinson's disease. Mol Brain 2025; 18:29. [PMID: 40186271 PMCID: PMC11971801 DOI: 10.1186/s13041-025-01200-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD), a globally prevalent neurodegenerative disorder, has been implicated with oxidative stress (OS) as a central pathomechanism. Excessive reactive oxygen species (ROS) trigger neuronal damage and may induce disulfidptosis-a novel cell death modality not yet characterized in PD pathogenesis. METHOD Integrated bioinformatics analyses were conducted using GEO datasets to identify PD-associated differentially expressed genes (DEGs). These datasets were subjected to: immune infiltration analysis, gene set enrichment analysis (GSEA), weighted gene co-expression network analysis (WGCNA), intersection analysis of oxidative stress-related genes (ORGs) and disulfidptosis-related genes (DRGs) for functional enrichment annotation. Following hub gene identification, diagnostic performance was validated using independent cohorts. LASSO regression was applied for feature selection, with subsequent experimental validation in MPTP-induced PD mouse models. Single-cell transcriptomic profiling and molecular docking studies were performed to map target gene expression and assess drug-target interactions. RESULT A total of 1615 PD DEGs and 200 WGCNA DEGs were obtained, and the intersection with ORGs and DRGs resulted in 202 DEORGs, 11 DEDRGs, and 5 DED-ORGs (NDUFS2, LRPPRC, NDUFS1, GLUD1, and MYH6). These genes are mainly associated with oxidative stress, the respiratory electron transport chain, the ATP metabolic process, oxidative phosphorylation, mitochondrial respiration, and the TCA cycle. 10 hub genes have good diagnostic value, including in the validation dataset (AUC ≥ 0.507). LASSO analysis of hub genes yielded a total of 6 target genes, ACO2, CYCS, HSPA9, SNCA, SDHA, and VDAC1. In the MPTP-induced PD mice model, the expression of ACO2, HSPA9, and SDHA was decreased while the expression of CYCS, SNCA, and VDAC1 was increased, and the expression of the 5 DED-ORGs was decreased. Additionally, it was discovered that N-Acetylcysteine (NAC) could inhibit the occurrence of disulfidptosis in the MPTP-induced PD model. Subsequently, the distribution of target genes with AUC > 0.7 in different cell types of the brain was analyzed. Finally, molecular docking was performed between the anti-PD drugs entering clinical phase IV and the target genes. LRPPRC has low binding energy and strong affinity with duloxetine and donepezil, with binding energies of -7.6 kcal/mol and - 8.7 kcal/mol, respectively. CONCLUSION This study elucidates the pathogenic role of OS-induced disulfidptosis in PD progression. By identifying novel diagnostic biomarkers (e.g., DED-ORGs) and therapeutic targets (e.g., LRPPRC), our findings provide a mechanistic framework for PD management and lay the groundwork for future therapeutic development.
Collapse
Affiliation(s)
- Junshi Zhang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- Department of Neurology, Huaihe Hospital of Henan Universtiy, Kaifeng, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Haojie Wu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Jianshe Wei
- Department of Neurology, Huaihe Hospital of Henan Universtiy, Kaifeng, 475004, China.
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
14
|
Tetorou K, Aghaeipour A, Singh S, Morgan JE, Muntoni F. The role of dystrophin isoforms and interactors in the brain. Brain 2025; 148:1081-1098. [PMID: 39673425 PMCID: PMC11967788 DOI: 10.1093/brain/awae384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 12/16/2024] Open
Abstract
Dystrophin is a protein crucial for maintaining the structural integrity of skeletal muscle. So far, attention has been focused on the role of dystrophin in muscle, in view of the devastating progression of weakness and early death that characterizes Duchenne muscular dystrophy. However, in the last few years, the role of shorter dystrophin isoforms, including development and adult expression-specific mechanisms, has been a greater focus. Within the cerebral landscape, various cell types, such as glia, oligodendrocytes and Purkinje, cerebellar granule and vascular-associated cells express a spectrum of dystrophin isoforms, including Dp427, Dp140, Dp71 and Dp40. The interaction of these isoforms with a multitude of proteins suggests their involvement in neurotransmission, influencing several circuit functions. This review presents the intricate interactions among dystrophin isoforms and diverse protein complexes across different cell types and brain regions, as well as the associated clinical complications. We focus on studies investigating protein interactions with dystrophin in the past 30 years at a biochemical level. In essence, the brain's dystrophin landscape is a thrilling exploration of diversity, challenging preconceptions and opening new avenues for understanding CNS physiology. It also holds potential therapeutic implications for neurological complications involving brain dystrophin deficiency. By revealing the molecular complexities related to dystrophin, this review paves the way for future investigations and therapeutic interventions for this CNS aspect of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Konstantina Tetorou
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Artadokht Aghaeipour
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Simran Singh
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Jennifer E Morgan
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Francesco Muntoni
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
15
|
Fu Q, Shen T, Qiu W, Liao Y, Yu M, Zhou Y. FOSB is a key factor in the genetic link between inflammatory bowel disease and acute myocardial infarction: multiple bioinformatics analyses and validation. BMC Med Genomics 2025; 18:63. [PMID: 40181401 PMCID: PMC11969767 DOI: 10.1186/s12920-025-02129-0] [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: 02/12/2025] [Accepted: 03/20/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Inflammatory Bowel Disease (IBD), which includes Crohn's disease and ulcerative colitis, is associated with an increased risk of Acute Myocardial Infarction (AMI). The genetic mechanisms underlying this link are not well understood. METHODS We downloaded IBD and AMI-related microarray datasets from the NCBI Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified and analyzed using enrichment analysis and Weighted Gene Co-expression Network Analysis (WGCNA). Machine learning techniques, including LASSO, random forest, and Boruta, were employed to screen for hub genes. These genes were validated through qRT-PCR and Western blotting. Single-cell sequencing was used to confirm findings. Additionally, potential therapeutic targets were identified using the Connectivity Map (CMap) database. RESULTS Five key hub genes-THBD, FOSB, ADGPR3, IL1R2, and PLAUR-were identified as significantly involved in both IBD and AMI pathogenesis. A diagnostic model for AMI constructed using these hub genes demonstrated high predictive accuracy. Single-cell sequencing analysis and several potential drugs targeting these hub genes were identified, offering new therapeutic avenues. CONCLUSION This study highlights the crucial role of FOSB and other hub genes in the comorbidity of IBD and AMI. The findings provide novel insights for early diagnosis and potential therapeutic strategies, emphasizing the importance of further investigation into these genetic links.
Collapse
Affiliation(s)
- Qingan Fu
- Cardiovascular Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianzhou Shen
- Cardiovascular Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Weihan Qiu
- School of Computer Science, South China Normal University, Guangzhou, China
| | - Yanhui Liao
- Cardiovascular Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Miao Yu
- Cardiovascular Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Yue Zhou
- Cardiovascular Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
16
|
Barnouin K, Tonoli E, Coveney C, Atkinson J, Sancho M, Skelton A, Boocock DJ, Huang L, Shephard J, Johnson TS, Verderio EAM, Twomey B. Identification of mechanistic CKD biomarkers in a rat SNx kidney fibrosis model by transcriptomics and proteomics detectable in biofluids. Sci Rep 2025; 15:11200. [PMID: 40169735 PMCID: PMC11962143 DOI: 10.1038/s41598-025-93894-6] [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: 01/20/2024] [Accepted: 03/10/2025] [Indexed: 04/03/2025] Open
Abstract
The rat sub-total nephrectomy (SNx) is a functional model of general chronic kidney disease (CKD) where the main pathological driver is glomerular hypertension representative of several subtypes of CKD. Comprehensive transcriptomics and proteomics analyses on the SNx rats were performed to identify biomarkers in plasma or urine that correlate with kidney disease and functional kidney loss. Kidneys were subjected to collagen I and III staining for fibrosis scoring, SWATH-MS proteomics and bulk RNA-sequencing transcriptomics, with SWATH-MS also performed on plasma and urine. Differential expression analysis demonstrated significant dysregulation of genes and proteins involved in fibrosis, metabolism, and immune response in the SNx rats compared to controls. Gene ontology analysis of the intersecting genes and proteins from both studies demonstrated common biology between animal cohorts that reached the predefined kidney disease thresholds (serum creatinine > two-fold or proteinuria > three-fold increase over sham-operated). Thirteen significantly differential molecules were detected with consistent directional changes in both omics datasets. These molecules were detected independently in kidney (both RNA and protein) and urine (protein only), but not in plasma. Bioinformatics analysis enabled the identification of mechanistic CKD biomarkers including lumican and collagen alpha-1(III) chain, whose co-expression has previously been both implicated in fibrosis and detected in urine in CKD patients.
Collapse
Affiliation(s)
- Karin Barnouin
- UCB Pharma, Slough, SL1 3WE, UK.
- MSD, London, EC2M 6UR, UK.
| | - Elisa Tonoli
- School of Science and Technology, Centre for Systems Health and Integrated Metabolic Research (SHiMR), Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Clare Coveney
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - John Atkinson
- UCB Pharma, Slough, SL1 3WE, UK
- Gilead Sciences, Oxford, OX4 4GE, UK
| | | | | | - David J Boocock
- John Van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Linghong Huang
- UCB Pharma, Slough, SL1 3WE, UK
- Mestag Therapeutics, Cambridge, CB10 1XL, UK
| | | | - Timothy S Johnson
- UCB Pharma, Slough, SL1 3WE, UK
- Experimental Renal Medicine, Oncology & Metabolism, University of Sheffield, Sheffield, S10 2RZ, UK
- Mestag Therapeutics, Cambridge, CB10 1XL, UK
| | - Elisabetta A M Verderio
- School of Science and Technology, Centre for Systems Health and Integrated Metabolic Research (SHiMR), Nottingham Trent University, Nottingham, NG11 8NS, UK.
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, BIGEA, 40126, Bologna, Italy.
| | | |
Collapse
|
17
|
Wang X, Gu J, Tang H, Gu L, Bi Y, Kong Y, Shan Q, Yin J, Lou M, Li S, Liu Y. Single-Cell Profiling and Proteomics-Based Insights Into mTORC1-Mediated Angio+TAMs Polarization in Recurrent IDH-Mutant Gliomas. CNS Neurosci Ther 2025; 31:e70371. [PMID: 40202138 PMCID: PMC11979715 DOI: 10.1111/cns.70371] [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: 11/19/2024] [Revised: 01/26/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND IDH mutant gliomas often exhibit recurrence and progression, with the mTORC1 pathway and tumor-associated macrophages potentially contributing to these processes. However, the precise mechanisms are not fully understood. This study seeks to investigate these relationships using proteomic, phosphoproteomic, and multi-dimensional transcriptomic approaches. METHODS This study established a matched transcriptomic, proteomic, and phosphoproteomic cohort of IDH-mutant gliomas with recurrence and progression, incorporating multiple glioma-related datasets. We first identified the genomic landscape of recurrent IDH-mutant gliomas through multi-dimensional differential enrichment, GSVA, and deconvolution analyses. Next, we explored tumor-associated macrophage subpopulations using single-cell sequencing in mouse models of IDH-mutant and wild-type gliomas, analyzing transcriptional changes via AddmodelScore and pseudotime analysis. We then identified these subpopulations in matched primary and recurrent IDH-mutant datasets, investigating their interactions with the tumor microenvironment and performing deconvolution to explore their contribution to glioma progression. Finally, spatial transcriptomics was used to map these subpopulations to glioma tissue sections, revealing spatial co-localization with mTORC1 and angiogenesis-related pathways. RESULTS Multi-dimensional differential enrichment, GSVA, and deconvolution analyses indicated that the mTORC1 pathway and the proportion of M2 macrophages are upregulated during the recurrence and progression of IDH-mutant gliomas. CGGA database analysis showed that mTORC1 activity is significantly higher in recurrent IDH-mutant gliomas compared to IDH-wildtype, with a correlation to M2 macrophage infiltration. KSEA revealed that AURKA is enriched during progression, and its inhibition reduces mTORC1 pathway activity. Single-cell sequencing in mouse models identified a distinct glioma subpopulation with upregulated mTORC1, exhibiting both M2 macrophage and angiogenesis transcriptional features, which increased after implantation of IDH-mutant tumor cells. Similarly, human glioma single-cell data revealed the same subpopulation, with cell-cell communication analysis showing active VEGF signaling. Finally, spatial transcriptomics deconvolution confirmed the co-localization of this subpopulation with mTORC1 and VEGFA in high-grade IDH-mutant gliomas. CONCLUSIONS Our findings suggest mTORC1 activation and Angio-TAMs play key roles in the recurrence and progression of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Xu Wang
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Jingyan Gu
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hongyu Tang
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lianping Gu
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Nanjing Medical UniversityJiangsuChina
| | - Yunke Bi
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Yue Kong
- Sun Yat‐Sen University Guanghua School of StomatologyGuangzhouChina
| | - Qiao Shan
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Jian Yin
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
| | - Meiqing Lou
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
- Nanjing Medical UniversityJiangsuChina
| | - Shouwei Li
- Department of NeurosurgeryCapital Medical University Sanbo Brain HospitalBeijingChina
| | - Yaohua Liu
- Department of NeurosurgeryShanghai General HospitalShanghaiChina
- Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
18
|
He H, Wang L, Ma M. MOGAN for LUAD Subtype Classification by Integrating Three Omics Data Types. CANCER INNOVATION 2025; 4:e160. [PMID: 40026873 PMCID: PMC11868734 DOI: 10.1002/cai2.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 03/05/2025]
Abstract
Background Lung adenocarcinoma (LUAD) is a highly heterogeneous cancer type with a poor prognosis. Accurate subtype identification can help guide its treatment. The traditional subtype identification methods using a single-omics approach make it difficult to comprehensively characterize the molecular features of LUAD. Identification of subtypes through multi-omics association strategies can effectively supplement the shortcomings of single-omics information. Methods In this study, we used the Generative Adversarial Network (GAN) to mine transcriptomic, proteomic, and epigenomic information and generate an integrated data set. The newly integrated data were then used to identify LUAD immune subtypes. In the improved GAN (MOGAN) method, we not only integrated multiple omics datasets but also included the interactions between proteins and genes and between methylation and genes. Thus, we achieved effective complementarity of multi-omics information. Results Two subtypes, MOGANTPM_S1 and MOGANTPM_S2, were identified using immune cell infiltration analysis and the integrated multi-omics data. MOGANTPM_S1 patients displayed higher immune cell infiltration, better prognosis, and sensitivity to immune checkpoint inhibitors (ICIs), while MOGANTPM_S2 had lower immune cell infiltration, poorer prognosis, and were insensitive to ICIs. Therefore, immunotherapy was more suitable for MOGANTPM_S1 patients in clinical practice. In addition, this study developed a LUAD subtype diagnostic model using the transcriptomic and proteomic features of five genes, which can be used to guide clinical subtype diagnosis. Conclusions In summary, the MOGAN method was applied to integrate three omics data types and successfully identify two LUAD immune subtypes with significant survival differences. This classification method may be useful for LUAD treatment decisions.
Collapse
Affiliation(s)
- Haibin He
- Chongqing Key Laboratory of Big Data for Bio IntelligenceChongqing University of Posts and TelecommunicationsChongqingChina
| | - Longxing Wang
- Chongqing Key Laboratory of Big Data for Bio IntelligenceChongqing University of Posts and TelecommunicationsChongqingChina
| | - Mingyue Ma
- Chongqing Key Laboratory of Big Data for Bio IntelligenceChongqing University of Posts and TelecommunicationsChongqingChina
- Institute of Life SciencesChongqing Medical UniversityChongqingChina
| |
Collapse
|
19
|
Yang Z, Tang Y, Wu X, Wang J, Yao W. MicroRNA-130b Suppresses Malignant Behaviours and Inhibits the Activation of the PI3K/Akt Signaling Pathway by Targeting MET in Pancreatic Cancer. Biochem Genet 2025; 63:1660-1685. [PMID: 38607540 PMCID: PMC11929638 DOI: 10.1007/s10528-024-10696-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/10/2024] [Indexed: 04/13/2024]
Abstract
There has been interested in the microRNAs' roles in pancreatic cancer (PC) cell biology, particularly in regulating pathways related to tumorigenesis. The study aimed to explore the hub miRNAs in PC and underlying mechanisms by bioinformatics and fundamental experiments. RNA datasets collected from the Gene Expression Omnibus were analysed to find out differentially expressed RNAs (DERNAs). The miRNA-mRNA and protein-protein interaction (PPI) networks were built. The clinicopathological features and expressions of hub miRNAs and hub mRNAs were explored. Dual-luciferase reporter gene assay was performed to assess the interaction between microRNA and target gene. RT-qPCR and western blot were employed to explore RNA expression. The roles of RNA were detected by CCK-8 test, wound healing, transwell, and flow cytometry experiment. We verified 40 DEmiRNAs and 1613 DEmRNAs, then detected a total of 69 final functional mRNAs (FmRNAs) and 23 DEmiRNAs. In the miRNA-mRNA networks, microRNA-130b (miR-130b) was the hub RNA with highest degrees. Clinical analysis revealed that miR-130b was considerably lower expressed in cancerous tissues than in healthy ones, and patients with higher-expressed miR-130b had a better prognosis. Mechanically, miR-130b directly targeted MET in PC cells. Cell functional experiments verified that miR-130b suppressed cell proliferation, migration, promoted apoptosis, and inhibited the PI3K/Akt pathway by targeting MET in PC cells. Our findings illustrated the specific molecular mechanism of miR-130b regulating PC progress. The miR-130b/MET axis may be an alternative target in the therapeutic intervention of PC and provide an opportunity to deepen our understanding of the pathogenesis of PC.
Collapse
Affiliation(s)
- Zilin Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuming Tang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuejiao Wu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiancheng Wang
- Department of General Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Weiyan Yao
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
20
|
Chen Y, Liu F, Shi S, Xiao S, Gong X. The Integrated Transcriptome Bioinformatics Analysis of Energy Metabolism-Related Profiles for Dorsal Root Ganglion of Neuropathic Pain. Mol Neurobiol 2025; 62:4149-4171. [PMID: 39406937 DOI: 10.1007/s12035-024-04537-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/07/2024] [Indexed: 03/05/2025]
Abstract
Neuropathic pain (NP) is a debilitating disease and is associated with energy metabolism alterations. This study aimed to identify energy metabolism-related differentially expressed genes (EMRDEGs) in NP, construct a diagnostic model, and analyze immune cell infiltration and single-cell gene expression characteristics of NP. GSE89224, GSE123919, and GSE134003 were downloaded from the Gene Expression Omnibus. Differentially expressed genes (DEGs) analysis and an intersection with highly energy metabolism-related modules in weighted gene co-expression network analysis (WGCNA) was performed in GSE89224. Least absolute shrinkage and selection operator (LASSO), random forest, and logistic regression were used for model genes selection. NP samples were divided into high- and low-risk groups and different disease subtypes based on risk score of LASSO algorithm and consensus clustering analysis, respectively. Immune cell composition was estimated in different risk groups and NP subtypes. Datasets 134,003 were performed for identification of single-cell DEGs and functional enrichment. Cell-cell communications and pseudo-time analysis to reveal the expression profile of NP. A total of 38 EMRDEGs were obtained and are majorly enriched in metabolism about glioma and inflammation. LASSO, random forest, and logistic regression identified 6 model genes, which were Itpr1, Gng8, Socs3, Fscn1, Cckbr, and Camk1. The nomogram, based on six model genes, had a good predictive ability, concordance, and diagnostic value. The comparisons between different risk groups and NP subtypes identified important pathways and different immune cells component. The immune infiltration results majorly associated with inflammation and energy metabolism. Single-cell analysis revealed cell-cell communications and cells differentiation characteristics of NP. In conclusion, our results not only elucidate the involvement of energy metabolism in NP but also provides a robust diagnostic tool with six model genes. These findings might give insight into the pathogenesis of NP and provide effective therapeutic regimens for the treatment of NP.
Collapse
Affiliation(s)
- Yongmei Chen
- Department of Laboratory, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, Xiangyang City, Hubei, China
| | - Fan Liu
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Shengnan Shi
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Shugen Xiao
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Xingrui Gong
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China.
| |
Collapse
|
21
|
Kashif M. Gene expression profiling to uncover prognostic and therapeutic targets in colon cancer, combined with docking and dynamics studies to discover potent anticancer inhibitor. Comput Biol Chem 2025; 115:108349. [PMID: 39813876 DOI: 10.1016/j.compbiolchem.2025.108349] [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: 09/30/2024] [Revised: 12/05/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
Drug resistance poses a major obstacle to the efficient treatment of colorectal cancer (CRC), which is one of the cancers that kill people most often in the United States. Advanced colorectal cancer patients frequently pass away from the illness, even with advancements in chemotherapy and targeted therapies. Developing new biomarkers and therapeutic targets is essential to enhancing prognosis and therapy effectiveness. My goal in this study was to use bioinformatics analysis of microarray data to find possible biomarkers and treatment targets for colorectal cancer. Using an ArrayExpress database, I examined a dataset on colon cancer to find genes that were differentially expressed (DEGs) in tumor versus healthy tissues. Integration of advanced bioinformatics tools provided robust insights into the identification and analysis of EGFR as a key player. STRING and Cytoscape enabled the construction and visualization of protein-protein interaction networks, highlighting EGFR as a hub gene due to its centrality and interaction profile. Functional enrichment analysis through DAVID revealed EGFR's involvement in critical biological pathways, as identified in GO and KEGG analyses. This underscores the power of combining computational tools to uncover significant biomarkers like EGFR. Autodock Vina screening of the NCI diversity dataset identified two potential EGFR inhibitors, ZINC13597410 and ZINC04896472. MD simulation data revealed that ZINC04896472 could be potential anticancer inhibitor. These findings serve as a basis for the creation of novel therapeutic approaches that target EGFR and other discovered pathways in CRC. The suggested strategy may improve the efficacy of CRC therapy and advance personalized medicine.
Collapse
Affiliation(s)
- Mohammad Kashif
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
22
|
Farrell C, Tandon K, Ferrari R, Lapborisuth K, Modi R, Snir S, Pellegrini M. The Multi-State Epigenetic Pacemaker enables the identification of combinations of factors that influence DNA methylation. GeroScience 2025; 47:2439-2454. [PMID: 39549198 PMCID: PMC11979089 DOI: 10.1007/s11357-024-01414-7] [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: 03/04/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024] Open
Abstract
Epigenetic clocks, DNA methylation-based predictive models of chronological age, are often utilized to study aging associated biology. Despite their widespread use, these methods do not account for other factors that also contribute to the variability of DNA methylation data. For example, many CpG sites show strong sex-specific or cell-type-specific patterns that likely impact the predictions of epigenetic age. To overcome these limitations, we developed a multidimensional extension of the Epigenetic Pacemaker, the Multi-state Epigenetic Pacemaker (MSEPM). We show that the MSEPM is capable of accurately modeling multiple methylation-associated factors simultaneously, while also providing site-specific models that describe the per site relationship between methylation and these factors. We utilized the MSEPM with a large aggregate cohort of blood methylation data to construct models of the effects of age-, sex-, and cell-type heterogeneity on DNA methylation. We found that these models capture a large faction of the variability at thousands of DNA methylation sites. Moreover, this approach allows us to identify sites that are primarily affected by aging and no other factors. An analysis of these sites reveals that those that lose methylation over time are enriched for CTCF transcription factor chip peaks, while those that gain methylation over time are associated with bivalent promoters of genes that are not expressed in blood. These observations suggest mechanisms that underlie age-associated methylation changes and suggest that age-associated increases in methylation may not have strong functional consequences on cell states. In conclusion, the MSEPM is capable of accurately modeling multiple methylation-associated factors, and the models produced can illuminate site-specific combinations of factors that affect methylation dynamics.
Collapse
Affiliation(s)
- Colin Farrell
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA.
| | - Keshiv Tandon
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Roberto Ferrari
- Dept. of Chemistry, Life Sciences and Environmental Sustainability, Laboratory of Molecular Cell Biology of the Epigenome (MCBE), University of Parma, Parma, Italy
| | - Kalsuda Lapborisuth
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Rahil Modi
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Sagi Snir
- Dept. of Evolutionary Biology, University of Haifa, Haifa, Israel
| | - Matteo Pellegrini
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA.
| |
Collapse
|
23
|
Akkus E, Yekedüz E, Ürün Y. Sodium-Glucose Cotransporter 2 (SGLT2) as a Potential Biomarker and Target in Papillary Renal Cell Carcinoma. Clin Genitourin Cancer 2025; 23:102314. [PMID: 40020263 DOI: 10.1016/j.clgc.2025.102314] [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: 01/09/2025] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND SGLT2 is selectively expressed in the human kidney. SGLT2 inhibitors have markedly changed diabetes, heart failure, and kidney disease treatment, and are under investigation in cancer. However, the role of SGLT2 in papillary renal cell carcinoma (pRCC) is not known. METHODS We investigated the SGLT2 gene expression, associated clinical-molecular features, and overall survival (OS) in pRCC. The Cancer Genome Atlas Program and Gene Expression Omnibus data were utilized. mRNA expression z-scores of the SGLT2 gene relative to normal samples (log-RNASeqV2-RSEM, threshold ± 2) were analyzed (low, unaltered, high expression). RESULTS 273 patients were involved. As per mRNA expression, 180 patients (66%) had low, and the remaining had unaltered expression. High correlation (r > 0.6) with SGLT2 was observed in IRX5, STRIP2, LINC00899, SATB2-AS1, FOXC1, IRX3, SLC22A8, SH3BP5 genes (P < .001,q < 0.001 for all) and with the HIF-2α (r:0.43, P < .001,q < 0.001). Tumor mutational burden (P = .365) and aneuploidy scores (P = .976) did not differ, however, among the genes with the highest alteration frequency, SETD2 alterations (15.63% vs. 1.07%, P < .00, q = 0.046) were more frequent in the unaltered-expression group. Differential protein expression analysis showed highly separated proteins (ERBB2, AR, MAPK14, VHL, TGM2 in the low and SHC1, SQSTM1, MYH14, and CDH1 in the unaltered group). The median OS has not reached the median in both groups [Hazard Ratio(HR) for the unaltered group:2.658, 95% Confidence Interval(CI):1.401-5.043, P = .003]. SGLT2 expression remained a significant prognostic factor in multivariable analysis [HR:2.446 (95%CI: 1.199-4.990), P = .014]. CONCLUSIONS This study reveals the first data that SGLT2 might have a role in pRCC as a pathogenic factor and biomarker. Confirmatory mechanistic studies are needed.
Collapse
Affiliation(s)
- Erman Akkus
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye; Cancer Research Institute, Ankara University, Ankara, Türkiye
| | - Emre Yekedüz
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye; Cancer Research Institute, Ankara University, Ankara, Türkiye; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Türkiye; Cancer Research Institute, Ankara University, Ankara, Türkiye.
| |
Collapse
|
24
|
Lin HY, Chu PY. Mitochondrial calcium uniporter as biomarker and therapeutic target for breast cancer: Prognostication, immune microenvironment, epigenetic regulation and precision medicine. J Adv Res 2025; 70:445-461. [PMID: 38663838 PMCID: PMC11976406 DOI: 10.1016/j.jare.2024.04.015] [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: 12/12/2023] [Revised: 03/24/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
INTRODUCTION Mitochondrial calcium uniporter (MCU) is a central subunit of MCU complex that regulate the levels of calcium ions within mitochondria. A comprehensive understanding the implications of MCU in clinical prognostication, biological understandings and therapeutic opportunity of breast cancer (BC) is yet to be determined. OBJECTIVES This study aims to investigate the role of MCU in predictive performance, tumor progression, epigenetic regulation, shaping of tumor immune microenvironment, and pharmacogenetics and the development of anti-tumor therapy for BC. METHODS The downloaded TCGA datasets were used to identify predictive ability of MCU expressions via supervised learning principle. Functional enrichment, mutation landscape, immunological profile, drug sensitivity were examined using bioinformatics analysis and confirmed by experiments exploiting human specimens, in vitro and in vivo models. RESULTS MCU copy numbers increase with MCU gene expression. MCU expression, but not MCU genetic alterations, had a positive correlation with known BC prognostic markers. Higher MCU levels in BC showed modest efficacy in predicting overall survival. In addition, high MCU expression was associated with known BC prognostic markers and with malignancy. In BC tumor and sgRNA-treated cell lines, enrichment pathways identified the involvement of cell cycle and immunity. miR-29a was recognized as a negative epigenetic regulator of MCU. High MCU levels were associated with increased mutation levels in oncogene TP53 and tumor suppression gene CDH1, as well as with an immunosuppressive microenvironment. Sigle-cell sequencing indicated that MCU mostly mapped on to tumor cell and CD8 T-cells. Inter-databases verification further confirmed the aforementioned observation. miR-29a-mediated knockdown of MCU resulted in tumor suppression and mitochondrial dysfunction, as well as diminished metastasis. Furthermore, MCU present pharmacogenetic significance in cellular docetaxel sensitivity and in prediction of patients' response to chemotherapeutic regimen. CONCLUSION MCU shows significant implication in prognosis, outcome prediction, microenvironmental shaping and precision medicine for BC. miR-29a-mediated MCU inhibition exerts therapeutic effect in tumor growth and metastasis.
Collapse
Affiliation(s)
- Hung-Yu Lin
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Pei-Yi Chu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
25
|
Han M, Wang J, Wu Y, Liao J, Guo J, Tang Z. CEBPA as a potential hub gene for cutaneous inflammation in type 2 diabetes mellitus. Int J Biol Macromol 2025; 298:140080. [PMID: 39837449 DOI: 10.1016/j.ijbiomac.2025.140080] [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: 09/19/2024] [Revised: 12/29/2024] [Accepted: 01/17/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND The role of inflammation in the development of type 2 diabetes mellitus (T2DM) related skin complications necessitates further investigation. This study aims to explore the correlation between inflammation and cutaneous alterations in T2DM, enhancing comprehension of underlying mechanism involved. METHODS Utilizing bioinformatics, the GSE38396 and GSE92724 datasets were employed to identify differentially expressed genes (DEGs) and potential hub genes in T2DM-related skin inflammation. Subsequently, gene functional enrichment analysis was employed for functional annotation. Finally, we validated the regulatory impact of hub gene on inflammation during high glucose incubation using the in vitro model. RESULTS A comprehensive analysis identified 742 DEGs, including 9 hub genes and 4 potential biomarkers. Compared to the CON group, the expression of M2 macrophages was significantly upregulated in the T2DM group, while resting dendritic cells and eosinophils showed notable decreases, indicating a significant correlation with CEBPA. Furthermore, functional enrichment analysis revealed significant enrichment of DEGs in pathways linked to immunity and diabetes pathogenesis. Interestingly, overexpression of CEBPA demonstrated anti-inflammatory effects under hyperglycemic conditions, while silencing CEBPA expression appeared to worsen inflammation. CONCLUSION CEBPA emerges as a potential hub gene for skin inflammation in T2DM, shedding light on the underlying mechanisms of this condition.
Collapse
Affiliation(s)
- Mingzheng Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jingchun Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yijin Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
26
|
Zehr S, Wolf S, Oellerich T, Leisegang MS, Brandes RP, Schulz MH, Warwick T. GeneCOCOA: Detecting context-specific functions of individual genes using co-expression data. PLoS Comput Biol 2025; 21:e1012278. [PMID: 40163580 PMCID: PMC11964461 DOI: 10.1371/journal.pcbi.1012278] [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: 06/26/2024] [Revised: 04/02/2025] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Extraction of meaningful biological insight from gene expression profiling often focuses on the identification of statistically enriched terms or pathways. These methods typically use gene sets as input data, and subsequently return overrepresented terms along with associated statistics describing their enrichment. This approach does not cater to analyses focused on a single gene-of-interest, particularly when the gene lacks prior functional characterization. To address this, we formulated GeneCOCOA, a method which utilizes context-specific gene co-expression and curated functional gene sets, but focuses on a user-supplied gene-of-interest (GOI). The co-expression between the GOI and subsets of genes from functional groups (e.g. pathways, GO terms) is derived using linear regression, and resulting root-mean-square error values are compared against background values obtained from randomly selected genes. The resulting p values provide a statistical ranking of functional gene sets from any collection, along with their associated terms, based on their co-expression with the gene of interest in a manner specific to the context and experiment. GeneCOCOA thereby provides biological insight into both gene function, and putative regulatory mechanisms by which the expression of the GOI is controlled. Despite its relative simplicity, GeneCOCOA outperforms similar methods in the accurate recall of known gene-disease associations. We furthermore include a differential GeneCOCOA mode, thus presenting the first implementation of a gene-focused approach to experiment-specific gene set enrichment analysis. GeneCOCOA is formulated as an R package for ease-of-use, available at https://github.com/si-ze/geneCOCOA.
Collapse
Affiliation(s)
- Simonida Zehr
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Sebastian Wolf
- Goethe University Frankfurt, University Hospital, Department of Medicine II, Haematology/Oncology, Frankfurt am Main, Germany
| | - Thomas Oellerich
- Goethe University Frankfurt, University Hospital, Department of Medicine II, Haematology/Oncology, Frankfurt am Main, Germany
| | - Matthias S Leisegang
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Ralf P Brandes
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| | - Marcel H Schulz
- German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
- Goethe University Frankfurt, Institute for Computational Genomic Medicine, Frankfurt am Main, Germany
| | - Timothy Warwick
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Rhine-Main, Frankfurt am Main, Germany
| |
Collapse
|
27
|
Ithurbide S, Buan N, Schulze S. Advancing archaeal research through FAIR resource and data sharing, and inclusive community building. Commun Biol 2025; 8:519. [PMID: 40157984 PMCID: PMC11954925 DOI: 10.1038/s42003-025-07962-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
Over the last two decades archaeal research has expanded into a wide-ranging research field, driven by a fairly small research community. Archaea are now recognized as important players in the One-Health approach and expertise on the biology of archaea has become crucial in the study of a broad range of topics and environments, including the host-associated microbiomes, major nutrient cycles, greenhouse gas metabolism, the cell biology and origin of eukaryotes, adaptation of life to extremes, as well as various biotechnological applications. Here, we summarize existing resources and ongoing efforts in the engaged broader archaeal scientific community to accelerate research and resource sharing guided by FAIR (findable, accessible, interoperable, reusable) data-sharing principles. We highlight ongoing community efforts that: (i) aim to share protocols and best practices for working with archaea (e.g. ARCHAEA.bio), (ii) combine large 'omics datasets for the dissemination of unified, system-wide results (e.g. Archaeal Proteome Project, KBase) and (iii) provide opportunities for scientists to present their work in a supportive environment and to forge connections and collaborations (e.g. Archaea Power Hour). Together, these resources and projects promise to spur and cross-fertilize research, making archaeal research more accessible to a broader and more diverse audience.
Collapse
Affiliation(s)
- Solenne Ithurbide
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC, Canada
| | - Nicole Buan
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Stefan Schulze
- Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, NY, USA.
| |
Collapse
|
28
|
Tang X, Yang Q, Dou Y, Zhang R, Yan M. Construction of a competing endogenous RNA regulatory network in pterygium and role of hsa_circ_0081682 in fibroblast proliferation, migration, and apoptosis. Exp Eye Res 2025; 255:110365. [PMID: 40164374 DOI: 10.1016/j.exer.2025.110365] [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: 01/23/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Pterygium is a fibrovascular growth associated with chronic inflammation, tissue remodeling, and angiogenesis, which invades the cornea. Circular RNAs (circRNAs) are emerging as pivotal role in many diseases, but their role in pterygium remains unclear. We performed circRNA and miRNA expression profiling on pterygium and conjunctival tissues, then the circRNA-miRNA-mRNA regulatory network was constructed. Bioinformatics was used to predict downstream pathways. Pterygium fibroblasts were used for experiments assessing proliferation (CCK8, EdU), migration (wound healing, transwell), and apoptosis (AnnexinV-FITC/PI). We identified 162 differentially expressed circRNAs and 96 miRNAs. Key pathways involved in pterygium pathogenesis, including focal adhesion and PI3K-Akt signaling, were predicted. Hsa_circ_0081862 was downregulated in pterygium tissues and fibroblasts, inhibiting fibroblast proliferation and migration while promoting apoptosis. This research constructed a ceRNA network and identified hsa_circ_0081682 as the potential diagnostic marker for pterygium. This research contributes to the understanding of biochemical basis of pterygium, which may facilitate the development of targeted strategies for its management and prevention.
Collapse
Affiliation(s)
- Xinyu Tang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiaodan Yang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yulian Dou
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ruiying Zhang
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Yan
- Department of Ophthalmology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
29
|
Wu M, Liu D, Xiong X, Su Q, Xiang Y, Shen L, An Z, Yang X. Analysis of the molecular mechanisms of ulcerative colitis and atherosclerosis by microarray data. Sci Rep 2025; 15:10715. [PMID: 40155679 PMCID: PMC11953266 DOI: 10.1038/s41598-025-95125-4] [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: 10/17/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
Adults can develop ulcerative colitis (UC), a chronic inflammatory illness of the colon, while atherosclerosis (AA) is a chronic inflammatory disease of the blood vessels caused by a range of risk factors. Prior research has demonstrated that UC increases the risk of AA, although the underlying pathological mechanisms are not entirely understood. The purpose of this work was to discover differentially expressed genes (DEGs) in UC and AA and investigate their molecular processes using a bioinformatics method. The UC (GSE36807) and AA (GSE28829) datasets were obtained from the Gene Expression Omnibus (GEO) database. Following the identification of genes that are differentially expressed in common with UC and AA, functional annotation, the construction of protein-protein interaction (PPI) networks and modules, the identification of hub genes, and co-expression analysis were carried out. A total of 105 (including 92 up-regulated and 13 down-regulated genes) DEGs were selected for correlation analysis in the above two datasets, and after Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional analysis immune responses, cytokines, and chemokines were found to play crucial roles in both diseases. Finally, a total of 16 hub genes were identified by CytoHubba and MCODE plugins in Cytoscape, including Chemokine (C-C motif) ligand 4(CCL4), Toll-like receptor 2 (TLR2), Integrin Beta 2(ITGB2), Chemokine (C-C motif) Receptor 1(CCR1), Toll-Like Receptor 8 (TLR8), Fc Fragment of IgG Receptor IIa (FCGR2A), Neutrophil Cytosolic Factor 2(NCF2), Leukocyte immunoglobulin-like receptor B2(LILRB2), FGR proto-oncogene, Src family tyrosine kinase(FGR), Intercellular Adhesion Molecule 1 (ICAM1), Caspase 1(CASP1), Matrix Metallopeptidase 9(MMP9), Cluster of Differentiation 163(CD163), Complement Component 5a Receptor 1 (C5AR1), Neutrophil Cytosolic Factor 4 (NCF4), Selectin P (SELP). This study discovered a link between UC and AA, as well as shared hub genes and pathways, which may bring new insights into the processes of UC and AA.
Collapse
Affiliation(s)
- Min Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China
| | - Dong Liu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China
| | - Xiaoman Xiong
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China
| | - Qiang Su
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China
| | - Yi Xiang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China
| | - Lang Shen
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China
| | - Zhenxiang An
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
| | - Xiaofang Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550002, China.
| |
Collapse
|
30
|
Cha J, Lee I. Single-cell network biology enabling cell-type-resolved disease genetics. Genomics Inform 2025; 23:10. [PMID: 40148916 PMCID: PMC11951680 DOI: 10.1186/s44342-025-00042-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Gene network models provide a foundation for graph theory approaches, aiding in the novel discovery of drug targets, disease genes, and genetic mechanisms for various biological functions. Disease genetics must be interpreted within the cellular context of disease-associated cell types, which cannot be achieved with datasets consisting solely of organism-level samples. Single-cell RNA sequencing (scRNA-seq) technology allows computational distinction of cell states which provides a unique opportunity to understand cellular biology that drives disease processes. Importantly, the abundance of cell samples with their transcriptome-wide profile allows the modeling of systemic cell-type-specific gene networks (CGNs), offering insights into gene-cell-disease relationships. In this review, we present reference-based and de novo inference of gene functional interaction networks that we have recently developed using scRNA-seq datasets. We also introduce a compendium of CGNs as a useful resource for cell-type-resolved disease genetics. By leveraging these advances, we envision single-cell network biology as the key approach for mapping the gene-cell-disease axis.
Collapse
Affiliation(s)
- Junha Cha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
31
|
Johnson Z, Anderson D, Cheung MS, Bohutskyi P. Gene network centrality analysis identifies key regulators coordinating day-night metabolic transitions in Synechococcus elongatus PCC 7942 despite limited accuracy in predicting direct regulator-gene interactions. Front Microbiol 2025; 16:1569559. [PMID: 40207147 PMCID: PMC11979508 DOI: 10.3389/fmicb.2025.1569559] [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: 02/01/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Synechococcus elongatus PCC 7942 is a model organism for studying circadian regulation and bioproduction, where precise temporal control of metabolism significantly impacts photosynthetic efficiency and CO2-to-bioproduct conversion. Despite extensive research on core clock components, our understanding of the broader regulatory network orchestrating genome-wide metabolic transitions remains incomplete. We address this gap by applying machine learning tools and network analysis to investigate the transcriptional architecture governing circadian-controlled gene expression. While our approach showed moderate accuracy in predicting individual transcription factor-gene interactions - a common challenge with real expression data - network-level topological analysis successfully revealed the organizational principles of circadian regulation. Our analysis identified distinct regulatory modules coordinating day-night metabolic transitions, with photosynthesis and carbon/nitrogen metabolism controlled by day-phase regulators, while nighttime modules orchestrate glycogen mobilization and redox metabolism. Through network centrality analysis, we identified potentially significant but previously understudied transcriptional regulators: HimA as a putative DNA architecture regulator, and TetR and SrrB as potential coordinators of nighttime metabolism, working alongside established global regulators RpaA and RpaB. This work demonstrates how network-level analysis can extract biologically meaningful insights despite limitations in predicting direct regulatory interactions. The regulatory principles uncovered here advance our understanding of how cyanobacteria coordinate complex metabolic transitions and may inform metabolic engineering strategies for enhanced photosynthetic bioproduction from CO2.
Collapse
Affiliation(s)
- Zachary Johnson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, United States
| | - David Anderson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Margaret S. Cheung
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Physics, University of Washington, Seattle, WA, United States
| | - Pavlo Bohutskyi
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, United States
| |
Collapse
|
32
|
Scialò C, Zhong W, Jagannath S, Wilkins O, Caredio D, Hruska-Plochan M, Lurati F, Peter M, De Cecco E, Celauro L, Aguzzi A, Legname G, Fratta P, Polymenidou M. Seeded aggregation of TDP-43 induces its loss of function and reveals early pathological signatures. Neuron 2025:S0896-6273(25)00180-1. [PMID: 40157355 DOI: 10.1016/j.neuron.2025.03.008] [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: 07/18/2024] [Revised: 12/24/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Neurodegeneration in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) results from both gain of toxicity and loss of normal function of the RNA-binding protein TDP-43, but their mechanistic connection remains unclear. Increasing evidence suggests that TDP-43 aggregates act as self-templating seeds, propagating pathology through the central nervous system via a prion-like cascade. We developed a robust TDP-43-seeding platform for quantitative assessment of TDP-43 aggregate uptake, cell-to-cell spreading, and loss of function within living cells, while they progress toward pathology. We show that both patient-derived and recombinant TDP-43 pathological aggregates were abundantly internalized by human neuron-like cells, efficiently recruited endogenous TDP-43, and formed cytoplasmic inclusions reminiscent of ALS/FTD pathology. Combining a fluorescent reporter of TDP-43 function with RNA sequencing and proteomics, we demonstrated aberrant cryptic splicing and a loss-of-function profile resulting from TDP-43-templated aggregation. Our data highlight known and novel pathological signatures in the context of seed-induced TDP-43 loss of function.
Collapse
Affiliation(s)
- Carlo Scialò
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Weijia Zhong
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Somanath Jagannath
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Oscar Wilkins
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK; The Francis Crick Institute, London, UK
| | - Davide Caredio
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | | | - Flavio Lurati
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Martina Peter
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Elena De Cecco
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Luigi Celauro
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zurich, Switzerland
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Trieste, Italy
| | - Pietro Fratta
- UCL Queen Square Motor Neuron Disease Centre, Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, UCL, London, UK; The Francis Crick Institute, London, UK
| | | |
Collapse
|
33
|
Jiang X, Wang M, Li H, Liu Y, Dong X. Identification of Oxidative Stress-Associated Biomarkers in Chronic Obstructive Pulmonary Disease: An Integrated Bioinformatics Analysis. Int J Chron Obstruct Pulmon Dis 2025; 20:841-855. [PMID: 40161396 PMCID: PMC11955178 DOI: 10.2147/copd.s485505] [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: 07/04/2024] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is among the three leading causes of death worldwide, with its prevalence, morbidity, and mortality rates increasing annually. Oxidative stress (OS) is a key mechanism in COPD development, making the identification of OS-related biomarkers beneficial for improving its diagnosis and treatment. Methods The genetic data from patients with COPD and controls were obtained from the Gene Expression Omnibus database to identify OS-related genes (OSRGs). Functional enrichment analysis was conducted using the Kyoto encyclopedia of genes and genomes signaling pathway and gene ontology (GO). Protein-protein interaction networks were constructed to identify the core genes, which were further evaluated using receiver operating characteristic (ROC) curves. Diagnostic models were developed based on the core genes. Besides, the correlation between the expression of the core genes and the immune cells was analyzed using single-sample gene set enrichment analysis. Drug-gene interactions were explored to predict target drugs, and related microribonucleic acid (miRNA) and transcription factors (TFs) were identified using miRNet. Results In this study, we identified 299 differential genes, including 16 OSRGs. Among these, five core genes-heat shock protein family A (Hsp70) member 1A (HSPA1A), glutamate-cysteine ligase modifier subunit, interleukin-1 beta (IL-1β), intercellular adhesion molecule 1 (ICAM1), and glutamate-cysteine ligase catalytic subunit (GCLC)-were screened and validated using ROC curve analysis. The results of GO enrichment analysis were mainly focused on the OS response, the negative regulation of the exogenous apoptosis signaling pathway, and the regulation of the apoptosis signaling pathway. Additionally, 33 target drugs were predicted, including ofloxacin, cisplatin, and pegolimumab, among others. Meanwhile, the regulatory networks comprising 33 miRNAs related to the core genes and 38 TFs associated with HSPA1A, IL-1β, ICAM1, and GCLC were constructed. A diagnostic model based on the five genes was constructed and validated with an area under the curve of 0.981 (95% confidence interval: 0.941-1.000). Conclusion This study identifies potential biomarkers for diagnosing COPD, new potential targets, and new directions for drug development and treatment.
Collapse
Affiliation(s)
- Xianwei Jiang
- National Regional TCM (Lung Disease) Diagnostic and Treatment Center, The First Affiliated Hospital of Henan University of CM, Zhengzhou, People’s Republic of China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Minghang Wang
- National Regional TCM (Lung Disease) Diagnostic and Treatment Center, The First Affiliated Hospital of Henan University of CM, Zhengzhou, People’s Republic of China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Henan University of CM, Zhengzhou, People’s Republic of China
| | - Huiru Li
- National Regional TCM (Lung Disease) Diagnostic and Treatment Center, The First Affiliated Hospital of Henan University of CM, Zhengzhou, People’s Republic of China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Yuanyuan Liu
- National Regional TCM (Lung Disease) Diagnostic and Treatment Center, The First Affiliated Hospital of Henan University of CM, Zhengzhou, People’s Republic of China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Xiaosheng Dong
- National Regional TCM (Lung Disease) Diagnostic and Treatment Center, The First Affiliated Hospital of Henan University of CM, Zhengzhou, People’s Republic of China
- First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| |
Collapse
|
34
|
John K, Huntress I, Smith E, Chou H, Tollison TS, Covarrubias S, Crisci E, Carpenter S, Peng X. Human long noncoding RNA VILMIR is induced by major respiratory viral infections and modulates the host interferon response. J Virol 2025:e0014125. [PMID: 40130878 DOI: 10.1128/jvi.00141-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) are a newer class of noncoding transcripts identified as key regulators of biological processes. Here, we aimed to identify novel lncRNA targets that play critical roles in major human respiratory viral infections by systematically mining large-scale transcriptomic data sets. Using bulk RNA-sequencing (RNA-seq) analysis, we identified a previously uncharacterized lncRNA, named virus-inducible lncRNA modulator of interferon response (VILMIR), that was consistently upregulated after in vitro influenza infection across multiple human epithelial cell lines and influenza A virus subtypes. VILMIR was also upregulated after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory syncytial virus (RSV) infections in vitro. We experimentally confirmed the response of VILMIR to influenza infection and interferon-beta (IFN-β) treatment in the A549 human epithelial cell line and found the expression of VILMIR was robustly induced by IFN-β treatment in a dose- and time-specific manner. Single-cell RNA-seq analysis of bronchoalveolar lavage fluid samples from coronavirus disease 2019 (COVID-19) patients uncovered that VILMIR was upregulated across various cell types, including at least five immune cells. The upregulation of VILMIR in immune cells was further confirmed in the human T cell and monocyte cell lines, SUP-T1 and THP-1, after IFN-β treatment. Finally, we found that knockdown of VILMIR expression reduced the magnitude of host transcriptional responses to both IFN-β treatment and influenza A virus infection in A549 cells. Together, our results show that VILMIR is a novel interferon-stimulated gene (ISG) that regulates the host interferon response and may be a potential therapeutic target for human respiratory viral infections upon further mechanistic investigation.IMPORTANCEIdentifying host factors that regulate the immune response to human respiratory viral infection is critical to developing new therapeutics. Human long noncoding RNAs (lncRNAs) have been found to play key regulatory roles during biological processes; however, the majority of lncRNA functions within the host antiviral response remain unknown. In this study, we identified that a previously uncharacterized lncRNA, virus-inducible lncRNA modulator of interferon response (VILMIR), is upregulated after major respiratory viral infections including influenza, severe acute respiratory syndrome coronavirus 2, and respiratory syncytial virus. We demonstrated that VILMIR is an interferon-stimulated gene that is upregulated after interferon-beta (IFN-β) in several human cell types. We also found that knockdown of VILMIR reduced the magnitude of host transcriptional responses to IFN-β treatment and influenza A infection in human epithelial cells. Our results reveal that VILMIR regulates the host interferon response and may present a new therapeutic target during human respiratory viral infections.
Collapse
Affiliation(s)
- Kristen John
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Genetics & Genomics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Ian Huntress
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Ethan Smith
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
| | - Hsuan Chou
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Tammy S Tollison
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Sergio Covarrubias
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Elisa Crisci
- Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, USA
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, North Carolina, USA
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
35
|
Chen W, Haoran C, Jinqiu D, Xinyi T, Dian Y, Yongpeng X, Xiaomin L. Candidate target genes in sepsis diagnosis and therapy: identifying hub genes with a spotlight on KLRB1. BMC Infect Dis 2025; 25:409. [PMID: 40133824 PMCID: PMC11938612 DOI: 10.1186/s12879-025-10818-5] [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: 01/17/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Sepsis, which causes systemic inflammation and organ failure, is one of the leading causes of death in the intensive care unit (ICU) and an urgent social health problem. However, the pathogenesis and molecular mechanism of sepsis are unclear. Therefore, this study aimed to identify candidate Hub genes during sepsis progression and the candidate target genes for sepsis diagnosis and treatment. METHODS GSE54514, GSE57065, GSE69528, GSE95233, and GSE131761 datasets were downloaded from public databases, and the differentially expressed genes (DEGs) between healthy and septic patients in each dataset were screened at adjusted P-value < 0.05 and| log2FC| ≥ 0.58. Subsequently, the obtained DEGs in each dataset were intersected to obtain the Hub genes. In addition, the DEGs between patients with better and poor prognoses in datasets GSE54514 and GSE95233 were analyzed after 28 days. The differential expression of Hub genes in septic patients with good and poor prognoses was detected at adjusted P-value < 0.05 and| log2FC| ≥ 0.58. Finally, real-time quantitative polymerase chain reaction (qRT-PCR) was used to verify the bioinformatics results. RESULTS In datasets GSE54514, GSE57065, GSE69528, GSE95233 and GSE131761, RNASE2, RNASE3, CTSG, SLPI, TNFAIP6, PGLYRP1 and BLOC1S1 were up-regulated in septic patients, and RPL10A and KLRB1 were down-regulated compared to healthy controls. qRT-PCR confirmed the expression trend of the hub genes except CTSG (which was not differentially expressed). Compared to septic patients with good prognoses, the differential expression of RNASE3 was higher in patients with poor prognoses. Furthermore, qRT-PCR revealed that KLRB1 was the only differentially expressed hub gene with down-regulated expression in sepsis patients with poor prognosis. CONCLUSIONS The candidate Hub genes closely related to sepsis include KLRB1, RNASE2, RNASE3, CTSG, SLPI, TNFAIP6, PGLYRP1, BLOC1S1, and RPL10A. KLRB1 is the most relevant candidate hub gene among these hub genes in the molecular underpinnings of sepsis, which could be targeted for sepsis detection and treatment.
Collapse
Affiliation(s)
- Wang Chen
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Chen Haoran
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
- Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Ding Jinqiu
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Tang Xinyi
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Yu Dian
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Xie Yongpeng
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China.
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China.
| | - Li Xiaomin
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China.
- Department of Emergency and Critical Care Medicine, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu, China.
| |
Collapse
|
36
|
Zhang H, Goedegebuure SP, Ding L, DeNardo D, Fields RC, Province M, Chen Y, Payne P, Li F. M3NetFlow: A multi-scale multi-hop graph AI model for integrative multi-omic data analysis. iScience 2025; 28:111920. [PMID: 40034855 PMCID: PMC11872513 DOI: 10.1016/j.isci.2025.111920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/17/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Multi-omic data-driven studies are at the forefront of precision medicine by characterizing complex disease signaling systems across multiple views and levels. The integration and interpretation of multi-omic data are critical for identifying disease targets and deciphering disease signaling pathways. However, it remains an open problem due to the complex signaling interactions among many proteins. Herein, we propose a multi-scale multi-hop multi-omic network flow model, M3NetFlow, to facilitate both hypothesis-guided and generic multi-omic data analysis tasks. We evaluated M3NetFlow using two independent case studies: (1) uncovering mechanisms of synergy of drug combinations (hypothesis/anchor-target guided multi-omic analysis) and (2) identifying biomarkers of Alzheimer's disease (generic multi-omic analysis). The evaluation and comparison results showed that M3NetFlow achieved the best prediction accuracy and identified a set of drug combination synergy- and disease-associated targets. The model can be directly applied to other multi-omic data-driven studies.
Collapse
Affiliation(s)
- Heming Zhang
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University in St. Louis, St. Louis, MO, USA
| | - S. Peter Goedegebuure
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Li Ding
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - David DeNardo
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ryan C. Fields
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Province
- Division of Statistical Genomics, Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
| | - Yixin Chen
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Philip Payne
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University in St. Louis, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University in St. Louis, St. Louis, MO, USA
- Division of Statistical Genomics, Department of Genetics, Washington University in St. Louis, St. Louis, MO, USA
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
37
|
Behrouzi Abady Pamsary S, Esmaeili F, Dehghanian F, Bahadori MH. Expression variation of long noncoding RNAs in dopaminergic cells-derived from stem cells and their MPP+ induced PD models. Hum Mol Genet 2025; 34:599-610. [PMID: 39820316 DOI: 10.1093/hmg/ddae192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/17/2024] [Indexed: 01/19/2025] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder characterized by the progressive loss of nigrostriatal dopaminergic neurons (DA) which can be caused by environmental and genetic factors. lncRNAs have emerged as an important regulatory layer in neurodegenerative disorders, including PD. In this study, we investigated and validated lncRNAs that may serve as diagnostic or therapeutic targets for PD. Key genes associated with midbrain and DA cells were screened by differential gene expression analysis on GSE213100 dataset and candidate lncRNAs were selected for further examination. P19 cells were differentiated into DA cells and received treatment with MPP+ to induce PD-like cytotoxic events, which were confirmed by light microscopy, RT-qPCR, immunofluorescence and flow cytometry. Then, the cells were used to investigate the changes of lncRNAs Malat1, Norad, Snhg1 and Meg3. Here we found that the neuronal phenotype was mainly observed on the 12th day of differentiation and the number of DA markers significantly decreased in PD model cells compared with the control group. Moreover, the expression levels of Meg3, Norad, and Snhg1 were decreased by MPP+ whereas Malat1 level was noticeably higher in MPP+ cells compared to DA cells and the control group. In conclusion, the expression level of lncRNAs was able to show a significant difference between differentiated dopaminergic cells and their Parkinsonian model, thereby improving our understanding of the molecular pathogenesis of PD.
Collapse
Affiliation(s)
- Setareh Behrouzi Abady Pamsary
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, Isfahan, Azadi Square, University of Isfahan, 8174673441, Iran
| | - Fariba Esmaeili
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, Isfahan, Azadi Square, University of Isfahan, 8174673441, Iran
| | - Fariba Dehghanian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, Azadi Square, University of Isfahan, Isfahan, 817467344, Iran
| | - Mohammad Hadi Bahadori
- Department of Histology & Embryology, Rasht - Parastar Street, Guilan University of Medical Sciences, 13111-41937, Iran
| |
Collapse
|
38
|
Ding YF, Ho KH, Lee WJ, Chen LH, Hsieh FK, Tung MC, Lin SH, Hsiao M, Yang SF, Yang YC, Chien MH. Cyclic increase in the histamine receptor H1-ADAM9-Snail/Slug axis as a potential therapeutic target for EMT-mediated progression of oral squamous cell carcinoma. Cell Death Dis 2025; 16:191. [PMID: 40113769 PMCID: PMC11926216 DOI: 10.1038/s41419-025-07507-1] [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: 08/20/2024] [Revised: 02/24/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
The intricate involvement of the histaminergic system, encompassing histamine and histamine receptors, in the progression of diverse neoplasias has attracted considerable scrutiny. Histamine receptor H1 (HRH1) was reported to be overexpressed in several cancer types, but its specific functional implications in oral squamous cell carcinoma (OSCC) predominantly remain unexplored. Our findings indicate that dysregulated high levels of HRH1 were correlated with lymph node (LN) metastasis and poor prognoses in OSCC patients. We identified a disintegrin and metalloprotease 9 (ADAM9) as a critical downstream target of HRH1, promoting protumorigenic and prometastatic characteristics both in vitro and in vivo. Molecular investigations revealed that the cyclic increase in the HRH1-ADAM9-Snail/Slug axis promoted progression of the epithelial-to-mesenchymal transition (EMT). Clinical analyses demonstrated significant correlations of HRH1 expression with ADAM9 and with EMT-related markers, with elevated ADAM9 also associated with LN metastasis in OSCC patients. Regarding therapeutic aspects, we discovered that activated STAT3 acts as a compensatory pathway for the long-term HRH1 signaling blockade in OSCC cells. Combining inhibition of HRH1 and STAT3 using their respective inhibitors or short hairpin (sh)RNAs enhanced the tumor-suppressive effects compared to HRH1 inhibition/depletion alone in OSCC cells and a xenograft model. In summary, HRH1 has emerged as a valuable biomarker for predicting OSCC progression, and combined targeting of HRH1 and STAT3 may represent a promising strategy for preventing OSCC progression.
Collapse
Grants
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
- MOST 112-2320-B-038-054 Ministry of Science and Technology, Taiwan (Ministry of Science and Technology of Taiwan)
Collapse
Affiliation(s)
- Yi-Fang Ding
- Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Otolaryngology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | - Kuo-Hao Ho
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Wei-Jiunn Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | - Li-Hsin Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Feng-Koo Hsieh
- The Genome Engineering & Stem Cell Center, School of Medicine, Washington University, St. Louis, MO, USA
| | - Min-Che Tung
- Department of Surgery, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan, ROC
| | - Shu-Hui Lin
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua, Taiwan, ROC
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan, ROC
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan, ROC
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan, ROC
| | - Yi-Chieh Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
- Department of Medical Research, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan, ROC.
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
- TMU Research Center for Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, ROC.
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital Taipei, Taipei, Taiwan, ROC.
| |
Collapse
|
39
|
Rahiyab M, Khan I, Ali SS, Hussain Z, Ali S, Iqbal A. Computational profiling of molecular biomarkers in congenital disorders of glycosylation Type-I and binding analysis of Ginkgolide A with P4HB. Comput Biol Med 2025; 190:110042. [PMID: 40117797 DOI: 10.1016/j.compbiomed.2025.110042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
AIMS Congenital disorders of glycosylation (CDG) comprise a diverse group of genetic diseases characterized by aberrant glycosylation that leads to severe multi-systematic effects. Despite advancements in understanding the underlying molecular mechanisms, curative options remain limited. This study employed computational methods to identify key molecular biomarkers for CDG-I and examine the pharmacological effects of Ginkgolide A (GA), a potent bioactive natural compound. METHODS We analyzed the GSE8440 microarray dataset to discover differentially expressed genes (DEGs) in patients compared to healthy individuals with CDG-I utilizing GEO2R. Functional enrichments, including gene ontologies (GO) and KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analyses, were conducted to contextualize the biological mechanisms and molecular signatures involved in CDG-I (Congenital Disorders of Glycosylation Type-1). The protein-protein interaction (PPI) network for DEGs was constructed using the STRING database, and the central hub genes within the PPI network were identified using Cytohubba. Furthermore, the 3D structure of the top hub gene (P4HB) was predicted by using the Robetta server. The CASTp was employed to evaluate the active sites. Molecular docking of P4HB with GA was carried out to investigate the binding affinity using the PyRx tool, and the stability of the docked complex was validated through MD simulation. The pharmacokinetics, toxicity, and bioactivity score of GA were comprehensively assessed using SwissADME, ProTox-II, and Molinspiration. RESULTS Our findings indicated 247 significant DEGs, including 146 up-regulated and 101 down-regulated genes. GO and KEGG pathway analyses confirmed that the up-regulated and hub genes were strongly associated with protein folding, glycoprotein processing in the endoplasmic reticulum, and endoplasmic reticulum stress (ER) pathways. P4HB emerged as the top hub gene in CDG-I, playing a significant role in protein folding and ER stress. The 3D structure of P4HB was refined and validated, achieving 95.8 % residues in the most favored region of the Ramachandran plot, with an overall quality of 92.97 %. The CASTp server predicted the largest active site with an area of 2243.660 Å2 and a volume of 3236.584 Å3. Molecular docking revealed that GA has a strong binding affinity with P4HB (-8.9 kcal/mol). The ADME (Absorption, Distribution, Metabolism, Excretion) and toxicity assessments confirmed promising drug-like characteristics, excellent bioavailability, and minimal toxicity risk. CONCLUSION This study emphasizes GA as a potential treatment possibility option to alleviated CDG-I pathology by targeting protein misfolding and ER stress, which are fundamental aspects of the disease. Additionally, our findings indicate that P4HB is a critical molecular target in CDG-I. These results pave the way for future preclinical and clinical investigations aimed at advancing the targeted and tailored treatments for CDG.
Collapse
Affiliation(s)
- Muhammad Rahiyab
- Center for Biotechnology and Microbiology, University of Swat, KPK, Pakistan
| | - Ishaq Khan
- Center for Biotechnology and Microbiology, University of Swat, KPK, Pakistan
| | - Syed Shujait Ali
- Center for Biotechnology and Microbiology, University of Swat, KPK, Pakistan
| | - Zahid Hussain
- Center for Biotechnology and Microbiology, University of Swat, KPK, Pakistan
| | - Shahid Ali
- Center for Biotechnology and Microbiology, University of Swat, KPK, Pakistan
| | - Arshad Iqbal
- Center for Biotechnology and Microbiology, University of Swat, KPK, Pakistan.
| |
Collapse
|
40
|
Fang T, Chen Y, Yuan F, Ma Y, Wang Q, Yao Y, Cai S, Jin W, Miao Q, Hu B. Multi-Omics Integration Reveals Mitochondrial Gene Regulation as a Determinant of Tuberculosis Susceptibility: A Mendelian Randomization Approach. Biomedicines 2025; 13:749. [PMID: 40149725 DOI: 10.3390/biomedicines13030749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Mitochondrial dysfunction has been implicated in the pathogenesis of tuberculosis (TB). Despite emerging evidence of the importance of mitochondrial gene regulation in the immune response, the specific role of mitochondrial-related genes in TB susceptibility remains to be fully elucidated. Methods: We employed a multi-omics approach integrating genetic, methylation, and protein-level data. Mendelian randomization (MR) and colocalization analyses were conducted to explore causal associations between mitochondrial gene features-expression quantitative trait loci (eQTL), methylation quantitative trait loci (mQTL), and protein quantitative trait loci (pQTL)-and TB susceptibility. Data were obtained from the FinnGen cohort and validated using independent datasets. Results: Our analyses identified several key mitochondrial genes (e.g., ACSF3, AK3, LYRM4, and PDHB) significantly associated with TB susceptibility. Random forest analysis and gene set enrichment analysis (GSEA) supported the predictive power of these genes. Furthermore, we observed significant correlations between mitochondrial gene expression and immune cell infiltration in TB patients, suggesting a role of these genes in modulating immune responses during infection. Receiver operating characteristic (ROC) analysis confirmed strong predictive accuracy for the identified feature genes, with area under the curve (AUC) values exceeding 0.7. Conclusions: This study demonstrates that mitochondrial-related gene regulation influences TB susceptibility across genetic, methylation, and protein levels. The integration of multi-omics data provides valuable insight into the molecular mechanisms underlying TB, highlighting the potential of mitochondrial genes as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Tingting Fang
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yu Chen
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Feifei Yuan
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yuyan Ma
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Qingqing Wang
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yumeng Yao
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Sishi Cai
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Wenting Jin
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Qing Miao
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Bijie Hu
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| |
Collapse
|
41
|
Tariq MH, Advani D, Almansoori BM, AlSamahi ME, Aldhaheri MF, Alkaabi SE, Mousa M, Kohli N. The Identification of Novel Therapeutic Biomarkers in Rheumatoid Arthritis: A Combined Bioinformatics and Integrated Multi-Omics Approach. Int J Mol Sci 2025; 26:2757. [PMID: 40141401 PMCID: PMC11943070 DOI: 10.3390/ijms26062757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 03/28/2025] Open
Abstract
Rheumatoid arthritis (RA) is a multifaceted autoimmune disease that is marked by a complex molecular profile influenced by an array of factors, including genetic, epigenetic, and environmental elements. Despite significant advancements in research, the precise etiology of RA remains elusive, presenting challenges in developing innovative therapeutic markers. This study takes an integrated multi-omics approach to uncover novel therapeutic markers for RA. By analyzing both transcriptomics and epigenomics datasets, we identified common gene candidates that span these two omics levels in patients diagnosed with RA. Remarkably, we discovered eighteen multi-evidence genes (MEGs) that are prevalent across transcriptomics and epigenomics, twelve of which have not been previously linked directly to RA. The bioinformatics analyses of the twelve novel MEGs revealed they are part of tightly interconnected protein-protein interaction networks directly related to RA-associated KEGG pathways and gene ontology terms. Furthermore, these novel MEGs exhibited direct interactions with miRNAs linked to RA, underscoring their critical role in the disease's pathogenicity. Overall, this comprehensive bioinformatics approach opens avenues for identifying new candidate markers for RA, empowering researchers to validate these markers efficiently through experimental studies. By advancing our understanding of RA, we can pave the way for more effective therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Muhammad Hamza Tariq
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Dia Advani
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates
| | - Buttia Mohamed Almansoori
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Maithah Ebraheim AlSamahi
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Maitha Faisal Aldhaheri
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Shahad Edyen Alkaabi
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
| | - Mira Mousa
- Department of Public Health and Epidemiology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates;
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Nupur Kohli
- Department of Biomedical Engineering and Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; (M.H.T.); (D.A.); (B.M.A.); (M.E.A.); (M.F.A.); (S.E.A.)
- Healthcare Engineering Innovation Group, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| |
Collapse
|
42
|
Turky MA, Youssef I, El Amir A. Identifying behavior regulatory leverage over mental disorders transcriptomic network hubs toward lifestyle-dependent psychiatric drugs repurposing. Hum Genomics 2025; 19:29. [PMID: 40102990 PMCID: PMC11921594 DOI: 10.1186/s40246-025-00733-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND There is a vast prevalence of mental disorders, but patient responses to psychiatric medication fluctuate. As food choices and daily habits play a fundamental role in this fluctuation, integrating machine learning with network medicine can provide valuable insights into disease systems and the regulatory leverage of lifestyle in mental health. METHODS This study analyzed coexpression network modules of MDD and PTSD blood transcriptomic profile using modularity optimization method, the first runner-up of Disease Module Identification DREAM challenge. The top disease genes of both MDD and PTSD modules were detected using random forest model. Afterward, the regulatory signature of two predominant habitual phenotypes, diet-induced obesity and smoking, were identified. These transcription/translation regulating factors (TRFs) signals were transduced toward the two disorders' disease genes. A bipartite network of drugs that target the TRFS together with PTSD or MDD hubs was constructed. RESULTS The research revealed one MDD hub, the CENPJ, which is known to influence intellectual ability. This observation paves the way for additional investigations into the potential of CENPJ as a novel target for MDD therapeutic agents development. Additionally, most of the predicted PTSD hubs were associated with multiple carcinomas, of which the most notable was SHCBP1. SHCBP1 is a known risk factor for glioma, suggesting the importance of continuous monitoring of patients with PTSD to mitigate potential cancer comorbidities. The signaling network illustrated that two PTSD and three MDD biomarkers were co-regulated by habitual phenotype TRFs. 6-Prenylnaringenin and Aflibercept were identified as potential candidates for targeting the MDD and PTSD hubs: ATP6V0A1 and PIGF. However, habitual phenotype TRFs have no leverage over ATP6V0A1 and PIGF. CONCLUSION Combining machine learning and network biology succeeded in revealing biomarkers for two notoriously spreading disorders, MDD and PTSD. This approach offers a non-invasive diagnostic pipeline and identifies potential drug targets that could be repurposed under further investigation. These findings contribute to our understanding of the complex interplay between mental disorders, daily habits, and psychiatric interventions, thereby facilitating more targeted and personalized treatment strategies.
Collapse
Affiliation(s)
| | - Ibrahim Youssef
- Faculty of Engineering, Biomedical Engineering Department, Cairo University, Giza, 12613, Egypt
| | - Azza El Amir
- Faculty of Science, Biotechnology Department, Cairo University, Giza, 12613, Egypt
| |
Collapse
|
43
|
Yeh YS, Evans TD, Iwase M, Jeong SJ, Zhang X, Liu Z, Park A, Ghasemian A, Dianati B, Javaheri A, Kratky D, Kawarasaki S, Goto T, Zhang H, Dutta P, Schopfer FJ, Straub AC, Cho J, Lodhi IJ, Razani B. Identification of lysosomal lipolysis as an essential noncanonical mediator of adipocyte fasting and cold-induced lipolysis. J Clin Invest 2025; 135:e185340. [PMID: 40091840 PMCID: PMC11910232 DOI: 10.1172/jci185340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/16/2025] [Indexed: 03/19/2025] Open
Abstract
Adipose tissue lipolysis is the process by which triglycerides in lipid stores are hydrolyzed into free fatty acids (FFAs), serving as fuel during fasting or cold-induced thermogenesis. Although cytosolic lipases are considered the predominant mechanism of liberating FFAs, lipolysis also occurs in lysosomes via lysosomal acid lipase (LIPA), albeit with unclear roles in lipid storage and whole-body metabolism. We found that adipocyte LIPA expression increased in adipose tissue of mice when lipolysis was stimulated during fasting, cold exposure, or β-adrenergic agonism. This was functionally important, as inhibition of LIPA genetically or pharmacologically resulted in lower plasma FFAs under lipolytic conditions. Furthermore, adipocyte LIPA deficiency impaired thermogenesis and oxygen consumption and rendered mice susceptible to diet-induced obesity. Importantly, lysosomal lipolysis was independent of adipose triglyceride lipase, the rate-limiting enzyme of cytosolic lipolysis. Our data suggest a significant role for LIPA and lysosomal lipolysis in adipocyte lipid metabolism beyond classical cytosolic lipolysis.
Collapse
Affiliation(s)
- Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Trent D. Evans
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Mari Iwase
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ziyang Liu
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ali Ghasemian
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Borna Dianati
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ali Javaheri
- Cardiovascular Division, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Hanrui Zhang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Partha Dutta
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| | - Francisco J. Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, Pennsylvania, USA
| | - Adam C. Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, Pennsylvania, USA
| | - Jaehyung Cho
- Division of Hematology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
44
|
Qi J, Wang Y, Chen H, Wu K, Zhou P, Dou Y, Xiong B, Zhou W. Advancing the Identification of Bioactive Molecules and the Construction of a Synergistic Drug Delivery System in Combating Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2407802. [PMID: 40091443 DOI: 10.1002/advs.202407802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 02/17/2025] [Indexed: 03/19/2025]
Abstract
In recent years, pneumonia caused by multiple viruses has posed a significant threat to public health, particularly affecting vulnerable populations such as the elderly and immunocompromised individuals. Current treatments primarily focused on antiviral medications, lacking "miracle cure" and innovative approaches for the pathological damage caused by viruses. Since 2019, Traditional Chinese Medicine (TCM) has shown remarkable efficacy in treating coronavirus disease 2019 (COVID-19). However, the application is hindered by intricate mechanisms, variable quality, and slow onset. Clinically, Ge-Gen Decoction (GGD) effectively reduced the severity in patients with viral infections. Taking COVID-19 as a case, the bioactive ingredients from GGD: glabridin (GLA) and puerarin (PUE) are identified. Interestingly, it was discovered that PUE can self-assemble into a 3D hydrogel structure upon heating and cooling, namely PUE@gel. This process mirrored the formation of gel-like precipitates in GGD post-decoction. Motivated by this phenomenon, a decoction-mimicking drug delivery system, glabridin─puerarin self-assembled hydrogel (GLA-PUE@gel) was constructed, which exhibits strong anti-inflammatory and antioxidant properties, comparable to GGD at the same dosage. Additionally, PUE that has a high biosafety threshold can competitively bind to angiotensin converting enzyme 2 (ACE2) on host cells, preventing SARS-CoV-2 from invading. This study offered a promising approach for treating virus-induced lung injury.
Collapse
Affiliation(s)
- Jianhong Qi
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Yanxia Wang
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Huan Chen
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Kaitian Wu
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Pei Zhou
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Yue Dou
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Bingqi Xiong
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| | - Wei Zhou
- Department of Pharmaceutics, China Pharmaceutical University, #24 Tong Jia Xiang, Gulou District, Nanjing, 210009, China
| |
Collapse
|
45
|
Pei YB, Yu ZY, Shen JS. Transfer learning for accelerated failure time model with microarray data. BMC Bioinformatics 2025; 26:84. [PMID: 40098088 PMCID: PMC11917065 DOI: 10.1186/s12859-025-06056-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: 09/05/2024] [Accepted: 01/17/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND In microarray prognostic studies, researchers aim to identify genes associated with disease progression. However, due to the rarity of certain diseases and the cost of sample collection, researchers often face the challenge of limited sample size, which may prevent accurate estimation and risk assessment. This challenge necessitates methods that can leverage information from external data (i.e., source cohorts) to improve gene selection and risk assessment based on the current sample (i.e., target cohort). METHOD We propose a transfer learning method for the accelerated failure time (AFT) model to enhance the fit on the target cohort by adaptively borrowing information from the source cohorts. We use a Leave-One-Out cross validation based procedure to evaluate the relative stability of selected genes and overall predictive power. CONCLUSION In simulation studies, the transfer learning method for the AFT model can correctly identify a small number of genes, its estimation error is smaller than the estimation error obtained without using the source cohorts. Furthermore, the proposed method demonstrates satisfactory accuracy and robustness in addressing heterogeneity across the cohorts compared to the method that directly combines the target and the source cohorts in the AFT model. We analyze the GSE88770 and GSE25055 data using the proposed method. The selected genes are relatively stable, and the proposed method can make an overall satisfactory risk prediction.
Collapse
Affiliation(s)
- Yan-Bo Pei
- School of Statistics, Capital University of Economics and Business, Beijing, China
| | - Zheng-Yang Yu
- School of Statistics, Capital University of Economics and Business, Beijing, China
| | - Jun-Shan Shen
- School of Statistics, Capital University of Economics and Business, Beijing, China.
| |
Collapse
|
46
|
Perez MF. CelEst: a unified gene regulatory network for estimating transcription factor activities in C. elegans. Genetics 2025; 229:iyae189. [PMID: 39705007 PMCID: PMC11912867 DOI: 10.1093/genetics/iyae189] [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: 09/27/2024] [Accepted: 11/02/2024] [Indexed: 12/21/2024] Open
Abstract
Transcription factors (TFs) play a pivotal role in orchestrating critical intricate patterns of gene regulation. Although gene expression is complex, differential expression of hundreds of genes is often due to regulation by just a handful of TFs. Despite extensive efforts to elucidate TF-target regulatory relationships in Caenorhabditis elegans, existing experimental datasets cover distinct subsets of TFs and leave data integration challenging. Here, I introduce CelEst, a unified gene regulatory network designed to estimate the activity of 487 distinct C. elegans TFs-∼58% of the total-from gene expression data. To integrate data from ChIP-seq, DNA-binding motifs, and eY1H screens, optimal processing of each data type was benchmarked against a set of TF perturbation RNA-seq experiments. Moreover, I showcase how leveraging TF motif conservation in target promoters across genomes of related species can distinguish highly informative interactions, a strategy which can be applied to many model organisms. Integrated analyses of data from commonly studied conditions including heat shock, bacterial infection, and sex differences validates CelEst's performance and highlights overlooked TFs that likely play major roles in coordinating the transcriptional response to these conditions. CelEst can infer TF activity on a standard laptop computer within minutes. Furthermore, an R Shiny app with a step-by-step guide is provided for the community to perform rapid analysis with minimal coding required. I anticipate that widespread adoption of CelEsT will significantly enhance the interpretive power of transcriptomic experiments, both present and retrospective, thereby advancing our understanding of gene regulation in C. elegans and beyond.
Collapse
Affiliation(s)
- Marcos Francisco Perez
- Instituto de Biología Molecular de Barcelona (IBMB), CSIC, Parc Científic de Barcelona, C. Baldiri Reixac, 4-8, 08028 Barcelona, Spain
| |
Collapse
|
47
|
Li H, Fan T, Qin M, Chen S, Zhou W, Wu P, Yuan Y, Tang X, Zeng T, Fang J, Yi T, Zhang J, Zou J, Li Y. ESM1 suppresses LncRNA GAS5/miR-23a-3p/PTEN axis to promote the cisplatin-chemotherapy resistance of ovarian cancer cells via activating the PI3K/AKT pathway. Discov Oncol 2025; 16:327. [PMID: 40089962 PMCID: PMC11911279 DOI: 10.1007/s12672-025-02113-1] [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: 12/01/2024] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Cisplatin chemotherapy is an important treatment for advanced ovarian cancer (OC). However, the development of cisplatin resistance greatly limits the survival time of OC patients. Endothelial cell-specific molecule 1 (ESM1) has been found to be an important proto-oncogene promoting OC, but its mediating OC cisplatin resistance remains unknown. METHODS We used quantitative polymerase chain reaction (qPCR) to measure transcription levels of ESM1, Growth arrest specific transcript 5 (GAS5), miR-23a-3p, and Phosphatase And Tensin Homolog (PTEN). A double luciferase reporter gene assay confirmed the direct binding of GAS5 to miR-23a-3p and miR-23a-3p to PTEN mRNA. The effects of ESM1, GAS5, miR-23a-3p, and PTEN on OC cisplatin resistance were tested with an Half Maximal Inhibitory Concentration (IC50) assay. Flow cytometry was used to assess the effects of ESM1, GAS5, and miR-23a-3p on cisplatin-induced OC apoptosis. Changes in apoptosis-related proteins and PI3K/AKT-related proteins were analyzed with western blot (WB). RESULTS ESM1 inhibits the levels of GAS5 and PTEN but increases miR-23a-3p. ESM1 and miR-23a-3p promote OC cisplatin resistance. GAS5 and miR-23a-3p promote cisplatin sensitivity for OC cells. Moreover, the main molecular mechanism is the ESM1/GAS5/miR-23a-3p/PTEN/PI3K/Akt signaling axis. CONCLUSION ESM1 promotes OC cisplatin resistance by activating the Phosphoinositide-3-Kinase (PI3K)/AKT Serine/Threonine Kinase (Akt) signaling pathway through the GAS5/miR-23a-3p/PTEN signaling axis. This suggests that prescriptive ESM1 regulates key downstream molecular mechanisms via non-coding RNA and can be used before neoadjuvant chemotherapy in OC is initiated.
Collapse
Affiliation(s)
- Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Tingyu Fan
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Mei Qin
- Department of Gynecology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Shuanghua Chen
- Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
- Department of Infectious Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Wenchao Zhou
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Peiting Wu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yuwei Yuan
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Tian Zeng
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Jiawen Fang
- Nursing Class of 2022, School of Nursing, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Ting Yi
- Trauma Center, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
| | - Juan Zou
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
48
|
Shang X, Wei R, Yang D, Yu B, Zhang W. Bioinformatics identification and validation of pyroptosis-related gene for ischemic stroke. BMC Med Genomics 2025; 18:53. [PMID: 40091022 PMCID: PMC11912741 DOI: 10.1186/s12920-025-02119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Ischemic stroke (IS) is one of the common and frequent diseases with extremely high lethality and disability in the world, and there is no effective treatment at present. This study aimed to screen hub genes involved in cerebral ischemia/reperfusion injury (CIRI) and pyroptosis, and explore promising intervention targets. METHODS CIRI-related genes (GSE202659 and GSE131193) and pyroptosis-related genes (PRGs) in mice were obtained from the Gene Expression Omnibus (GEO) and GeneCards database. We screened for LASSO regression to construct a prognostic model of GSE131193 and PRGs and examined by GSE137482. The functional enrichment analysis of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA) were performed on pyroptosis-related differentially expressed genes (PRDEGs) of GSE202659.The key modules for CIRI and pyroptosis were identified by Weight Gene Co-expression Network Analysis (WGCNA). Subsequently, Protein-protein Interaction (PPI) network and the Cytoscape was constructed to screen out hub genes. Used the starBase to predict miRNA interacting with hub genes and constructed mRNA-miRNA-lncRNA interaction networks. CIRI-related Molecular Subtypes were constructed for hub genes. The relationship between immune cells and hub genes was verified via CIBERSORT. Finally, we selected C57BL/6 mice to construct models to confirm hub genes by enzyme linked immunosorbent assay (ELISA), reverse transcription-polymerase chain reaction (RT-PCR), western blot, and Immunofluorescence. RESULTS A total of 272 PRGs and 35 PRDEGs were screened. An eight-gene risk prediction models were established (AUC = 0.868). GO, KEGG, GSEA and GSVA analyses revealed that PRDEGs were mainly involved in positive regulation of cytokine production, and NOD-like receptor signaling pathway. And then, seven hub genes (Irf1, Icam1, Tlr2, Tnf, Cebpb, Il1rn, and Casp8) were identified by PPI. Icam1, Tnf, Cebpb, Il1rn, and Casp8 had high expression profiles in Cluster2 by hierarchical clustering. The immune infiltration analysis results showed that among the hub genes, Cebpb, Il1rn, and Casp8, showed a significant positive correlation with the degree of NK.Actived, and Icam1 showed a significant negative correlation with B.Cells.Memory. The results of animal experiments significantly demonstrated an upregulation of Irf1, Icam1, Tlr2, Cebpb, and Il1rn. CONCLUSION Our finding indicated that Irf1, Icam1, Tlr2, Cebpb, and Il1rn are hub genes associated with pyroptosis, and these genes are all associated with different immune cells, so as to provide new targets for the prevention and treatment of IS from the perspective of pyroptosis.
Collapse
Affiliation(s)
- Xinying Shang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Rui Wei
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Di Yang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Bawei Yu
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China
| | - Wei Zhang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, No. 295 Xichang Road, Wu Hua District, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
49
|
Hu K, Shi A, Shu Y, Sudesh S, Ling J, Chen Y, Hua F, Yu S, Zhang J, Yu P. Novel Identification of CD74 as a Biomarker for Diagnosing and Prognosing Sepsis Patients. J Inflamm Res 2025; 18:3829-3842. [PMID: 40115322 PMCID: PMC11922779 DOI: 10.2147/jir.s509089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/04/2025] [Indexed: 03/23/2025] Open
Abstract
Purpose Sepsis, a life-threatening inflammatory condition due to an imbalanced response to infections, has been a major concern. Necroptosis, a newly discovered programmed cell death form, plays a crucial role in various inflammatory diseases. Our study aims to identify necroptosis - related genes (NRGs) and explore their potential for sepsis diagnosis. Patients and methods We used weighted gene co-expression network analysis to identify gene modules associated with sepsis. Cox regression and Kaplan-Meier methods were employed to assess the diagnostic and prognostic value of these genes. Single-cell and immune infiltration analyses were carried out to explore the immune environment in sepsis. Plasma CD74 protein levels were quantified in our samples, and relevant clinical data from electronic patient records were analyzed for correlation. Results CD74 was identified through the intersection of the hub genes of sepsis and NRGs related modules. Septic patients had lower CD74 expression compared to healthy controls. The CD74-based diagnostic model showed better performance in the training dataset (AUC, 0.79 [95% CI, 0.75-0.84]), was cross-validated in external datasets, and demonstrated better performances than other published diagnostic models. Pathway analysis and single-cell profiling supported further exploration of CD74-related inflammation and immune response in sepsis. Conclusion This study presents the first quantitative assessment of human plasma CD74 in sepsis patients. CD74 levels were significantly lower in the sepsis cohort. CD74 warrants further exploration as a potential prognostic and therapeutic target for sepsis.
Collapse
Affiliation(s)
- Kaibo Hu
- Department of Endocrinology and Metabolism, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- The second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Ao Shi
- Faculty of Medicine, St George's University of London, London, UK
| | - Yuan Shu
- Department of Endocrinology and Metabolism, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- The second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Shivon Sudesh
- Faculty of Medicine, St George's University of London, London, UK
| | - Jitao Ling
- Department of Endocrinology and Metabolism, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
- The second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Yixuan Chen
- The second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
- Department of Anesthesiology, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Fuzhou Hua
- Department of Anesthesiology, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Shuchun Yu
- Department of Anesthesiology, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jing Zhang
- Department of Anesthesiology, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Peng Yu
- Department of Endocrinology and Metabolism, second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
50
|
Hosseinpour Z, Rezaei-Tavirani M, Akbari ME, Farahani M. Developing a gene expression classifier for breast cancer diagnosis. Med Biol Eng Comput 2025:10.1007/s11517-025-03329-7. [PMID: 40080330 DOI: 10.1007/s11517-025-03329-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/10/2025] [Indexed: 03/15/2025]
Abstract
Breast cancer (BC) is the most common type of cancer in women worldwide. Solid tumors are complex structures composed of many cell types and extracellular matrix components. Understanding solid tumors is crucial for developing effective treatments. This study aimed to develop a gene expression classifier to predict BC with high accuracy. The study first identified the most important genes for cancer through differential expression analysis (DEA) between breast cancer and adjacent normal breast samples. The R package STRINGdb was then used to create a protein-protein interaction network (PPI) to examine upregulated genes and find clusters. Enrichment analyses were performed to identify overrepresented biological functions and pathways. A logistic regression prediction model was developed using a breast cancer dataset from TCGA and evaluated using discrimination and calibration measures. BUB1 expression in breast cancer was also investigated using quantitative analysis. Two significant clusters were identified, with cell cycle checkpoints and M phase key pathways in one cluster and extracellular matrix organization in the other. A prediction model using the hub gene set (COMP, FN1, SDC1, BUB1, TTK, and NUSAP1) showed high sensitivity (97.2%) and specificity (96.1%), and an AUC of 0.994. Three hub genes (COMP, FN1, and SDC1) were identified through the PPI network, strongly linked to extracellular matrix organization (BUB1, TTK, and NUSAP1) as hub genes involved in M phase and cell cycle checkpoints. Overall, the study identified hub pathways and genes that accurately distinguish between cancer and normal samples, presenting promising new possibilities for early cancer detection and improved BC therapy.
Collapse
Affiliation(s)
- Zahra Hosseinpour
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad-Esmaeil Akbari
- Surgical Oncology, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Farahani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|