1
|
Hart M, Diener C, Rheinheimer S, Kehl T, Keller A, Lenhof HP, Meese E. Expanding the immune-related targetome of miR-155-5p by integrating time-resolved RNA patterns into miRNA target prediction. RNA Biol 2025; 22:1-9. [PMID: 39760255 PMCID: PMC11730359 DOI: 10.1080/15476286.2025.2449775] [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: 08/30/2024] [Revised: 12/14/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025] Open
Abstract
The lack of a sufficient number of validated miRNA targets severely hampers the understanding of their biological function. Even for the well-studied miR-155-5p, there are only 239 experimentally validated targets out of 42,554 predicted targets. For a more complete assessment of the immune-related miR-155 targetome, we used an inverse correlation of time-resolved mRNA profiles and miR-155-5p expression of early CD4+ T cell activation to predict immune-related target genes. Using a high-throughput miRNA interaction reporter (HiTmIR) assay we examined 90 target genes and confirmed 80 genes as direct targets of miR-155-5p. Our study increases the current number of verified miR-155-5p targets approximately threefold and exemplifies a method for verifying miRNA targetomes as a prerequisite for the analysis of miRNA-regulated cellular networks.
Collapse
Affiliation(s)
- Martin Hart
- Institute of Human Genetics, Saarland University (USAAR), Homburg, Germany
- Center of Human and Molecular Biology (ZHMB), Saarland University (USAAR), Saarbrücken, Germany
| | - Caroline Diener
- Institute of Human Genetics, Saarland University (USAAR), Homburg, Germany
| | | | - Tim Kehl
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University (USAAR), Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University (USAAR), Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)–Helmholtz Centre for Infection Research (HZI), Saarland University Campus, Saarbrücken, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University (USAAR), Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University (USAAR), Homburg, Germany
| |
Collapse
|
2
|
Tang S, Long X, Li F, Jiang S, Fu Y, Liu J. Identification of RUVBL2 as a novel biomarker to predict the prognosis and drug sensitivity in multiple myeloma based on ferroptosis genes. Hematology 2025; 30:2467499. [PMID: 39985176 DOI: 10.1080/16078454.2025.2467499] [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: 10/21/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a hematological malignancy with the proliferation of malignant plasma cells. Numerous studies have highlighted the critical role of ferroptosis in MM. However, how to use ferroptosis-related genes (FRGs) for prognostic prediction and treatment guidance in MM remains unknown. METHODS By analysis of GEO databases, the prognostic gene was identified and a therapeutic strategy for MM patients based on FRGs was explored. A total of 12 FRGs were identified, utilizing the STRING database and Cytoscape software, and the PPI networks were constructed to identify hub genes and further functional enrichment analyses. Based on the aforementioned data, this study analyzed the expression of RUVBL2 in MM patients by qRT-PCR and Western blotting. To validate the functional role of RUVBL2 in the MM cells, cellular experiments were ultimately conducted. RESULTS The analysis highlighted six hub genes, including TP53, MCM5, TLR4, RUVBL2, GCLM and ITGA6, and functional enrichment analyses indicating enrichment in DNA replication, regulation of apoptotic signaling pathway and PI3K/AKT signaling pathway. Prognostic analysis indicated that TP53, RUVBL2, and MCM5 are associated with MM prognosis, with RUVBL2 displaying a notable area under the curve (AUC) of 0.823 in ROC analysis. The study first determined that RUVBL2 is highly expressed in MM, siRUVBL2-mediated deletion of RUVBL2 inhibited proliferation, promoted apoptosis and increased the sensitivity of BTZ in MM cells, and also overcame BTZ resistance in CD138+ primary cells from MM patients. CONCLUSIONS Our study first suggested that RUVBL2 may be regarded as potential therapeutic targets and prognostic value in MM.
Collapse
Affiliation(s)
- Sishi Tang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xinyi Long
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Fangfang Li
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Siyi Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
3
|
Yang C, Camargo Tavares L, Lee HC, Steele JR, Ribeiro RV, Beale AL, Yiallourou S, Carrington MJ, Kaye DM, Head GA, Schittenhelm RB, Marques FZ. Faecal metaproteomics analysis reveals a high cardiovascular risk profile across healthy individuals and heart failure patients. Gut Microbes 2025; 17:2441356. [PMID: 39709554 DOI: 10.1080/19490976.2024.2441356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
The gut microbiota is a crucial link between diet and cardiovascular disease (CVD). Using fecal metaproteomics, a method that concurrently captures human gut and microbiome proteins, we determined the crosstalk between gut microbiome, diet, gut health, and CVD. Traditional CVD risk factors (age, BMI, sex, blood pressure) explained < 10% of the proteome variance. However, unsupervised human protein-based clustering analysis revealed two distinct CVD risk clusters (low-risk and high-risk) with different blood pressure (by 9 mmHg) and sex-dependent dietary potassium and fiber intake. In the human proteome, the low-risk group had lower angiotensin-converting enzymes, inflammatory proteins associated with neutrophil extracellular trap formation and auto-immune diseases. In the microbial proteome, the low-risk group had higher expression of phosphate acetyltransferase that produces SCFAs, particularly in fiber-fermenting bacteria. This model identified severity across phenotypes in heart failure patients and long-term risk of cardiovascular events in a large population-based cohort. These findings underscore multifactorial gut-to-host mechanisms that may underlie risk factors for CVD.
Collapse
Affiliation(s)
- Chaoran Yang
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Leticia Camargo Tavares
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Han-Chung Lee
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Joel R Steele
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | | | - Anna L Beale
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
| | - Stephanie Yiallourou
- Preclinical Disease and Prevention Unit, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Melinda J Carrington
- Preclinical Disease and Prevention Unit, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - David M Kaye
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, Australia
- School of Translational Medicine, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Geoffrey A Head
- Neuropharmacology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Pharmacology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
- Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
- Victorian Heart Institute, Monash University, Clayton, Australia
| |
Collapse
|
4
|
Dong Z, Wang X, Hu G, Huang Q, Zhang Y, Jia Y, Du S, Zhu C, Wei F, Zhang D, Wang Y, Cai Q. A KSHV-targeted small molecule efficiently blocks SARS-CoV-2 infection via inhibiting expression of EGFR and Cyclin A2. Emerg Microbes Infect 2025; 14:2440490. [PMID: 39655540 DOI: 10.1080/22221751.2024.2440490] [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: 08/17/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic has led to numerous cases of co-infection with SARS-CoV-2 and other viruses, including Kaposi's sarcoma-associated herpesvirus (KSHV), worldwide. This co-infection has increased patient mortality due to the lack of efficient bi-targeted drugs. Cambogin, a bioactive natural product, has been shown to effectively induce regression of KSHV-latently infected tumours in xenograft mice models; however, its impact on SARS-CoV-2 infection remains unclear. Here, we report that Cambogin targets 46 host genes commonly affected by both SARS-CoV-2 and KSHV infections, as identified through bioinformatics analysis. These genes are related with 14 key upstream signalling pathways, particularly those involved in inflammation regulation, protein phosphorylation, metabolic processes, and cellular stress response. Within the transcriptional factor (TF)-miRNA co-regulatory network, ten out of 46 hub-target genes are closely linked to Cambogin and KSHV/SARS-CoV-2. Importantly, Cambogin not only efficiently blocks the replication and virion production of SARS-CoV-2 in vitro and in vivo by reducing the expression of EGFR and Cyclin A2, but also simultaneously inhibits both SARS-CoV-2 infection and the growth of KSHV-induced tumours in vivo using a murine xenograft model. These findings provide an alternative strategy for the potential use of Cambogin in the treatment of SARS-CoV-2 patients, particularly those with KSHV co-infection.
Collapse
Affiliation(s)
- Zhongwei Dong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Xinyu Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Gaowei Hu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Qingye Huang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yulin Zhang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yuping Jia
- Shandong Academy of Pharmaceutical Sciences, Jinan, People's Republic of China
| | - Shujuan Du
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Caixia Zhu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Fang Wei
- ShengYushou Center of Cell Biology and Immunology, Joint International Research Laboratory of Metabolic & Development Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Daizhou Zhang
- Shandong Academy of Pharmaceutical Sciences, Jinan, People's Republic of China
| | - Yuyan Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Qiliang Cai
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Expert Workstation, Baoji Central Hospital, Baoji, People's Republic of China
- Qidong-Fudan Innovative Institute of Medical Science, Qidong, People's Republic of China
| |
Collapse
|
5
|
Zhou S, Li T, Zhang W, Wu J, Hong H, Quan W, Qiao X, Cui C, Qiao C, Zhao W, Shen Y. The cGAS-STING-interferon regulatory factor 7 pathway regulates neuroinflammation in Parkinson's disease. Neural Regen Res 2025; 20:2361-2372. [PMID: 39359093 PMCID: PMC11759022 DOI: 10.4103/nrr.nrr-d-23-01684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00026/figure1/v/2024-09-30T120553Z/r/image-tiff Interferon regulatory factor 7 plays a crucial role in the innate immune response. However, whether interferon regulatory factor 7-mediated signaling contributes to Parkinson's disease remains unknown. Here we report that interferon regulatory factor 7 is markedly up-regulated in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse model of Parkinson's disease and co-localizes with microglial cells. Both the selective cyclic guanosine monophosphate adenosine monophosphate synthase inhibitor RU.521 and the stimulator of interferon genes inhibitor H151 effectively suppressed interferon regulatory factor 7 activation in BV2 microglia exposed to 1-methyl-4-phenylpyridinium and inhibited transformation of mouse BV2 microglia into the neurotoxic M1 phenotype. In addition, siRNA-mediated knockdown of interferon regulatory factor 7 expression in BV2 microglia reduced the expression of inducible nitric oxide synthase, tumor necrosis factor α, CD16, CD32, and CD86 and increased the expression of the anti-inflammatory markers ARG1 and YM1. Taken together, our findings indicate that the cyclic guanosine monophosphate adenosine monophosphate synthase-stimulator of interferon genes-interferon regulatory factor 7 pathway plays a crucial role in the pathogenesis of Parkinson's disease.
Collapse
Affiliation(s)
- Shengyang Zhou
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Ting Li
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Wei Zhang
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Jian Wu
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Hui Hong
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Wei Quan
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Xinyu Qiao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Chun Cui
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Chenmeng Qiao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Weijiang Zhao
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| | - Yanqin Shen
- Laboratory of Neurodegenerative and Neuroinjury Diseases, Wuxi Medicine School, Jiangnan University, Wuxi, Jiangsu Province, China
| |
Collapse
|
6
|
Yang J, Wu J, Xie X, Xia P, Lu J, Liu J, Bai L, Li X, Yu Z, Li H. Perilipin-2 mediates ferroptosis in oligodendrocyte progenitor cells and myelin injury after ischemic stroke. Neural Regen Res 2025; 20:2015-2028. [PMID: 39254564 PMCID: PMC11691472 DOI: 10.4103/nrr.nrr-d-23-01540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/17/2024] [Accepted: 02/27/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00024/figure1/v/2024-09-09T124005Z/r/image-tiff Differentiation of oligodendrocyte progenitor cells into mature myelin-forming oligodendrocytes contributes to remyelination. Failure of remyelination due to oligodendrocyte progenitor cell death can result in severe nerve damage. Ferroptosis is an iron-dependent form of regulated cell death caused by membrane rupture induced by lipid peroxidation, and plays an important role in the pathological process of ischemic stroke. However, there are few studies on oligodendrocyte progenitor cell ferroptosis. We analyzed transcriptome sequencing data from GEO databases and identified a role of ferroptosis in oligodendrocyte progenitor cell death and myelin injury after cerebral ischemia. Bioinformatics analysis suggested that perilipin-2 (PLIN2) was involved in oligodendrocyte progenitor cell ferroptosis. PLIN2 is a lipid storage protein and a marker of hypoxia-sensitive lipid droplet accumulation. For further investigation, we established a mouse model of cerebral ischemia/reperfusion. We found significant myelin damage after cerebral ischemia, as well as oligodendrocyte progenitor cell death and increased lipid peroxidation levels around the infarct area. The ferroptosis inhibitor, ferrostatin-1, rescued oligodendrocyte progenitor cell death and subsequent myelin injury. We also found increased PLIN2 levels in the peri-infarct area that co-localized with oligodendrocyte progenitor cells. Plin2 knockdown rescued demyelination and improved neurological deficits. Our findings suggest that targeting PLIN2 to regulate oligodendrocyte progenitor cell ferroptosis may be a potential therapeutic strategy for rescuing myelin damage after cerebral ischemia.
Collapse
Affiliation(s)
- Jian Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Xueshun Xie
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Pengfei Xia
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiale Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Roney M, Uddin MN, Khan AA, Fatima S, Mohd Aluwi MFF, Hamim SMI, Ahmad A. Repurposing of dipeptidyl peptidase FDA-approved drugs in Alzheimer's disease using network pharmacology and in-silico approaches. Comput Biol Chem 2025; 116:108378. [PMID: 39938415 DOI: 10.1016/j.compbiolchem.2025.108378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/01/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) have similar clinical characteristics in the brain and islet, as well as an increased incidence with ageing and familial susceptibility. Therefore, in recent years there has been a great desire for research that elucidates how anti-diabetic drugs affect AD. This work attempts to first elucidate the possible mechanism of action of DPP-IV inhibitors in the treatment of AD by employing techniques from network pharmacology, molecular docking, molecular dynamic simulation, principal component analysis, and MM/PBSA. A total of 463 targets were identified from the SwissTargetPrediction and 784 targets were identified from the SuperPred databases. 79 common targets were screened using the PPI network. The GO and KEGG analyses indicated that the activity of DPP-IV against AD potentially involves the hsa04080 neuroactive ligand-receptor interaction signalling pathway, which contains 17 proteins, including CHRM2, CHRM3, CHRNB1, CHRNB4, CHRM1, PTGER2, CHRM4, CHRM5, TACR2, HTR2C, TACR1, F2, GABRG2, MC4R, HTR7, CHRNG, and DRD3. Molecular docking demonstrated that sitagliptin had the greatest binding affinity of -10.7 kcal/mol and established hydrogen bonds with the Asp103, Ser107, and Asn404 residues in the active site of the CHRM2 protein. Molecular dynamic simulation, PCA, and MM/PBSA were performed for the complex of sitagliptin with the above-mentioned proteins, which revealed a stable complex throughout the simulation. The work identifies the active component and possible molecular mechanism of sitagliptin in the treatment of AD and provides a theoretical foundation for future fundamental research and practical implementation.
Collapse
Affiliation(s)
- Miah Roney
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia; Centre for Bio-aromatic Research, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia
| | - Md Nazim Uddin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research, Dhaka 1205, Bangladesh
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Sabiha Fatima
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 12371, Saudi Arabia
| | - Mohd Fadhlizil Fasihi Mohd Aluwi
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia; Centre for Bio-aromatic Research, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia
| | - S M Istiaque Hamim
- Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Lebuhraya Persiaran Tun Khalil Yaakob, Gambang 26300, Kuantan, Pahang, Malaysia
| | - Asrar Ahmad
- Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC, USA
| |
Collapse
|
8
|
Sun H, Liu X, Lu J, Fan H, Lu D, Sun H, Zhou Z, Li Y, Yin X, Song Y, Wang S, Xin T. A multi-omics target study for glioblastoma multiforme (GBM) based on Mendelian randomization analysis. IBRO Neurosci Rep 2025; 18:400-408. [PMID: 40124114 PMCID: PMC11928806 DOI: 10.1016/j.ibneur.2025.02.011] [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: 12/29/2024] [Accepted: 02/23/2025] [Indexed: 03/25/2025] Open
Abstract
Background Glioblastoma multiforme (GBM) is the most frequent type of primary malignant brain tumor. This study utilized Mendelian randomization (MR) analysis to explore the causal link between proteins in plasma and cerebrospinal fluid and GBM. Aims This study aimed to identify proteins in both plasma and cerebrospinal fluid (CSF) that could serve as potential therapeutic targets for GBM. Methods We employed previously published protein quantitative trait loci (pQTL) data from CSF and plasma as the exposure data, alongside aggregated Genome-Wide Association Study (GWAS) data on GBM for our MR analysis. Furthermore, we conducted Bayesian co-localization analysis and examined the protein-protein interaction (PPI) networks of CSF and plasma proteins related to GBM risk. Results MR identified three key proteins linked to GBM risk: ribophorin I (RPN1) in plasma, von Willebrand factor (vWF) and macrophage-stimulating protein (MSP). in CSF. Elevated RPN1 and MSP were associated with decreased GBM risk, while increased vWF was linked to higher risk. External validation confirmed that RPN1 served as a key protein in GBM development. Bayesian co-localization showed a 10.35 % probability of a shared causal variant between RPN1 and GBM. Protein-protein interaction analysis further highlighted related proteins for RPN1. Conclusions In summary, the plasma protein RPN1 and the CSF proteins vWF and MSP are causally associated with the risk of GBM. Further research is needed to clarify the roles of these candidate proteins in GBM. Notably, RPN1 may serve as a potential therapeutic target for GBM. Future clinical studies on GBM treatment could explore drugs targeting RPN1.
Collapse
Affiliation(s)
- Hao Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Xiangyin Liu
- Department of Pharmacy, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Engineering and Technology Research Center for Pediatric Drug Development, Shandong Medicine and Health Key Laboratory of Clinical Pharmacy, Jinan 250000, China
| | - Jiaze Lu
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
| | - Hao Fan
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Dongxiao Lu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Haohan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Zijian Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Yuming Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Xianyong Yin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Yuwen Song
- Department of Opthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Tao Xin
- School of Clinical Medicine, Shandong Second Medical University, Weifang 261053, China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250014, China
- Shandong Engineering Research Center of Precision Diagnosis and Treatment Technology for Neuro-oncology, Jinan 250014, China
- Laboratory of Basic and Translational Neuromedicine, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan 250117, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| |
Collapse
|
9
|
Liu T, Nie H, Huo Z, Yan X. Genome-wide identification of aquaporin and their potential role in osmotic pressure regulation in Ruditapes philippinarum. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101436. [PMID: 39929021 DOI: 10.1016/j.cbd.2025.101436] [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: 10/30/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 02/12/2025]
Abstract
Aquaporins (AQPs) are specialized membrane proteins that create selective water channels, facilitating the transport of water across cell membranes and playing a vital role in maintaining water balance and regulating osmotic pressure in aquatic animals. This study identified 9 aquaporin genes from the genome of R. philippinarum, and a comprehensive analysis was conducted on their gene structure, phylogenetic relationships, protein structure, and chromosome localization. RNA-seq data analysis revealed that aquaporin genes were differentially expressed at different developmental stages, in tissue distribution, and in response to salinity stress. In addition, qPCR results revealed that the expression levels of aquaporin genes (AQP1, AQP4d, and AQP3) were significantly elevated in response to both acute low and high salinity stress, suggesting their important role in osmotic pressure regulation in R. philippinarum. This study's results offer an important reference for further investigations into the regulation of osmotic pressure and salinity adaptation of aquaporin in R. philippinarum.
Collapse
Affiliation(s)
- Tao Liu
- College of Fisheries and Life Science, Dalian Ocean University, 116023 Dalian, China; Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, 116023 Dalian, China
| | - Hongtao Nie
- College of Fisheries and Life Science, Dalian Ocean University, 116023 Dalian, China; Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, 116023 Dalian, China.
| | - Zhongming Huo
- College of Fisheries and Life Science, Dalian Ocean University, 116023 Dalian, China; Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, 116023 Dalian, China
| | - Xiwu Yan
- College of Fisheries and Life Science, Dalian Ocean University, 116023 Dalian, China; Engineering Research Center of Shellfish Culture and Breeding in Liaoning Province, Dalian Ocean University, 116023 Dalian, China
| |
Collapse
|
10
|
Jia Z, Jiang N, Lin L, Li B, Liang X. Integrative proteomic analysis reveals the potential diagnostic marker and drug target for the Type-2 diabetes mellitus. J Diabetes Metab Disord 2025; 24:55. [PMID: 39850446 PMCID: PMC11754769 DOI: 10.1007/s40200-025-01562-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025]
Abstract
Objective The escalating prevalence of Type-2 diabetes mellitus (T2DM) poses a significant global health challenge. Utilizing integrative proteomic analysis, this study aimed to identify a panel of potential protein markers for T2DM, enhancing diagnostic accuracy and paving the way for personalized treatment strategies. Methods Proteome profiles from two independent cohorts were integrated: cohort 1 composed of 10 T2DM patients and 10 healthy controls (HC), and cohort 2 comprising 87 T2DM patients and 60 healthy controls. Differential expression analysis, functional enrichment analysis, receiver operating characteristic (ROC) analysis, and classification error matrix analysis were employed. Results Comparative proteomic analysis identified the differential expressed proteins (DEPs) and changes in biological pathways associated with T2DM. Further combined analysis refined a group of protein panel (including CA1, S100A6, and DDT), which were significantly increased in T2DM in both two cohorts. ROC analysis revealed the area under curve (AUC) values of 0.94 for CA1, 0.87 for S100A6, and 0.97 for DDT; the combined model achieved an AUC reaching 1. Classification error matrix analysis demonstrated the combined model could reach an accuracy of 1 and 0.875 in the 60% training set and 40% testing set. Conclusions This study incorporates different cohorts of T2DM, and refines the potential markers for T2DM with high accuracy, offering more reliable markers for clinical translation. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-025-01562-3.
Collapse
Affiliation(s)
- Zhen Jia
- Department of Peripheral Vascular Diseases, First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Ning Jiang
- Department of Cardiovascular Medicine, First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Lin Lin
- Department of Radiology, First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Bing Li
- Department of Peripheral Vascular Diseases, First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Xuewei Liang
- Department of Peripheral Vascular Diseases, First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
11
|
Liu X, Wang W, Zhao H, Wang Y, Jiang L, Zhang E, Feng Y, Wang X, Qu J, Yang J, Li Z. Transcriptome profiling of triploid Crassostrea gigas gills indicates the host immune mechanism against bacterial infection. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101392. [PMID: 39647257 DOI: 10.1016/j.cbd.2024.101392] [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: 08/26/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
As an important member of global aquaculture, oysters (Crassostrea gigas) have significant economic value. With the development of commercial aquaculture, the frequent occurrence of diseases caused by Vibrio alginolyticus has become a hindrance to high-density aquaculture. Gill tissue, as an important component of immune system of the oysters, plays the key point in the face of invasion by foreign substances. Compared to the diploid oyster, the triploid oyster presents a higher growth rate and lower growth investment, making it a more ideal model for studying oyster immune defense. In this study, triploid oysters were as the research subject, and gill tissues attacked by V. alginolyticus were sequenced. By analyzing samples from different time points, 1746 DEGs were obtained. The KEGG and GO functional enrichment analysis showed that gill tissues mainly participate in immune function through the PIK3-Akt signaling pathway and the MAPK signaling pathway. The protein interaction network revealed three genes (CASP8, CASP9 and PIK3CA) that play core roles in immune defense by analyzing the interaction relationship between genes. Finally, qRT-PCR verified the expression of key genes. This study provides a more effective scientific basis for disease prevention and control of oysters and other bivalve shellfish, and helps to promote the sustainable development of aquaculture.
Collapse
Affiliation(s)
- Xiumei Liu
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Weijun Wang
- Shandong Engineering Research Center of Oyster Germplasm Creation and Efficient Culture, Yantai 264025, China; School of Fisheries, Ludong University, Yantai 264025, China
| | - Haitao Zhao
- Dongying Marine Development Research Institute, Dongying 257091, China
| | - Yongjie Wang
- School of Fisheries, Ludong University, Yantai 264025, China
| | - Liming Jiang
- Yantai Marine Economic Research Institute, Yantai 264003, China
| | - Enshuo Zhang
- School of Fisheries, Ludong University, Yantai 264025, China
| | - Yanwei Feng
- Shandong Engineering Research Center of Oyster Germplasm Creation and Efficient Culture, Yantai 264025, China; School of Fisheries, Ludong University, Yantai 264025, China
| | - Xumin Wang
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Jiangyong Qu
- College of Life Sciences, Yantai University, Yantai 264005, China
| | - Jianmin Yang
- Shandong Engineering Research Center of Oyster Germplasm Creation and Efficient Culture, Yantai 264025, China; School of Fisheries, Ludong University, Yantai 264025, China
| | - Zan Li
- Shandong Engineering Research Center of Oyster Germplasm Creation and Efficient Culture, Yantai 264025, China; School of Fisheries, Ludong University, Yantai 264025, China.
| |
Collapse
|
12
|
Zhilin L, Haobo F, Juan W, AiRui X, XiaoDong L, Yuan Y, Junguo D. Investigating the therapeutic potential of Ganoderma lucidum in treating optic nerve atrophy through network pharmacology and experimental validation. Biochem Biophys Res Commun 2025; 760:151702. [PMID: 40158404 DOI: 10.1016/j.bbrc.2025.151702] [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: 10/30/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE The aim of this study is to employ network pharmacology to identify potential therapeutic targets for Ganoderma lucidum in the treatment of optic atrophy, and elucidate the underlying pharmacological mechanism. METHODS This study is mainly divided into two parts. In the first part, the chemical composition and Target of Ganoderma lucidum compound were predicted by TCMSP and Swiss Target Prediction, and the crossover gene between OA and Ganoderma lucidum target gene was screened based on GeneCards and OMIM database. Then, the target genes were enriched and the main pathways of action were analyzed to discover the possible mechanism of action for the treatment of optic atrophy. Finally, the selected core compounds and core targets were interfaced to understand the main binding patterns and affinity. The second part mainly verifies whether Ganoderma lucidum polysaccharide has protective effect on RGC. Firstly, CCK8 method was used to detect the proliferation and virulence analysis of RGC-5 cells with different concentrations of Ganoderma lucidum polysaccharide, and then RGC-5 cells were cultured in subgroups for 12 h, and then put into anaerobic encapsulation to make molds. After 24 h of continuous culture, cells were removed and collected for subsequent RT-PCR and WB detection. RESULTS Through screening target genes of Ganoderma lucidum and OA, 85 potential therapeutic targets were obtained by intersection. Through PPI network analysis of 85 potential targets, it was found that the degree values of TP53, TNF, CASP3, IL6, EGFR, MTOR, ESR1 and other targets were higher. (+)-Ganoderic acid Mf, (+)-Methyl ganolucidate A, epoxyganoderiol A, Ergosta-4,7, 22-Trien-3, 6-Dione and other compounds play a key role in the whole network. It may be the key compound of ganoderma lucidum in treating OA. Through enrichment pathway analysis, it was found that the number of genes was enriched in AGE-RAGE signaling pathway, cAMP signaling pathway, inflammation and cancer pathways, and the structure of TP53, TNF, CASP3, and IL6 binding to the above compounds was stable and the binding activity was high. CONCLUSIONS The findings suggest that Ganoderma lucidum may exert its therapeutic effects on optic atrophy by targeting TP53, TNF, CASP3, and IL6. Additionally, it may also be involved in the AGE-RAGE signaling pathway and cAMP signaling pathway. These results provide reference for the clinical application of ganoderma lucidum in the treatment of OA.
Collapse
Affiliation(s)
- Li Zhilin
- Eye School of Chengdu University of TCM, China; Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, China; Retinal Image Technology and Chronic Vascular Disease Prevention&Control and Collaborative Innovation Center, China
| | - Fan Haobo
- Eye School of Chengdu University of TCM, China
| | - Wen Juan
- Ineye Hospital of Chengdu University of TCM, China
| | - Xie AiRui
- Ineye Hospital of Chengdu University of TCM, China
| | - Li XiaoDong
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, China
| | - Ying Yuan
- Chengdu Coma Ren Far Technology Co., LTD, China
| | - Duan Junguo
- Eye School of Chengdu University of TCM, China; Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, China; Retinal Image Technology and Chronic Vascular Disease Prevention&Control and Collaborative Innovation Center, China; Ineye Hospital of Chengdu University of TCM, China.
| |
Collapse
|
13
|
Wang H, Mei Q, Mei P. Comprehensive analysis of the role of Caspases in glioma. Brain Res 2025; 1855:149529. [PMID: 40032044 DOI: 10.1016/j.brainres.2025.149529] [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: 09/15/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Caspases (CASPs) are attractive targets for cancer therapy. Many prognostic models based on gene signatures include genes from the CASPs family in diffuse glioma. CASP3, CASP4 and CASP6 in glioma have been studied individually. However, specialized comprehensive analysis of the roles of CASPs family in glioma is lacking. Therefore, this study utilized bioinformatics methods to investigate this issue. CASP1-10 expressionlevels were significantly up-regulated in LGG and GBM and glioma, and varied significantly across different clinical subgroups of glioma and LGG and various cell types, and most of CASP1-10 members showed significant differences in recurrence status of LGG. 10 signatures (CASP1-10) were associated with poor overall survival (OS) in glioma and LGG and GBM. However, pan-cancer survival analysis showed that CASP1-10 were associated with the prognosis of LGG, but not GBM. CASP1-10 were related to poor prognosis of glioma and LGG, except for CASP9, which was the opposite of a protective factor. CASP1-10 were independent prognostic factors for OS in glioma and LGG, except for CASP5, and also for recurrence-free survival (RFS) in LGG. Most of CASP1-10 were also independent prognostic factors for disease-specific survival (DSS) and progression-free interval (PFI) and had diagnostic value in glioma and LGG. Genetic alterations of CASP1-10 genes set were associated with poor prognosis in LGG. CASP1-10 were involved in immune infiltration and programmed cell death in glioma and LGG and GBM, and might promote the apoptosis of immune cells. Compared to GBM, CASP1-10 had a more significant impact on the prognosis, cancer-related pathways, and immune infiltration in LGG, indicating that CASP1-10 might play important roles in the recurrence and progression of LGG, and might be promising therapeutic targets for LGG. Therefore, it is speculated that natural caspase inhibitor p35 may be a promising drug for the treatment of glioma, especially for LGG.
Collapse
Affiliation(s)
- Heming Wang
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresource, Hainan University, Haikou 570228, China
| | - Qunfang Mei
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Pengying Mei
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
14
|
Liang X, Tan S, Chen Y, Wei C, Qin Z. Bioinformatics exploration of SPHKAP's role in IDH-mutant glioma involving energy metabolism, prognosis, and immune modulation. J Neuroimmunol 2025; 402:578570. [PMID: 40058165 DOI: 10.1016/j.jneuroim.2025.578570] [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: 08/07/2024] [Revised: 09/14/2024] [Accepted: 02/22/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND The current understanding of glioma pathogenesis is limited by the lack of comprehensive insights into the metabolic reprogramming associated with isocitrate dehydrogenase (IDH) mutations. This study aims to contribute a step to this gap by investigating the role of energy metabolism-related genes in glioma. Our objective is to identify key molecular markers that could serve as prognostic markers and potential therapeutic targets for more effective treatment strategies in IDH-mutant glioma patients. METHODS We conducted an in-depth analysis of gene expression data from TCGA, CGGA, and GEO databases, employing Weighted Gene Co-expression Network Analysis (WGCNA) and differential gene expression analysis to pinpoint candidate genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to elucidate the biological pathways implicated by these genes. Protein-Protein Interaction (PPI) and Gene Multiple Association Network Integration Algorithm (GeneMANIA) networks were constructed to map gene interactions, and survival analysis and Cox regression models were utilized to assess the prognostic value of the identified genes. Additionally, CIBERSORT was used to evaluate immune cell infiltration in the tumor microenvironment. RESULTS Our findings identified SPHKAP as a gene significantly downregulated in glioma tissues compared to control samples. Specifically, low SPHKAP expression was associated with a poorer prognosis of patients with IDH-mutant glioma and linked to the expression of key enzymes involved in energy metabolism. Meanwhile, in IDH-mutant gliomas, reduced SPHKAP expression was correlated with increased macrophage infiltration, enhanced T cell response, and upregulation of immune checkpoint genes, highlighting its role as an independent prognostic marker. CONCLUSION This study reveals the differential expression of SPHKAP in glioma, suggesting its potential as a prognostic marker for IDH-mutant gliomas, providing information for future studies aimed at developing targeted therapies for glioma patients.
Collapse
Affiliation(s)
- Xi Liang
- Department of Neurosurgery, Guangxi Hospital, the First Affiliated Hospital of Sun Yat-sen University, Qingxiu District, Nanning 530022, PR China.
| | - Shi Tan
- Department of Neurosurgery, Guigang City People's Hospital, Gangbei District, Guigang 537100, PR China
| | - Yuecheng Chen
- Department of Neurosurgery, Guigang City People's Hospital, Gangbei District, Guigang 537100, PR China
| | - Cuirong Wei
- Department of Pathology, Guigang City People's Hospital, Gangbei District, Guigang 537100, PR China
| | - Zhongqiao Qin
- Department of Neurosurgery, Guigang City People's Hospital, Gangbei District, Guigang 537100, PR China.
| |
Collapse
|
15
|
Miller CJ, Golovina E, Gokuladhas S, Wicker JS, Jacobsen JC, O'Sullivan JM. Unraveling ADHD: genes, co-occurring traits, and developmental dynamics. Life Sci Alliance 2025; 8:e202403029. [PMID: 40000109 PMCID: PMC11861640 DOI: 10.26508/lsa.202403029] [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: 09/02/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a heterogeneous neurodevelopmental condition with a high prevalence of co-occurring conditions, contributing to increased difficulty in long-term management. Genome-wide association studies have identified variants shared between ADHD and co-occurring psychiatric disorders; however, the genetic mechanisms are not fully understood. We integrated gene expression and spatial organization data into a two-sample Mendelian randomization study for putatively causal ADHD genes in fetal and adult cortical tissues. We identified four genes putatively causal for ADHD in cortical tissues (fetal: ST3GAL3, PTPRF, PIDD1; adult: ST3GAL3, TIE1). Protein-protein interaction databases seeded with the causal ADHD genes identified biological pathways linking these genes with conditions (e.g., rheumatoid arthritis) and biomarkers (e.g., lymphocyte counts) known to be associated with ADHD, but without previously shown genetic relationships. The analysis was repeated on adult liver tissue, where putatively causal ADHD gene ST3GAL3 was linked to cholesterol traits. This analysis provides insight into the tissue-dependent temporal relationships between ADHD, co-occurring traits, and biomarkers. Importantly, it delivers evidence for the genetic interplay between co-occurring conditions, both previously studied and unstudied, with ADHD.
Collapse
Affiliation(s)
- Catriona J Miller
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Evgeniia Golovina
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Sreemol Gokuladhas
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Joerg S Wicker
- School of Computer Science, University of Auckland, Auckland, New Zealand
| | - Jessie C Jacobsen
- School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Justin M O'Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- Garvan Institute of Medical Research, Sydney, Australia
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
16
|
Wen H, He Y, Tang Y, Zhu L, Tao Q, Jin B, Luo T, Peng Y, Wei Y, Lei J, Wang L, Wang F, Ling F, Gao Y, Han L. Altered immune response is associated with sex difference in vulnerability to Alzheimer's disease in human prefrontal cortex. Brain Pathol 2025; 35:e13318. [PMID: 39497354 PMCID: PMC11961208 DOI: 10.1111/bpa.13318] [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: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 04/03/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with a higher risk incidence in females than in males, and there are also differences in AD pathophysiology between sexes. The role of sex in the pathogenesis of AD may be crucial, yet the cellular and molecular basis remains unclear. Here, we performed a comprehensive analysis using four public transcriptome datasets of AD patients and age-matched control individuals in prefrontal cortex, including bulk transcriptome (295 females and 402 males) and single-nucleus RNA sequencing (snRNA-seq) data (224 females and 219 males). We found that the transcriptomic profile in female control was similar to those in AD. To characterize the key features associated with both the pathogenesis of AD and sex difference, we identified a co-expressed gene module that positively correlated with AD, sex, and aging, and was also enriched with immune-associated pathways. Using snRNA-seq datasets, we found that microglia (MG), a resident immune cell in the brain, demonstrated substantial differences in several aspects between sexes, such as an elevated proportion of activated MG, altered transcriptomic profile and cell-cell interaction between MG and other brain cell types in female control. Additionally, genes upregulated in female MG, such as TLR2, MERTK, SPP1, SLA, ACSL1, and FKBP5, had high confidence to be identified as biomarkers to distinguish AD status, and these genes also interacted with some approved drugs for treatment of AD. These findings underscore the altered immune response in female is associated with sex difference in susceptibility to AD, and the necessity of considering sex factors when developing AD biomarkers and therapeutic strategies, providing a scientific basis for further in-depth studies on sex differences in AD.
Collapse
Affiliation(s)
- Huiying Wen
- BGI ResearchHangzhouChina
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
- BGI ResearchShenzhenChina
| | - Youzhe He
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Yuanchun Tang
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Langjian Zhu
- BGI ResearchHangzhouChina
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
- BGI ResearchShenzhenChina
| | - Quyuan Tao
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Bufan Jin
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Ting Luo
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yujie Peng
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yanrong Wei
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Junjie Lei
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Lifang Wang
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Fan Wang
- Department of Pathology of Sir Run Run Shaw Hospital, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
- Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
| | - Fei Ling
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | - Yue Gao
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- Department of Pathology of Sir Run Run Shaw Hospital, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
- Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
| | - Lei Han
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| |
Collapse
|
17
|
Serpente M, Delvecchio G, Fenoglio C, Di Consoli L, Giudici G, Borracci V, Rotondo E, Arcaro M, Sacchi L, Pintus M, Ghezzi L, Ferro A, Prunas C, Callari A, Scola E, Triulzi FM, Arighi A, Brambilla P, Galimberti D. Differential miRNA expression in neural-enriched extracellular vesicles as potential biomarker for frontotemporal dementia and bipolar disorder. Neurobiol Dis 2025; 208:106867. [PMID: 40064379 DOI: 10.1016/j.nbd.2025.106867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025] Open
Abstract
Behavioral variant of Frontotemporal Dementia (bvFTD) and Bipolar Disorder (BD) share overlapping symptoms, complicating diagnosis. BvFTD, especially linked to C9orf72 expansions, often mimics BD, highlighting the need for reliable biomarkers. This study aimed to differentiate bvFTD from BD using miRNA profiles in neural-enriched extracellular vesicles (NEVs). A cohort of 100 subjects was analyzed: 40 bvFTD (20 sporadic, 20 C9orf72 carriers), 40 BD, and 20 healthy controls. NEVs were isolated from plasma and profiled using real-time PCR. Among 754 miRNAs, 11 were significantly deregulated in bvFTD and BD. MiR-152-5p was downregulated in sporadic bvFTD, while let-7b, let-7e, miR-18b, and miR-142-5p were altered in C9orf72 carriers. BD patients showed distinct patterns in miR-331-5p, miR-335, and miR-345 compared to bvFTD. Bioinformatics analyses revealed that let-7e, let-7b, miR-18b, and miR-142-5p share common long non-coding RNA (lncRNA) targets, including XIST, NEAT1, and OIP5-AS1, suggesting their involvement in molecular networks relevant to C9orf72-related bvFTD. These miRNA signatures can differentiate bvFTD from BD, especially in C9orf72-related cases, and offer insights into disease pathways. Further research is needed to validate these findings and explore their clinical application.
Collapse
Affiliation(s)
- Maria Serpente
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy.
| | - Giuseppe Delvecchio
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Lorena Di Consoli
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Giulia Giudici
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Vittoria Borracci
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Emanuela Rotondo
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Marina Arcaro
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sacchi
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Manuela Pintus
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Laura Ghezzi
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Adele Ferro
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Cecilia Prunas
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Antonio Callari
- Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy
| | - Elisa Scola
- Neuroradiology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio M Triulzi
- Neuroradiology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Dept of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Andrea Arighi
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Brambilla
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Neurosciences and Mental Health, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Mila, Italy; Dept of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione Ca' Granda, IRCSS Ospedale Maggiore Policlinico, Milan, Italy; Dept of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| |
Collapse
|
18
|
Li C, Zhang K, Zhao J. Genome-wide Mendelian randomization mapping the influence of plasma proteome on major depressive disorder. J Affect Disord 2025; 376:1-9. [PMID: 39892755 DOI: 10.1016/j.jad.2025.01.140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Plasma proteins play critical roles in a series of biological processes and represent a major source of translational biomarkers and drug targets. In this study, we performed Mendelian randomization (MR) to explore potential causal associations of protein quantitative trait loci (pQTL, n = 54,219) with major depressive disorder (MDD) using summary statistics from the PGC (n = 143,265) and further replicated in FinnGen cohort (n = 406,986). Subsequently, gene expression quantitative trait loci (eQTL) of identified proteins were leveraged to validate the primary findings in both PGC and FinnGen cohorts. We implemented reverse causality detection using bidirectional MR analysis, Steiger test, Bayesian co-localization and phenotype scanning to further strengthen the MR findings. In primary analyses, MR analysis revealed 2 plasma protein significantly associated with MDD risk at Bonferroni correction (P < 3.720 × 10-5), including butyrophilin subfamily 2 member A1 (BTN2A1, OR = 0.860; 95 % CI, 0.825-0.895; P = 1.79 × 10-5) and butyrophilin subfamily 3 member A2 (BTN3A2, OR = 1.071; 95 % CI, 1.056-1.086; P = 3.89 × 10-6). Both the identified proteins had no reverse causality. Bayesian co-localization indicated that BTN2A1 (coloc.abf-PPH4 = 0.620) and BTN3A2 (coloc.abf-PPH4 = 0.872) exhibited a shared variant with MDD, a finding that was subsequently validated by HEIDI test. In the replication stage, BTN2A1 and BTN3A2 were successfully validated in the FinnGen cohort. This study genetically determined BTN2A1 and BTN3A2 were associated with MDD and these findings may have clinical implications for MDD prevention.
Collapse
Affiliation(s)
- Chong Li
- Department of Psychiatry, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue Zhong, Guangzhou, Guangdong 510220, China
| | - Kunxue Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Dadao Road North, Guangzhou, Guangdong 510515, China
| | - Jiubo Zhao
- Department of Psychiatry, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue Zhong, Guangzhou, Guangdong 510220, China; Department of Psychology, School of Public Health, Southern Medical University, No. 1838 Guangzhou Dadao Road North, Guangzhou, Guangdong 510220, China.
| |
Collapse
|
19
|
Xing L, Wu S, Xue S, Li X. A Novel Neutrophil Extracellular Trap Signature Predicts Patient Chemotherapy Resistance and Prognosis in Lung Adenocarcinoma. Mol Biotechnol 2025; 67:1939-1957. [PMID: 38734842 DOI: 10.1007/s12033-024-01170-1] [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: 01/09/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024]
Abstract
Chemoresistance is a key obstacle in the long-term survival of patients with locally and advanced lung adenocarcinoma (LUAD). This study used bioinformatic analysis to reveal the chemoresistance of gene-neutrophil extracellular traps (NETs) associated with LUAD. RNA sequencing data and LUAD expression patterns were obtained from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, respectively. The GeneCards database was used to identify NETosis-related genes (NRGs). To identify hub genes with significant and consistent expression, differential analysis was performed using the TCGA-LUAD and GEO datasets. LUAD subtypes were determined based on these hub genes, followed by prognostic analysis. Immunological scoring and infiltration analysis were conducted using NETosis scores (N-scores) derived from the TCGA-LUAD dataset. A clinical prognostic model was established and analyzed, and its clinical applications explored. Twenty-two hub genes were identified, and consensus clustering was used to identify two subgroups based on their expression levels. The Kaplan-Meier (KM) curves demonstrated statistically significant differences in prognosis between the two LUAD subtypes. Based on the median score, patients were further divided into high and low N-score groups, and KM curves showed that the N-scores were more precise at predicting the prognosis of patients with LUAD for overall survival (OS). Immunological infiltration analysis revealed significant differences in the abundances of 10 immune cell infiltrates between the high and low N-score groups. Risk scores indicated significant differences in prognosis between the two extreme score groups. The risk scores for the prognostic model also indicated significant differences between the two groups. The results provide new insights into NETosis-related differentially expressed genes (NRDEGs) associated with chemotherapy resistance in patients with LUAD. The established prognostic model is promising and could help with clinical applications to evaluate patient survival and therapeutic efficiency.
Collapse
Affiliation(s)
- Long Xing
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China
- Department of Oncology, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shuangli Wu
- Department of Special Examination, Affiliated Hospital of Qingdao Binhai University, Qingdao, Shandong, China
| | - Shiyue Xue
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Xingya Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
20
|
Pan T, Wu J, Qiu X, Zhu D, Wang J, Li T, Wang Z, Feng F, Xu Y, Zhou X. Identification of potential mechanisms of Schisandrin B in the treatment of idiopathic pulmonary fibrosis by integrating network pharmacology and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5389-5403. [PMID: 39549058 DOI: 10.1007/s00210-024-03605-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a worsening fibrotic condition characterized by a short survival rate and limited treatment options. This study evaluates the potential anti-fibrotic properties of Schisandrin B (Sch B) through network pharmacology and experimental validation. A mouse model of bleomycin-induced pulmonary fibrosis was established, and the modeled mice were treated with Sch B at three doses (20 mg/kg/day, 40 mg/kg/day, and 80 mg/kg/day). A fibrotic model was developed in NIH/3T3 cells by treating them with TGF-β (10 ng/mL) and administering Sch B at various concentrations (10, 20, and 40 µM). The results revealed that Sch B treatment delayed the development of bleomycin-induced pulmonary fibrosis and substantially decreased the transcription levels of collagen I and α-SMA in TGF-β-induced fibroblasts. Core targets were screened with protein-protein interaction network analysis, molecular complex detection (MCODE), and CytoHubba plugin. The application of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and molecular docking highlighted the significance of the HIF-1α signaling pathway in the potential mechanism of Sch B in IPF therapy. Western blot, PCR, and immunofluorescence were performed to validate the effects of Sch B on HIF-1α. In vivo and in vitro, Sch B administration reduced HIF-1α expression. These outcomes provide valuable insights into the potential mechanism by which Sch B delays IPF development, with HIF-1α potentially serving as a key target. However, further investigation is warranted to assess the safety and efficacy of Sch B in clinical settings.
Collapse
Affiliation(s)
- Tingyu Pan
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jieyu Wu
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xirui Qiu
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Dongwei Zhu
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jing Wang
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Tingyuan Li
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhichao Wang
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Fanchao Feng
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yong Xu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Xianmei Zhou
- Department of Pulmonary and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
21
|
Xu J, Lei L, Li P, Huang ZC, Meng Y, He B, Kuang JL. Specnuezhenide and ecliptasaponin A from Ligustrum lucidum Ait and Ecliptae Herba improved premature ovarian failure by targeting the ESR1. J Pharmacol Sci 2025; 158:13-26. [PMID: 40121053 DOI: 10.1016/j.jphs.2025.02.011] [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: 12/23/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
This study was designed to investigate the role of Ligustrum lucidum Ait and Ecliptae Herba on premature ovarian failure (POF) and the underlying mechanisms. In the POF mouse model constructed using cyclophosphamide (CTX), Ligustrum lucidum Ait and Ecliptae Herba increased ovarian index and estradiol (E2) levels and curtailed motility and follicle-stimulating hormone (FSH). Ligustrum lucidum Ait and Ecliptae Herba alleviated ovarian pathological damage in POF mice and promoted the expression of ovarian CD31 and Vascular Endothelial Growth Factor A (VEGFA). Through high-performance liquid chromatography-mass spectrometry (HPLC-MS) and network pharmacology, Specnuezhenide and ecliptasaponin A were identified as the key components of Ligustrum lucidum Ait and Ecliptae Herba in anti-POF action. The important target associated with these components is Estrogen Receptor (ESR) 1. Molecular docking and in vitro experiments showed that Specnuezhenide and ecliptasaponin A can both bind to the ESR protein; knocking down ESR1 inhibited the anti-apoptotic effect of Specnuezhenide and ecliptasaponin A on CTX-induced POF cells. In conclusion, the key components of Ligustrum lucidum Ait and Ecliptae Herba that alleviate POF are Specnuezhenide and ecliptasaponin A, which improve the condition by upregulating ESR1.
Collapse
Affiliation(s)
- Jia Xu
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, PR China
| | - Lei Lei
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Ping Li
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, PR China
| | - Zi-Chun Huang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, PR China
| | - Ying Meng
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, PR China
| | - Bing He
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, PR China.
| | - Ji-Lin Kuang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410005, PR China.
| |
Collapse
|
22
|
Tuo Y, Peng S, Li Y, Dang J, Feng Z, Ding L, Du S, Liu X, Wang L. Quinoa protein and its hydrolysate improve the fatigue resistance of mice: a potential mechanism to relieve oxidative stress and inflammation and improve energy metabolism. J Nutr Biochem 2025; 139:109863. [PMID: 39952621 DOI: 10.1016/j.jnutbio.2025.109863] [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: 09/21/2024] [Revised: 01/05/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Fatigue is commonly marked by reduced endurance and impaired function, often linked to overexertion and chronic conditions. Quinoa (Chenopodium quinoa Willd.), with its rich amino acids and resilience to harsh conditions, offers a novel strategy for combating fatigue. This study explored the antifatigue effects of quinoa protein (QPro) and its hydrolysate (QPH) in weight-loaded swimming mice. After 4 weeks of oral administration, QPro and QPH significantly prolonged swimming duration, reduced serum fatigue biomarkers (lactic acid, urea nitrogen, lactate dehydrogenase, creatine kinase), and elevated glycogen reserves in the liver and muscle. RT-qPCR analysis indicated that QPH activated hepatic gluconeogenesis via G6Pase and PEPCK signaling and enhanced mitochondrial function through PGC-1α/NRF1/TFAM signaling in muscle. Additionally, QPro and QPH boosted antioxidant defenses by improving antioxidant enzyme activity, reducing malondialdehyde through the Nrf2/HO-1 pathway, and suppressing inflammation by reducing TNF-α and IL-6 levels. Network pharmacology identified 31 key targets involved in energy metabolism and inflammation, providing novel insights into the molecular mechanisms underlying the antifatigue properties of quinoa peptides. These findings highlight the potential of QPro and QPH as natural and bioactive ingredients in functional foods for enhancing endurance and mitigating fatigue.
Collapse
Affiliation(s)
- Yuanrong Tuo
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Siwang Peng
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Yiju Li
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Jiamin Dang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Zhi Feng
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Long Ding
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China.
| | - Shuangkui Du
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China; Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, Yangling, Shaanxi, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Liying Wang
- College of Food Science and Engineering, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China; Engineering Research Center of Grain and Oil Functionalized Processing, Universities of Shaanxi Province, Yangling, Shaanxi, China.
| |
Collapse
|
23
|
Joshi CP, Baldi A, Kumar N, Pradhan J. Harnessing network pharmacology in drug discovery: an integrated approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4689-4703. [PMID: 39621088 DOI: 10.1007/s00210-024-03625-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 04/11/2025]
Abstract
Traditional drug discovery approach is based on one drug-one target, that is associated with very lengthy timelines, high costs and very low success rates. Network pharmacology (NP) is a novel method of drug designing, that is based on a multiple-target approach. NP integrates systems such as biology, pharmacology and computational techniques to address the limitations of traditional methods of drug discovery. With help of mapping biological networks, it provides deep insights into biological molecules' interactions and enhances our understanding to the mechanism of drugs, polypharmacology and disease etiology. This review explores the theoretical framework of network pharmacology, discussing the principles and methodologies that enable the construction of drug-target and disease-gene networks. It highlights how data mining, bioinformatics tools and computational models are utilised to predict drug behaviour, repurpose existing drugs and identify novel therapeutic targets. Applications of network pharmacology in the treatment of complex diseases-such as cancer, neurodegenerative disorders, cardiovascular diseases and infectious diseases-are extensively covered, demonstrating its potential to identify multi-target drugs for multifaceted disease mechanisms. Despite the promising results, NP faces challenges due to incomplete and quality of biological data, computational complexities and biological system redundancy. It also faces regulatory challenges in drug approval, demanding revision in regulatory guidelines towards multi-target therapies. Advancements in AI and machine learning, dynamic network modelling and global collaboration can further enhance the efficacy of network pharmacology. This integrative approach has the potential to revolutionise drug discovery, offering new solutions for personalised medicine, drug repurposing and tackling the complexities of modern diseases.
Collapse
Affiliation(s)
- Chandra Prakash Joshi
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Ashish Baldi
- Pharma Innovation Lab, Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India.
| | - Neeraj Kumar
- B N College of Pharmacy, B. N. University, Udaipur, Rajasthan, India
| | - Joohee Pradhan
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan, India.
| |
Collapse
|
24
|
Sharma K, Rai P, Maurya SK, Tapadia MG. Anti-diabetic drug pioglitazone reduces Islet amyloid aggregation overload in the Drosophila neuronal cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6031-6041. [PMID: 39636405 DOI: 10.1007/s00210-024-03632-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Amyloid-proteinopathy is observed in type 2 diabetes, where Islet amyloid polypeptide is secreted atypically and impedes cellular homeostasis. The thiazolidinediones family is reported to influence amyloid-beta aggregations. However, research on drug-based stimulation of insulin signaling to alleviate Islet amyloid aggregations is lacking. To understand the impact of pioglitazone on islet amyloid aggregation, we conducted an in vivo and in silico analysis. For in vivo analysis, we generated a transgenic Drosophila harboring the preproform of human Islet amyloid polypeptide (IAPP) that can be ectopically expressed in a spatio-temporal manner. We show that the unprocessed form of IAPP also has the propensity to form aggregates and cause degeneration. Pioglitazone feeding effectively reduces the burden of Islet amyloid aggregations in the larval brain. In silico analysis shows that there is a higher protein-ligand binding energy for IAPP with pioglitazone than amyloid-beta. These results suggests that pioglitazone might be repurposed as a drug to cure islet amyloidogenesis.
Collapse
Affiliation(s)
- Khushboo Sharma
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Pooja Rai
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Faculty of Science, University of Delhi, Delhi, 110007, India
| | - Madhu G Tapadia
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
25
|
Oyabu M, Ohira Y, Fujita M, Yoshioka K, Kawaguchi R, Kubo A, Hatazawa Y, Yukitoshi H, Ortuste Quiroga HP, Horii N, Miura F, Araki H, Okano M, Hatada I, Gotoh H, Yoshizawa T, Fukada SI, Ogawa Y, Ito T, Ishihara K, Ono Y, Kamei Y. Dnmt3a overexpression disrupts skeletal muscle homeostasis, promotes an aging-like phenotype, and reduces metabolic elasticity. iScience 2025; 28:112144. [PMID: 40151644 PMCID: PMC11937683 DOI: 10.1016/j.isci.2025.112144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/10/2024] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Mammalian aging is reportedly driven by the loss of epigenetic information; however, its impact on skeletal muscle aging remains unclear. This study shows that aging mouse skeletal muscle exhibits increased DNA methylation, and overexpression of DNA methyltransferase 3a (Dnmt3a) induces an aging-like phenotype. Muscle-specific Dnmt3a overexpression leads to an increase in central nucleus-positive myofibers, predominantly in fast-twitch fibers, a shift toward slow-twitch fibers, elevated inflammatory and senescence markers, mitochondrial OXPHOS complex I reduction, and decreased basal autophagy. Dnmt3a overexpression resulted in reduced muscle mass and strength and impaired endurance exercise capacity with age, accompanied by an enhanced inflammatory signature. In addition, Dnmt3a overexpression reduced not only sensitivity to starvation-induced muscle atrophy but also the restorability from muscle atrophy. These findings suggest that increased DNA methylation disrupts skeletal muscle homeostasis, promotes an aging-like phenotype, and reduces muscle metabolic elasticity.
Collapse
Affiliation(s)
- Mamoru Oyabu
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Yuto Ohira
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Mariko Fujita
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Kiyoshi Yoshioka
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
- Institute for Research on Productive Aging (IRPA), Tokyo, Japan
| | - Runa Kawaguchi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Atsushi Kubo
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yukino Hatazawa
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Hinako Yukitoshi
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Huascar Pedro Ortuste Quiroga
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Naoki Horii
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Masaki Okano
- Department of Pluripotent Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
- Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Maebashi 371-8511, Japan
| | - Hitoshi Gotoh
- Cell Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan
| | - Tatsuya Yoshizawa
- Cell Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 606-0823, Japan
| | - So-ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Kengo Ishihara
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University, Shiga 520-2194, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo 173-0015, Japan
| | - Yasutomi Kamei
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| |
Collapse
|
26
|
Wang YX, Fei CJ, Shen C, Ou YN, Liu WS, Yang L, Wu BS, Deng YT, Feng JF, Cheng W, Yu JT. Exome sequencing identifies protein-coding variants associated with loneliness and social isolation. J Affect Disord 2025; 375:192-204. [PMID: 39842675 DOI: 10.1016/j.jad.2025.01.096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/31/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Loneliness and social isolation are serious yet underappreciated public health problems, with their genetic underpinnings remaining largely unknown. We aimed to explore the role of protein-coding variants in the manifestation of loneliness and social isolation. METHODS We conducted the first exome-wide association analysis on loneliness and social isolation, utilizing 336,115 participants of white-British ancestry for loneliness and 346,115 for social isolation. Sensitivity analyses were performed to validate the genetic findings. We estimated the genetic burden heritability of loneliness and social isolation and provided biological insights into them. RESULTS We identified six novel risk genes (ANKRD12, RIPOR2, PTEN, ARL8B, NF1, and PIMREG) associated with loneliness and two (EDARADD and GIGYF1) with social isolation through analysis of rare coding variants. Brain-wide association analysis uncovered 47 associations between identified genes and brain structure phenotypes, many of which are critical for social processing and interaction. Phenome-wide association analysis established significant links between these genes and phenotypes across five categories, mostly blood biomarkers and cognitive measures. LIMITATIONS The measurements of loneliness and social isolation in UK Biobank are brief for these multi-layer social factors, some relevant aspects may be missed. CONCLUSIONS Our study revealed 13 risk genes associated with loneliness and 6 with social isolation, with the majority being novel discoveries. These findings advance our understanding of the genetic basis of these two traits. The study provides a foundation for future studies aimed at exploring the functional mechanisms of these genes and their potential implications for public health interventions targeting loneliness and social isolation.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Chen-Jie Fei
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Chun Shen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
Lin Q, Zhang D, Gruber PJ, Tam PKH, Lui VCH, Wu Z, Hong H, Chien KR, Sham PC, Tang CSM. Multifaceted analysis of noncoding and coding de novo variants implicates NOTCH signaling pathway in tetralogy of Fallot in Chinese population. HGG ADVANCES 2025; 6:100414. [PMID: 39921258 PMCID: PMC11910093 DOI: 10.1016/j.xhgg.2025.100414] [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: 09/17/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
Tetralogy of Fallot (TOF) is the most common cyanotic heart defect in neonates. While there is compelling evidence of genetic contribution to the etiology of TOF, the contribution of noncoding variants to the development of the defect remains unexplored. Potentially damaging noncoding de novo variants (NC DNVs) were detected from 141 Chinese nonsyndromic TOF trios (CHN-TOF) and compared with those detected in the Pediatric Cardiac Genomics Consortium (PCGC). Bioinformatic analyses on noncoding and previously detected coding DNVs were performed to identify developmental pathways affected in TOF. Chinese but not PCGC-TOF patients showed a notably increased burden of putative damaging NC DNVs (n = 249). In Chinese, NC and coding DNVs were predominantly associated with cardiomyocyte differentiation and with chamber/valve/aorta development, respectively, producing a combined enrichment in NOTCH signaling (p = 1.1 × 10-6) and outflow tract morphogenesis (p = 2.2 × 10-5). Genes with NC DNVs (e.g., EFNB2, HEY2, and PITX2) interacted with NOTCH1 and FLT4 in a tight STRING protein-protein interaction (PPI) network. During the in vitro cardiac differentiation process, these noncoding candidate genes, which harbored potentially damaging regulatory NC DNVs, exhibited co-expression with NOTCH signaling genes and demonstrated dysregulated gene expression at various differentiation stages following NOTCH1 downregulation. In summary, our findings highlight a significant contribution of NC DNVs to TOF and suggest the presence of population genetic heterogeneity. Integrative analyses implicate dysregulation of NOTCH signaling, with converging influences from both coding and noncoding variants, in TOF within the Chinese population.
Collapse
Affiliation(s)
- Qiongfen Lin
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Detao Zhang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Peter J Gruber
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Paul Kwong-Hang Tam
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Faculty of Medicine, Macau University of Science and Technology, Macao, China
| | - Vincent Chi-Hang Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhongluan Wu
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Haifa Hong
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kenneth R Chien
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Pak Chung Sham
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Clara Sze-Man Tang
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Dr Li Dak-Sum Research Centre, The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong, China.
| |
Collapse
|
28
|
Schaffer LV, Hu M, Qian G, Moon KM, Pal A, Soni N, Latham AP, Pontano Vaites L, Tsai D, Mattson NM, Licon K, Bachelder R, Cesnik A, Gaur I, Le T, Leineweber W, Palar A, Pulido E, Qin Y, Zhao X, Churas C, Lenkiewicz J, Chen J, Ono K, Pratt D, Zage P, Echeverria I, Sali A, Harper JW, Gygi SP, Foster LJ, Huttlin EL, Lundberg E, Ideker T. Multimodal cell maps as a foundation for structural and functional genomics. Nature 2025:10.1038/s41586-025-08878-3. [PMID: 40205054 DOI: 10.1038/s41586-025-08878-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 03/10/2025] [Indexed: 04/11/2025]
Abstract
Human cells consist of a complex hierarchy of components, many of which remain unexplored1,2. Here we construct a global map of human subcellular architecture through joint measurement of biophysical interactions and immunofluorescence images for over 5,100 proteins in U2OS osteosarcoma cells. Self-supervised multimodal data integration resolves 275 molecular assemblies spanning the range of 10-8 to 10-5 m, which we validate systematically using whole-cell size-exclusion chromatography and annotate using large language models3. We explore key applications in structural biology, yielding structures for 111 heterodimeric complexes and an expanded Rag-Ragulator assembly. The map assigns unexpected functions to 975 proteins, including roles for C18orf21 in RNA processing and DPP9 in interferon signalling, and identifies assemblies with multiple localizations or cell type specificity. It decodes paediatric cancer genomes4, identifying 21 recurrently mutated assemblies and implicating 102 validated new cancer proteins. The associated Cell Visualization Portal and Mapping Toolkit provide a reference platform for structural and functional cell biology.
Collapse
Affiliation(s)
- Leah V Schaffer
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mengzhou Hu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gege Qian
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
| | - Kyung-Mee Moon
- Department of Biochemistry & Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Abantika Pal
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Neelesh Soni
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Andrew P Latham
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | | | - Dorothy Tsai
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nicole M Mattson
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Katherine Licon
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robin Bachelder
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anthony Cesnik
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Ishan Gaur
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Trang Le
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | | | - Aji Palar
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Ernst Pulido
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Yue Qin
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Xiaoyu Zhao
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Christopher Churas
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joanna Lenkiewicz
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Chen
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Keiichiro Ono
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Dexter Pratt
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Peter Zage
- Department of Pediatrics, Division of Hematology-Oncology, University of California San Diego, La Jolla, CA, USA
| | - Ignacia Echeverria
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, USA
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Leonard J Foster
- Department of Biochemistry & Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| | - Emma Lundberg
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA.
- Department of Pathology, Stanford University, Palo Alto, CA, USA.
- Science for Life Laboratory, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
29
|
Jerabkova-Roda K, Peralta M, Huang KJ, Mousson A, Bourgeat Maudru C, Bochler L, Busnelli I, Karali R, Justiniano H, Lisii LM, Carl P, Mittelheisser V, Asokan N, Larnicol A, Lefebvre O, Lachuer H, Pichot A, Stemmelen T, Molitor A, Scheid L, Frenger Q, Gros F, Hirschler A, Delalande F, Sick E, Carapito R, Carapito C, Lipsker D, Schauer K, Rondé P, Hyenne V, Goetz JG. Peripheral positioning of lysosomes supports melanoma aggressiveness. Nat Commun 2025; 16:3375. [PMID: 40204688 PMCID: PMC11982396 DOI: 10.1038/s41467-025-58528-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Emerging evidence suggests that the function and position of organelles are pivotal for tumor cell dissemination. Among them, lysosomes stand out as they integrate metabolic sensing with gene regulation and secretion of proteases. Yet, how their function is linked to their position and how this controls metastasis remains elusive. Here, we analyze lysosome subcellular distribution in patient-derived melanoma cells and patient biopsies and show that lysosome spreading scales with melanoma aggressiveness. Peripheral lysosomes promote matrix degradation and cell invasion which is directly linked to the lysosomal and cell transcriptional programs. Using chemo-genetical control of lysosome positioning, we demonstrate that perinuclear clustering impairs lysosome secretion, matrix degradation and invasion. Impairing lysosome spreading significantly reduces invasive outgrowth in two in vivo models, mouse and zebrafish. Our study provides a direct demonstration that lysosome positioning controls cell invasion, illustrating the importance of organelle adaptation in carcinogenesis and suggesting its potential utility for diagnosis of metastatic melanoma.
Collapse
Affiliation(s)
- Katerina Jerabkova-Roda
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
- Institut Curie, PSL, CNRS, UMR144, Paris, France.
| | - Marina Peralta
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, 00015, Rome, Italy
| | - Kuang-Jing Huang
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Antoine Mousson
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Clara Bourgeat Maudru
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Louis Bochler
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Ignacio Busnelli
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Rabia Karali
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Hélène Justiniano
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Lucian-Mihai Lisii
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Philippe Carl
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Vincent Mittelheisser
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Nandini Asokan
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Annabel Larnicol
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Olivier Lefebvre
- Tumor Biomechanics, Strasbourg, France
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France
| | - Hugo Lachuer
- Institut Curie, PSL, CNRS, UMR144, Paris, France
- Institut Gustave Roussy, INSERM UMR1279, Université Paris-Saclay, Villejuif, France
- Université de Paris, CNRS, Institut Jacques Monod, 75013, Paris, France
| | - Angélique Pichot
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Tristan Stemmelen
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
| | - Anne Molitor
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | - Léa Scheid
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Quentin Frenger
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Frédéric Gros
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Aurélie Hirschler
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - François Delalande
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Emilie Sick
- Université de Strasbourg, Strasbourg, France
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France
| | - Raphaël Carapito
- INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Plateforme GENOMAX, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Fédération Hospitalo-Universitaire OMICARE, Strasbourg, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | - Christine Carapito
- Laboratoire de Spectrométrie de Masse Bio-Organique (LSMBO), IPHC, UMR 7178, CNRS, Université de Strasbourg, Infrastructure Nationale de Protéomique ProFI, FR2048, Strasbourg, France
| | - Dan Lipsker
- Faculté de Médecine, Université de Strasbourg et Clinique Dermatologique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Kristine Schauer
- Institut Curie, PSL, CNRS, UMR144, Paris, France.
- Institut Gustave Roussy, INSERM UMR1279, Université Paris-Saclay, Villejuif, France.
| | - Philippe Rondé
- Université de Strasbourg, Strasbourg, France.
- CNRS UMR7021, Faculté de Pharmacie, Illkirch, France.
| | - Vincent Hyenne
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
- CNRS, SNC5055, Strasbourg, France.
| | - Jacky G Goetz
- Tumor Biomechanics, Strasbourg, France.
- INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Strasbourg, France.
| |
Collapse
|
30
|
Zheng J, Jiao Z, Yang X, Ruan Q, Huang Y, Jin C, Gui S, Xuan Z, Jia X. Network pharmacology-based exploration of the mechanism of Wenweishu granule in treating chronic atrophic gastritis with spleen-stomach cold deficiency syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119591. [PMID: 40054637 DOI: 10.1016/j.jep.2025.119591] [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: 12/12/2024] [Revised: 02/14/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wenweishu (WWS) is a traditional Chinese medicine compound formulated for chronic atrophic gastritis (CAG) treatment by warming the stomach and alleviating pain. However, its pharmacological mechanisms remain underexplored. AIM OF THE STUDY This study investigated the therapeutic effects and potential mechanisms of WWS on CAG with spleen-stomach cold deficiency syndrome (SSCDS). METHODS To achieve this, an SSCDS-CAG rat model and a human gastric mucosal epithelial cells (GES-1) cell model were established using multi-factor modeling and N-Methyl-N'-nitro-N-nitrosoguanidine (MNNG) induction, respectively. WWS's effects on gastric injury were evaluated through pathology, inflammation, serum biomarkers, and apoptosis. Additionally, MNNG's effects on GES-1 cells were analyzed. Network pharmacology, involving protein-protein interaction networks, GO/KEGG enrichment, and molecular docking, was employed to predict WWS's potential targets and mechanisms in SSCDS-CAG. Mechanistic insights were further validated using immunohistochemistry, quantitative reverse transcription polymerase chain reaction, and western blotting. RESULTS In vivo results showed that WWS alleviated symptoms in SSCDS-CAG rats, lowering symptom scores and improving gastric histopathology. It modulated serum biomarkers and reduced inflammation and apoptosis in both in vivo and in vitro studies. Network pharmacology results revealed 263 overlapping targets between WWS and SSCDS-CAG, associated with apoptosis, inflammation, and the PI3K/AKT pathway. Molecular docking revealed strong binding affinity between the core target and active WWS components. In SSCDS-CAG rats and GES-1 cells, WWS inhibited PI3K/AKT phosphorylation, increased PTEN expression, and regulated Bcl-2, Bax, and cleaved caspase-3 levels. CONCLUSION WWS reduces inflammation and apoptosis in multi-factor CAG rats and MNNG-induced GES-1 cells by modulating the PTEN/PI3K/AKT signaling pathway and apoptosis-related proteins.
Collapse
Affiliation(s)
- Jia Zheng
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Zhiyong Jiao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Xinyu Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Qing Ruan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Yuzhe Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Cheng Jin
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Shuangying Gui
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Zihua Xuan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China
| | - Xiaoyi Jia
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Bioactive Natural Products, Hefei, 230012, China.
| |
Collapse
|
31
|
Li Y, Yu X, Liu Y, Miao S, Liu X, Wang Z, Zhou H. Pharmacodynamic components and molecular mechanism of Gastrodia elata Blume in treating hypertension: Absorbed components, network pharmacology analysis, molecular docking and in vivo experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119583. [PMID: 40058475 DOI: 10.1016/j.jep.2025.119583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/18/2025] [Accepted: 03/02/2025] [Indexed: 03/21/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rhizome of Gastrodia elata Blume (RGE) is a valuable traditional Chinese Medicine (TCM) in the clinical practice. The Compendium of Materia Medica records that RGE has the effect of flatting liver wind out. It has sedative, analgesic, hypnotic, anticonvulsant, anti-hypertensive, anti-myocardial ischemia, anti-arrhythmic and anti-platelet aggregation effects. RGE is often used to relieve and treat vertigo, headache, hypertension, convulsions, and epilepsy in TCM clinic for thousands of years. Accumulated evidences have suggested that hypertension disease is related to the renin-angiotensin-aldosterone system (RAAS) disturbance. However, the potential pharmacodynamic components and anti-hypertensive mechanisms of RGE are unclear now. AIM OF THE STUDY The active component and mechanism of RGE in treating hypertension were elucidated to strengthen the quality control and development of anti-hypertensive drugs. MATERIALS AND METHODS The anti-hypertensive active components of RGE were analyzed by multi-dimensional qualitative analysis method including ethanol extract, in-vitro intestinal absorption, in-vivo plasma. The ultra high performance liquid chromatography-mass spectrometry (UPLC-Q-Exactive MS/MS) analysis technology was adopted to identify these components. Network pharmacology was applied to predicted anti-hypertensive active components, target proteins and pathways. Molecular docking was used to evaluate the potential molecular binding modes between 68 components and nine proteins. Spontaneously hypertensive rats (SHR) model was adopted to evaluate the activity of reducing systolic and diastolic blood pressure (SBP and DBP). Levels of renin, angiotcnsin II (Ang II) and aldosterone (ALD) in serum were determined by Elisa kit. Immunohistochemical were adopted to compare the changes of Ang II receptor 1 (AT1R) protein levels in SHR model and RGE groups. RESULTS The multi-dimensional components qualitative analysis method of RGE was established. The results showed that 79, 70 and 30 components were identified in RGE ethanol extract, in-vitro intestinal absorption and in-vivo plasma, respectively. These components were mainly parishins, nucleosides, amino acids, phenolic acids, flavonoids, organic acids et al. Network pharmacology results showed that anti-hypertensive active components were nucleosides and organic acids. It was speculated that RGE could exert its anti-hypertensive effect by regulating aldosterone-regulated sodium reabsorption, renin-angiotensin system pathways and related target proteins. Molecular docking results showed that 21 components including parishins, nucleosides and phenolic acids were potential active components of anti-hypertensive. Taking together, parishin A, B, E, C, D, adenosine, N6-(4-hydroxybenzyl) adenosine, guanosine, ferulic acid were the main anti-hypertensive active components of RGE. Pharmacodynamic results showed that RGE (0.7 g·kg-1) at low dosage could reduce SBP and DBP of SHR in vivo. Meanwhile, RGE (1.4 g·kg-1) markedly reduced the contents of renin, angiotcnsin II and ALD (p < 0.05) of SHR. Immunohistochemical data demonstrated that RGE (0.7 g·kg-1) could downregulate the protein expression of AT1R. In general, RGE can significantly reduce blood pressure of SHR by regulating RAAS. CONCLUSION The multi-dimensional components qualitative analysis combining network pharmacology and molecular docking technology provide a new perspective for discovering potential anti-hypertensive components of RGE. RGE possess anti-hypertensive activity by regulating multiple targets of RAAS. Thus, it has the potential to develop into the novel raw material of anti-hypertensive drugs.
Collapse
Affiliation(s)
- Yun Li
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Xiaofei Yu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yezhi Liu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shuxin Miao
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaoqian Liu
- National Engineering Laboratory for Quality Control Technology of Chinese Herbal Medicines, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhimin Wang
- National Engineering Laboratory for Quality Control Technology of Chinese Herbal Medicines, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Honglei Zhou
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
32
|
Gao R, Zhang X, Ju H, Zhou Y, Yin L, Yang L, Wu P, Sun X, Fang H. Telocyte-derived exosomes promote angiogenesis and alleviate acute respiratory distress syndrome via JAK/STAT-miR-221-E2F2 axis. MOLECULAR BIOMEDICINE 2025; 6:21. [PMID: 40198510 PMCID: PMC11979044 DOI: 10.1186/s43556-025-00259-6] [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: 08/21/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by severe respiratory failure and significant inflammation, leading to vascular and epithelial cell damage. The absence of effective pharmacologic treatments underscores the need for novel therapeutic approaches. Telocytes (TCs), a newly identified type of interstitial cells, have shown potential in tissue repair and angiogenesis, particularly through the release of exosomal microRNAs (miRNAs). Exosomes were isolated from LPS (lipopolysaccharide)-stimulated TCs and characterized using western blotting and nanoparticle tracking analysis. The role of exosomal miR-221 in angiogenesis was assessed through tube formation, migration, and proliferation assays in mouse vascular endothelial cells (MVECs). The JAK/STAT pathway's involvement in miR-221 regulation was determined using western blotting and qRT-PCR. A dual-luciferase assay confirmed E2F2 as a direct target of miR-221. ARDS mouse model was established via LPS instillation, and the therapeutic effects of TCs-derived exosomes were evaluated by histopathological scoring, cytokine analysis, and endothelial barrier integrity assays. Our findings demonstrated that exosomes from LPS-stimulated TCs significantly promoted angiogenesis, proliferation, and migration in MVECs. These effects were mediated by miR-221, which downregulated E2F2 expression, an important regulator of endothelial cell functions. The JAK/STAT pathway played a crucial role in miR-221 production, with pathway inhibition reducing miR-221 levels and attenuating its pro-angiogenic effects. In vivo, TCs-derived exosomes reduced lung inflammation and tissue damage in ARDS mice, effects that were reversed by miR-221 inhibition. These results suggested that TCs-derived exosomes promoted angiogenesis and alleviated ARDS through the JAK/STAT-miR-221-E2F2 axis.
Collapse
Affiliation(s)
- Rongrong Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Clinical Center for Biotherapy at Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Zhang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Huihui Ju
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yile Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Luoyue Yin
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liuke Yang
- College of Plant Protection, Nanjing Agricultural University, Nanjing, 210000, China
| | - Pinwen Wu
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Xia Sun
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai, China.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai, China.
| |
Collapse
|
33
|
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
|
34
|
Lai Y, Su B, Wang X, Zeng C, Chu H, Zhou L, Bing D. Transcriptomic analysis reveals the function of m6A regulators in aged cochlea. Braz J Otorhinolaryngol 2025; 91:101578. [PMID: 40198960 DOI: 10.1016/j.bjorl.2025.101578] [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: 09/18/2024] [Revised: 01/22/2025] [Accepted: 02/03/2025] [Indexed: 04/10/2025] Open
Abstract
OBJECTIVE Presbycusis is a prevalent health issue among the elderly. Previous studies have shown mechanisms related to this condition, but the underlying mechanisms of presbycusis remain elusive. N6-methyladenosine (m6A) modification in regulating gene expression and cellular functions has been implicated in the development of various diseases. Nevertheless, the potential role of m6A regulators in presbycusis is still unclear. In this study, we aim to determine the expression of m6A regulators in the cochleae of young and old mice, and to investigate their potential role in aging. METHODS We sequenced the transcriptome from the cochleae of six young (2-mo) and six old mice (24-mo) bioinformatics analysis. Differential expression analysis and downstream functional analysis was performed to identify m6A regulators. Association of m6A regulators with protein-protein interaction and transcription factor-miRNA networks were constructed to explore their regulatory mechanisms. RESULTS ALKBH5 and YTHDC1 were found upregulated in the aged cochleae. They were strongly correlated with immune-related pathways, immune molecular subtypes, and immune infiltration levels in old mice, suggesting their potential involvement in immune-related mechanisms of presbycusis. Receiver Operating Characteristic (ROC) curve analysis demonstrated the high diagnostic potential molecules of AlkB Homolog 5 (ALKBH5) and YTHDC1. CONCLUSION This study has established a molecular foundation and introduce a novel perspective on the role of m6A regulators in presbycusis, emphasizing ALKBH5 and YTHDC1 as potential markers. LEVEL OF EVIDENCE Acknowledging methodological similarities with Level 3 (non-randomized controlled cohort or case-control studies) in clinical research, we reference Level 3 as a comparative framework, while recognizing the distinct differences between clinical and animal research settings.
Collapse
Affiliation(s)
- Yanbing Lai
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Bo Su
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Xiaodi Wang
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Chenghui Zeng
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Hanqi Chu
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Liangqiang Zhou
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China
| | - Dan Bing
- Tongji Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Department of Otorhinolaryngology Head and Neck Surgery, Wuhan, China.
| |
Collapse
|
35
|
Lee JJ, Yang L, Kotzin JJ, Ahimovic D, Bale MJ, Nigrovic PA, Josefowicz SZ, Mathis D, Benoist C. Early transcriptional effects of inflammatory cytokines reveal highly redundant cytokine networks. J Exp Med 2025; 222:e20241207. [PMID: 39873673 DOI: 10.1084/jem.20241207] [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: 07/15/2024] [Revised: 09/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
Inflammatory cytokines are fundamental mediators of the organismal response to injury, infection, or other harmful stimuli. To elucidate the early and mostly direct transcriptional signatures of inflammatory cytokines, we profiled all immunologic cell types by RNAseq after systemic exposure to IL1β, IL6, and TNFα. Our results revealed a significant overlap in the responses, with broad divergence between myeloid and lymphoid cells, but with very few cell-type-specific responses. Pathway and motif analysis identified several main controllers (NF-κB, IRF8, and PU.1), but the largest portion of the response appears to be mediated by MYC, which was also implicated in the response to γc cytokines. Indeed, inflammatory and γc cytokines elicited surprisingly similar responses (∼50% overlap in NK cells). Significant overlap with interferon-induced responses was observed, paradoxically in lymphoid but not myeloid cell types. These results point to a highly redundant cytokine network, with intertwined effects between disparate cytokines and cell types.
Collapse
Affiliation(s)
- Juliana J Lee
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Liang Yang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Jonathan J Kotzin
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dughan Ahimovic
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences , New York, NY, USA
| | - Michael J Bale
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences , New York, NY, USA
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences , New York, NY, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard , Cambridge, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard , Cambridge, MA, USA
| |
Collapse
|
36
|
Chen H, Charles PD, Gu Q, Liberatori S, Robertson DL, Palmarini M, Wilson SJ, Mohammed S, Castello A. Omics analyses uncover host networks defining virus-permissive and -hostile cellular states. Mol Cell Proteomics 2025:100966. [PMID: 40204275 DOI: 10.1016/j.mcpro.2025.100966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/24/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025] Open
Abstract
The capacity of host cells to sustain or restrict virus infection is influenced by their proteome. Understanding the compendium of proteins defining cellular permissiveness is key to many questions in fundamental virology. Here, we apply a multiomic approach to determine the proteins that are associated with highly permissive, intermediate, and hostile cellular states. We observed two groups of differentially regulated genes: i) with robust changes in mRNA and protein levels, and ii) with protein/RNA discordances. Whereas many of the latter are classified as interferon stimulated genes (ISGs), most exhibit no antiviral effects in overexpression screens. This suggest that IFN-dependent protein changes can be better indicators of antiviral function than mRNA levels. Phosphoproteomics revealed an additional regulatory layer involving non-signalling proteins with altered phosphorylation. Indeed, we confirmed that several permissiveness-associated proteins with changes in abundance or phosphorylation regulate infection fitness. Altogether, our study provides a comprehensive and systematic map of the cellular alterations driving virus susceptibility.
Collapse
Affiliation(s)
- Honglin Chen
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland (UK); Department of Biochemistry, University of Oxford, South Parks Road, Oxford, UK
| | | | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland (UK)
| | - Sabrina Liberatori
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, UK
| | - David L Robertson
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland (UK)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland (UK)
| | - Sam J Wilson
- Cambridge Institute of Therapeutic Immunol & Infect Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Puddicombe Way, UK
| | - Shabaz Mohammed
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, UK; The Rosalind Franklin Institute, Oxfordshire, UK; Department of Chemistry, University of Oxford, Mansfield Road, Oxford, UK.
| | - Alfredo Castello
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland (UK).
| |
Collapse
|
37
|
Cao F, Zhao X, Fu X, Jin Y. Computational insights into exploring the potential effects of environmental contaminants on human health. Sci Rep 2025; 15:11779. [PMID: 40189682 PMCID: PMC11973197 DOI: 10.1038/s41598-025-96193-2] [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: 10/31/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
With rapid industrialization and urbanization, the increasing prevalence of air and water pollutants poses a significant threat to public health. Traditional research methods, such as epidemiological studies and in vitro/in vivo experiments, provide valuable biological insights but are often costly, time-consuming, and limited in scale. To address this gap, this study develops a machine learning-based approach to predict the carcinogenicity of pollutants. Using the dataset of carcinogenic and non-carcinogenic molecules that we collected, the pretrained KPGT model trained with molecular fingerprints and descriptors achieved an AUC of 0.83, surpassing traditional machine learning models. To validate this model, common pollutants from air and water sources were analyzed. Further clustering classified these pollutants into five distinct groups. Target prediction analysis identified key genes associated with representative pollutant molecules, such as MAPK1, MTOR, and PTPN11. GO and KEGG pathway analyses, along with survival analysis, revealed potential carcinogenic mechanisms and prognostic implications. Our findings contribute to improved pollution risk assessment and evidence-based environmental policy development, ultimately aiding in the mitigation of pollutant-related health risks.
Collapse
Affiliation(s)
- Fuyan Cao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xinyue Zhao
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xueqi Fu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, China
- National Engineering Laboratory of AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Yue Jin
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, China.
- Edmond H. Fischer Signal Transduction Laboratory, School of Life Sciences, Jilin University, Changchun, 130012, China.
- National Engineering Laboratory of AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China.
| |
Collapse
|
38
|
Potemkin N, Cawood SMF, Guévremont D, Mockett B, Treece J, Stanton JAL, Williams JM. Whole Transcriptome RNA-Seq Reveals Drivers of Pathological Dysfunction in a Transgenic Model of Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04878-6. [PMID: 40186694 DOI: 10.1007/s12035-025-04878-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 03/20/2025] [Indexed: 04/07/2025]
Abstract
Alzheimer's disease (AD) affects more than 55 million people worldwide, yet current theories cannot fully explain its aetiology. Accordingly, gene expression profiling has been used to provide a holistic view of the biology underpinning AD. Focusing primarily on protein-coding genes, such approaches have highlighted a critical involvement of microglia-related inflammatory processes. Simultaneous investigation of transcriptional regulators and noncoding RNA (ncRNA) can offer further insight into AD biology and inform the development of disease-modifying therapies. We previously described a method for whole transcriptome sampling to simultaneously investigate protein-coding genes and ncRNA. Here, we use this technique to explore transcriptional changes in a murine model of AD (15-month-old APP/PS1 mice). We confirmed the extensive involvement of microglia-associated genes and gene networks, consistent with literature. We also report a wealth of differentially-expressed non-coding RNA - including microRNA, long non-coding RNA, small nuclear and small nucleolar RNA, and pseudogenes - many of which have been overlooked previously. Transcription factor analysis determined that six transcription factors likely regulate gene expression changes in this model (Irf8, Junb, c-Fos, Lmo2, Runx1, and Nfe2l2). We then utilised validated miRNA-target interactions, finding 60 interactions between 15 miRNA and 42 mRNA (messenger RNA) with largely consistent directionality. Furthermore, we found that eight transcription factors (Clock, Lmo2, Runx1, Nfe2l2, Egr2, c-Fos, Junb, and Nr4a1) are likely responsible for the regulation of miRNA expression. Taken together, these data indicate a complex interplay of coding and non-coding RNA, driven by a small number of specific transcription factors, contributing to transcriptional changes in 15-month-old APP/PS1 mice.
Collapse
Affiliation(s)
- Nikita Potemkin
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Sophie M F Cawood
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Diane Guévremont
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
| | - Bruce Mockett
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand
- Department of Psychology, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Jackson Treece
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Jo-Ann L Stanton
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand
| | - Joanna M Williams
- Department of Anatomy, School of Biomedical Sciences, University of Otago, P.O. Box 56, Dunedin, New Zealand.
- Brain Health Research Centre, Brain Research New Zealand-Rangahau Roro Aotearoa, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
39
|
Candia J, Fantoni G, Moaddel R, Delgado-Peraza F, Shehadeh N, Tanaka T, Ferrucci L. Effects of in vitro hemolysis and repeated freeze-thaw cycles in protein abundance quantification using the SomaScan and Olink assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.21.613295. [PMID: 40166260 PMCID: PMC11956925 DOI: 10.1101/2024.09.21.613295] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
SomaScan and Olink are affinity-based platforms that aim to estimate the relative abundance of thousands of human proteins with a broad range of endogenous concentrations. In this study, we investigated the effects of in vitro hemolysis and repeated freeze-thaw cycles in protein abundance quantification across 10,776 (11K SomaScan) and 1472 (Olink Explore 1536) analytes, respectively. Using SomaScan, we found two distinct groups, each one consisting of 4% of all aptamers, affected by either hemolysis or freeze-thaw cycles. Using Olink, we found 6% of analytes affected by freeze-thaw cycles and nearly half of all measured probes significantly impacted by hemolysis. Moreover, we observed that Olink probes affected by hemolysis target proteins with a larger number of annotated protein-protein interactions. We found that Olink probes affected by hemolysis were significantly associated with the erythrocyte proteome, whereas SomaScan probes were not. Given the extent of the observed nuisance effects, we propose that unbiased, quantitative methods of evaluating hemolysis, such as the hemolysis index successfully implemented in many clinical laboratories, should be adopted in proteomics studies. We provide detailed results for each SomaScan and Olink probe in the form of extensive Supplementary Data files to be used as resources for the growing user communities of both platforms.
Collapse
|
40
|
Sun X, Hu X, Wei J, An H. Uncovering leading compounds for alzheimer's disease treatment: mendelian randomization and virtual screening insights into plasma protein modulation. Biol Res 2025; 58:19. [PMID: 40186323 PMCID: PMC11971886 DOI: 10.1186/s40659-025-00598-2] [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: 11/07/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder influenced by both genetic and environmental factors. Identifying therapeutic targets and interventions remains challenging. This study utilized Mendelian Randomization (MR) to investigate causal relationships between plasma proteins, lifestyle factors, and AD, along with virtual screening to identify potential drug compounds. A two-sample MR analysis assessed associations between plasma proteins, identified through genome-wide association studies (GWAS), and AD risk. Co-localization analysis (CA) confirmed the overlap between protein expression and AD susceptibility loci, and reverse MR ruled out reverse causality. A protein-protein interaction (PPI) network was constructed to explore therapeutic targets, followed by virtual screening to identify small-molecule inhibitors for selected proteins. The analysis found significant associations between eight plasma proteins and AD, with five proteins (GSTP1, BIN1, Siglec-3, SERPINF2, and GRN) showing strong evidence of involvement in AD pathogenesis. Virtual screening identified six compounds as potential inhibitors of GSTP1 and four compounds as potential inhibitors of BIN1. Furthermore, MR analysis of lifestyle factors, such as dietary behaviors and smoking cessation, indicated they may influence AD risk through their effects on specific proteins. These findings offer novel insights into the genetic mechanisms underlying AD and highlight the potential of combining MR with virtual screening to identify therapeutic targets. The study also suggests that lifestyle modifications could offer alternative prevention and treatment strategies for AD. Future research should focus on the experimental validation of the identified compounds and further explore the mechanisms linking lifestyle factors to AD.
Collapse
Affiliation(s)
- Xiaohan Sun
- School of Science, Hong Kong University of Science and Technology, Hong Kong, People's Republic of China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Haoyu An
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
41
|
Wang Q, Zhong L, Hua L, Pang S, Li Y, Zhang Z, Zhao J, Huang H. Molecular Mechanisms in Idiopathic Mitral Valve Chordae Tendineae Rupture: Insights from Transcriptome Analysis and Inflammation Evaluation. J Inflamm Res 2025; 18:4771-4783. [PMID: 40206810 PMCID: PMC11980926 DOI: 10.2147/jir.s510525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
Objective This study investigates the molecular mechanisms and hub genes in idiopathic rupture of mitral valve chordae tendineae (iRCT). Methods Histological changes were assessed via pathological staining, and transcriptome sequencing was performed on samples from 8 iRCT patients and 6 controls. Differentially expressed genes (DEGs), functional enrichment, PPI networks, and immune cell infiltration were analyzed. Hub gene expression was validated using RT-qPCR. Results iRCT samples exhibited cell proliferation, disorganized collagen fibers, and elastin fiber rupture. Immunohistochemical analysis further confirmed that activated fibroblasts, macrophages, dendritic cells, and T cells were increased in iRCT samples compared to normal samples. Additionally, iRCT samples exhibited an increased content of collagen fibers and elastin fibers. Transcriptome analysis identified 208 DEGs (109 upregulated, 99 downregulated) linked to inflammation, immune activation, and extracellular matrix remodeling. Conclusion iRCT involves ECM remodeling, inflammation, and immune dysregulation, with identified hub genes offering potential therapeutic targets.
Collapse
Affiliation(s)
- Qiuji Wang
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, People’s Republic of China
| | - Lishan Zhong
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
| | - Linbin Hua
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, People’s Republic of China
| | - Shanwen Pang
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, People’s Republic of China
| | - Yuxin Li
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
| | - Zhaolong Zhang
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
| | - Junfei Zhao
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
| | - Huanlei Huang
- Department of Cardiac Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, People’s Republic of China
- Guangdong Cardiovascular Institute, Guangzhou, 510030, People’s Republic of China
- Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangzhou, People’s Republic of China
| |
Collapse
|
42
|
Singh J, Pradhan P, Kataria A, Sinha S, Ehtesham NZ, Monk PN, Hasnain SE. Conservation of Putative Liquid-Liquid Phase Separating Proteins in Multiple Drug-Resistant Mycobacterium tuberculosis: Role in Host-Pathogen Interactions? ACS Infect Dis 2025. [PMID: 40183374 DOI: 10.1021/acsinfecdis.4c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
We observed a high proportion of proteins in pathogenic Mycobacterium species that can potentially undergo liquid-liquid phase separation (LLPS) mediated biomolecular condensate formation, compared to nonpathogenic species. These proteins mainly include the PE-PPE and PE-PGRS families of proteins that have nucleic acid and protein-protein binding functions, typical of LLPS proteins. We also mapped identified LLPS proteins in M. tuberculosis (M.tb) drug-resistant databases PubMLST and TBProfiler, based upon the WHO 2023 catalogue of resistance-associated mutations. High sequence conservation of LLPS-associated proteins in various multiple drug-resistant M.tb isolates points to their potentially important role in virulence and host-pathogen interactions during pathogenic evolution. This analysis provides a perspective on the role of protein phase separation in the evaluation of M.tb pathogenesis and offers avenues for future research aimed at developing innovative strategies to combat M.tb infection.
Collapse
Affiliation(s)
- Jasdeep Singh
- Department of Chemistry and Biochemistry, University of Denver, Denver, Colorado 80210, United States
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi 110016, India
| | - Prashant Pradhan
- Laboratory of Nuclear Organization, Cecil H. and Ida Green Center for Reproductive Biology Sciences, Division of Basic Research, Department of Obstetrics and Gynecology, Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9096, United States
| | - Arti Kataria
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Hamilton, Montana 59840, United States
| | - Sanjeev Sinha
- Department of Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Nasreen Z Ehtesham
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
| | - Peter N Monk
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, U.K
| | - Seyed E Hasnain
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201310, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, New Delhi 110016, India
| |
Collapse
|
43
|
Li K, Zhang Y, Diao Y, Fan S. Paeonol regulates the DDIT4-mTOR signaling pathway in macrophages to promote diabetic wound healing. Int Immunopharmacol 2025; 151:114347. [PMID: 39987633 DOI: 10.1016/j.intimp.2025.114347] [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: 01/22/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Diabetic foot ulcers are a common complication in people with diabetes, and patients with severe disease are at risk of amputation. Current studies have found that one of the reasons for the difficulty in healing diabetic foot ulcers is the Abnormal polarization of the M1/M2 phenotype of macrophages, which leads to a prolonged inflammatory period of the wound. The aim of this study was to investigate whether paeonol can promote the polarization of macrophages towards the M2 type and whether M2 type macrophages can regulate the DDIT4-mTOR signaling pathway and slow down the inflammatory response of diabetic foot ulcers. METHODS C57BL/6 mice were used to establish an animal model of diabetic foot ulcers and the effect of paeonol on wound healing was investigated. The effects of paeonol on wound healing of foot ulcer in diabetic mice were evaluated using histological staining and immunohistochemistry. The molecular mechanism of refractory healing of foot ulcers was speculated through network pharmacology. The effects of Paeonol on phenotypic polarization of macrophages and the mechanism of inhibiting inflammation were studied by q-PCR, ELISA, immunofluorescence and Western. RESULTS Paeonol can effectively promote wound healing in diabetic mice. HE staining showed that paeonol could improve the inflammatory infiltration in the ulcer wound of diabetic mice; Masson trichromatic staining showed that paeonol could increase the increase of muscle fibers and collagen in the wound tissue of diabetic mice; immunofluorescence results showed that paeonol could increase the angiogenesis in the wound tissue of diabetic mice. Network pharmacological analysis showed that the molecular mechanism of paeonol in treating diabetic wound healing may be through DDIT4-mTOR signaling pathway. q-PCR, ELISA, immunofluorescence and Western blot showed that paeonol could reduce the expression of the signature protein CD86 and inflammatory factors in M1 macrophages, and promote the phenotypic polarization of M2 macrophages, which is the mechanism of inhibiting inflammation by activating DDIT4-mTOR signaling pathway. CONCLUSION Paeonol can promote the polarization of macrophages towards M2 type, reduce inflammatory response and accelerate wound surface healing through DDIT4-mTOR signaling pathway, providing a new therapeutic strategy for the treatment of diabetic foot ulcers.
Collapse
Affiliation(s)
- Kun Li
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China.
| | - Yingying Zhang
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian 116044, China; Dalian anti-infective traditional Chinese Medicine Development Engineering Technology Research Center, China
| | - Shuyuan Fan
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China.
| |
Collapse
|
44
|
Huo Z, Fan C, Li K, Xu C, Niu Y, Wang F. Identification and validation of hub m7G-related genes and infiltrating immune cells in osteoarthritis based on integrated computational and bioinformatics analysis. BMC Musculoskelet Disord 2025; 26:333. [PMID: 40186163 PMCID: PMC11971809 DOI: 10.1186/s12891-025-08539-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a joint disease closely associated with synovial tissue inflammation, with the severity of synovitis impacting disease progression. m7G RNA methylation is critical in RNA processing, metabolism, and function, but its role in OA synovial tissue is not well understood. This study explores the relationship between m7G methylation and immune infiltration in OA. METHODS Data were obtained from the GEO database. Hub genes related to m7G were identified using differential expression and LASSO-Cox regression analysis, and a diagnostic model was developed. Functional enrichment, drug target prediction, and target gene-related miRNA prediction were performed for these genes. Immune cell infiltration was analyzed using the CIBERSORT algorithm, and unsupervised clustering analysis was conducted to examine immune infiltration patterns. RT-qPCR was used to validate hub gene expression. RESULTS Seven m7G hub genes (SNUPN, RNMT, NUDT1, LSM1, LARP1, CYFIP2, and CYFIP1) were identified and used to develop a nomogram for OA risk prediction. Functional enrichment indicated involvement in mRNA metabolism and RNA transport. Differences in macrophage and T-cell infiltration were observed between OA and normal groups. Two distinct m7G immune infiltration patterns were identified, with significant microenvironment differences between clusters. RT-qPCR confirmed differential hub gene expression. CONCLUSION A diagnostic model based on seven m7G hub genes was developed, highlighting these genes as potential biomarkers and significant players in OA pathogenesis.
Collapse
Affiliation(s)
- Zhenhui Huo
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Chongyi Fan
- Department of Orthopedics, Aerospace Central Hospital, Beijing, 100049, China
| | - Kehan Li
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Chenyue Xu
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| | - Yingzhen Niu
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China.
| | - Fei Wang
- Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China.
| |
Collapse
|
45
|
Nagarajan P, Winkler TW, Bentley AR, Miller CL, Kraja AT, Schwander K, Lee S, Wang W, Brown MR, Morrison JL, Giri A, O'Connell JR, Bartz TM, de Las Fuentes L, Gudmundsdottir V, Guo X, Harris SE, Huang Z, Kals M, Kho M, Lefevre C, Luan J, Lyytikäinen LP, Mangino M, Milaneschi Y, Palmer ND, Rao V, Rauramaa R, Shen B, Stadler S, Sun Q, Tang J, Thériault S, van der Graaf A, van der Most PJ, Wang Y, Weiss S, Westerman KE, Yang Q, Yasuharu T, Zhao W, Zhu W, Altschul D, Ansari MAY, Anugu P, Argoty-Pantoja AD, Arzt M, Aschard H, Attia JR, Bazzanno L, Breyer MA, Brody JA, Cade BE, Chen HH, Chen YDI, Chen Z, de Vries PS, Dimitrov LM, Do A, Du J, Dupont CT, Edwards TL, Evans MK, Faquih T, Felix SB, Fisher-Hoch SP, Floyd JS, Graff M, Gu C, Gu D, Hairston KG, Hanley AJ, Heid IM, Heikkinen S, Highland HM, Hood MM, Kähönen M, Karvonen-Gutierrez CA, Kawaguchi T, Kazuya S, Kelly TN, Komulainen P, Levy D, Lin HJ, Liu PY, Marques-Vidal P, McCormick JB, Mei H, Meigs JB, Menni C, Nam K, Nolte IM, Pacheco NL, Petty LE, Polikowsky HG, Province MA, Psaty BM, Raffield LM, Raitakari OT, Rich SS, Riha RL, Risch L, Risch M, Ruiz-Narvaez EA, Scott RJ, Sitlani CM, Smith JA, Sofer T, Teder-Laving M, Völker U, Vollenweider P, Wang G, Willems van Dijk K, Wilson OD, Xia R, Yao J, Young KL, Zhang R, Zhu X, Below JE, Böger CA, Conen D, Cox SR, Dörr M, Feitosa MF, Fox ER, Franceschini N, Gharib SA, Gudnason V, Harlow SD, He J, Holliday EG, Kutalik Z, Lakka TA, Lawlor DA, Lee S, Lehtimäki T, Li C, Liu CT, Mägi R, Matsuda F, Morrison AC, Penninx BW, Peyser PA, Rotter JI, Snieder H, Spector TD, Wagenknecht LE, Wareham NJ, Zonderman AB, North KE, Fornage M, Hung AM, Manning AK, Gauderman J, Chen H, Munroe PB, Rao DC, van Heemst D, Redline S, Noordam R, Wang H. A large-scale genome-wide study of gene-sleep duration interactions for blood pressure in 811,405 individuals from diverse populations. Mol Psychiatry 2025:10.1038/s41380-025-02954-w. [PMID: 40181193 DOI: 10.1038/s41380-025-02954-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025]
Abstract
Although both short and long sleep duration are associated with elevated hypertension risk, our understanding of their interplay with biological pathways governing blood pressure remains limited. To address this, we carried out genome-wide cross-population gene-by-short-sleep and long-sleep duration interaction analyses for three blood pressure traits (systolic, diastolic, and pulse pressure) in 811,405 individuals from diverse population groups. We discovered 22 novel gene-sleep duration interaction loci for blood pressure, mapped to 23 genes. Investigating these genes' functional implications shed light on neurological, thyroidal, bone metabolism, and hematopoietic pathways that necessitate future investigation for blood pressure management that caters to sleep health lifestyle. Non-overlap between short sleep (12) and long sleep (10) interactions underscores the plausible nature of distinct influences of both sleep duration extremes in cardiovascular health. Several of our loci are specific towards a particular population background or sex, emphasizing the importance of addressing heterogeneity entangled in gene-environment interactions, when considering precision medicine design approaches for blood pressure management.
Collapse
Affiliation(s)
- Pavithra Nagarajan
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Clint L Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Aldi T Kraja
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Karen Schwander
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Songmi Lee
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Wenyi Wang
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - John L Morrison
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Ayush Giri
- Division of Quantitative and Clinical Sciences, Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Lisa de Las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Valborg Gudmundsdottir
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, Department of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sarah E Harris
- Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Mart Kals
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Minjung Kho
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Christophe Lefevre
- Department of Data Sciences, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
- National Heart & Lung Institute, Cardiovascular Genomics and Precision Medicine, Imperial College London, London, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC/Vrije universiteit, Amsterdam, Netherlands
- GGZ inGeest, Amsterdam, Netherlands
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Varun Rao
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Botong Shen
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Stefan Stadler
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jingxian Tang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC, Canada
| | - Adriaan van der Graaf
- Statistical Genetics Group, Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Yujie Wang
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stefan Weiss
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Kenneth E Westerman
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Qian Yang
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tabara Yasuharu
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Wei Zhao
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Wanying Zhu
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Drew Altschul
- Department of Psychology, The University of Edinburgh, Edinburgh, UK
- School of Psychology, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Md Abu Yusuf Ansari
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pramod Anugu
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Anna D Argoty-Pantoja
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Hugues Aschard
- Department of Computational Biology, F-75015 Paris, France Institut Pasteur, Université Paris Cité, Paris, France
- Department of Epidemiology, Harvard TH School of Public Health, Boston, MA, USA
| | - John R Attia
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Lydia Bazzanno
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Max A Breyer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hung-Hsin Chen
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Zekai Chen
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Latchezar M Dimitrov
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anh Do
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jiawen Du
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles T Dupont
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Todd L Edwards
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tariq Faquih
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Stephan B Felix
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Susan P Fisher-Hoch
- School of Public Health, The University of Texas Health Science Center at Houston (UTHealth), Brownsville, TX, USA
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Charles Gu
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Dongfeng Gu
- Shenzhen Key Laboratory of Cardiovascular Health and Precision Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Kristen G Hairston
- Department of Endocrinology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Anthony J Hanley
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Heather M Highland
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michelle M Hood
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | | | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Setoh Kazuya
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | | | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Henry J Lin
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peter Y Liu
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Joseph B McCormick
- School of Public Health, The University of Texas Health Science Center at Houston (UTHealth), Brownsville, TX, USA
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Kisung Nam
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Natasha L Pacheco
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Lauren E Petty
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hannah G Polikowsky
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, and Department of Clinical Physiology and Nuclear Medicine, University of Turku, and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Renata L Riha
- Department of Sleep Medicine, The University of Edinburgh, Edinburgh, UK
| | - Lorenz Risch
- Faculty of Medical Sciences, Institute for Laboratory Medicine, Private University in the Principality of Liechtenstein, Vaduz, Liechtenstein
- Center of Laboratory Medicine, Institute of Clinical Chemistry, University of Bern and Inselspital, Bern, Switzerland
| | - Martin Risch
- Central Laboratory, Cantonal Hospital Graubünden, Chur, Switzerland
- Medical Laboratory, Dr. Risch Anstalt, Vaduz, Liechtenstein
| | | | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Guanchao Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden, Netherlands
| | - Otis D Wilson
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rui Xia
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kristin L Young
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ruiyuan Zhang
- Department of Epidemiology, O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jennifer E Below
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Nephrology and Rheumatology, Kliniken Südostbayern, Traunstein, Germany
- KfH Kidney Centre Traunstein, Traunstein, Germany
| | - David Conen
- Population Health Research Institute, Medicine, McMaster University, Hamilton, ON, Canada
| | - Simon R Cox
- Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Cardiology, Pneumology, Infectious Diseases, Intensive Care Medicine, Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ervin R Fox
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nora Franceschini
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sina A Gharib
- Pulmonary, Critical Care and Sleep Medicine, Medicine, University of Washington, Seattle, WA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, Department of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sioban D Harlow
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jiang He
- Department of Epidemiology, O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth G Holliday
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW, Australia
| | - Zoltan Kutalik
- Statistical Genetics Group, Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Center for Primary Care and Public Health, University of Lausanne, Lausanne, Switzerland
| | - Timo A Lakka
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Seunggeun Lee
- Graduate School of Data Science, Seoul National University, Seoul, South Korea
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere, Finland
| | - Changwei Li
- Department of Epidemiology, O'Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Brenda Wjh Penninx
- Department of Psychiatry, Amsterdam UMC/Vrije universiteit, Amsterdam, Netherlands
- GGZ inGeest, Amsterdam, Netherlands
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Kari E North
- Department of Epidemiology, UNC Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Adriana M Hung
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626), Department of Veterans Affairs, Nashville, TN, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James Gauderman
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Patricia B Munroe
- Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London, UK
| | - Dabeeru C Rao
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, Netherlands
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Ju F, Wang J, Xu K, Xu Q, Liu X, Tian T, Du Z, Wang J, Liao Z, Wang B, Zhang H. Genome-wide insights into the nomenclature, evolution and expression of tobacco TIFY/JAZ genes. PLANTA 2025; 261:103. [PMID: 40183817 DOI: 10.1007/s00425-025-04676-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
MAIN CONCLUSION A systematic nomenclature for tobacco TIFY/JAZ proteins was established via genome-wide analysis, and the gene transcription patterns and potential functions of these proteins were analyzed as well. Intensive studies focused on the plant-specific JAZ regulators of jasmonate (JA) signaling in tobacco due to their critical roles in regulating JA-mediated development, secondary metabolism, and stress responses. JAZs comprise a subfamily of the TIFY proteins, yet the reported TIFY/JAZ regulators of tobacco spp. are tangled in naming confusion, which resulted in nomenclature chaos. Here, we identified 32 TIFY/JAZ proteins via genome-wide analysis of tobacco cultivar TN90 and obtained their homologues in Nicotiana sylvestris and Nicotiana tomentosiformis. By bioinformatic analysis, these TIFY/JAZ regulators were classified into 4 subfamilies (i.e., 21 JAZs, 5 ZIM & ZMLs, 2 TIFY8s, and 4 PPDs) based on their phylogenetic relationship to establish a systematic nomenclature, which indicated gene loss or genomic rearrangement during the formation of common tobacco. Analysis of JA-induced expression revealed that these TIFY/JAZ genes displayed distinct expression patterns in the leaves and roots upon JA treatment. Further microarray and metabolomics assays observed that 5 TIFY/JAZ genes were differentially expressed in the plants with dysfunction of COI1, the receptor protein of JA hormone and that the abundance of a series of primary and secondary metabolites was altered as well. A predicted protein interaction network of tobacco TIFY/JAZ proteins was also constructed, and it indicated that 120 proteins may interact with these regulators. Findings of this work provide valuable information about TIFY/JAZ proteins in regulating JA responses and metabolic processes in tobacco and may contribute greatly to future studies on tobacco TIFY/JAZ proteins.
Collapse
Affiliation(s)
- Fuzhu Ju
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Jiahao Wang
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Ke Xu
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Qing Xu
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Xiaofeng Liu
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Tian Tian
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Zaifeng Du
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Jialin Wang
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China
| | - Zhihua Liao
- SWU-TAAHC Medicinal Plant Joint R&D Centre, School of Life Sciences, Southwest University, Chongqing, 400716, China
| | - Bingwu Wang
- Tobacco Breeding and Biotechnology Research Center, Yunnan Academy of Tobacco Agricultural Sciences, Kunming, 650021, China.
| | - Hongbo Zhang
- Key Laboratory of Synthetic Biology of Ministry of Agriculture and Rural Affairs, Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao, 266101, China.
| |
Collapse
|
47
|
Gomes FP, Durbin KR, Schauer K, Nwachukwu JC, R Kobylski R, Njeri JW, Seath CP, Saviola AJ, McClatchy DB, Diedrich JK, Garrett PT, Papa AB, Ciolacu I, Kelleher NL, Nettles KW, Yates JR. Native top-down proteomics enables discovery in endocrine-resistant breast cancer. Nat Chem Biol 2025:10.1038/s41589-025-01866-8. [PMID: 40186031 DOI: 10.1038/s41589-025-01866-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/24/2025] [Indexed: 04/07/2025]
Abstract
Oligomerization of proteoforms produces functional protein complexes. Characterization of these assemblies within cells is critical to understanding the molecular mechanisms involved in disease and to designing effective drugs. Here we present a native top-down proteomics (nTDP) strategy to identify protein assemblies (≤70 kDa) in breast cancer cells and in cells that overexpress epidermal growth factor receptor (EGFR), which serves as a resistance model of estrogen receptor-alpha (ER)-targeted therapies. This nTDP approach identified ~104 complexoforms from 17 protein complexes, which revealed several molecular features of the breast cancer proteome, including EGFR-induced dissociation of nuclear transport factor 2 (NUTF2) assemblies that modulate ER activity. We found that the K4 and K55 post-translational modification sites discovered with nTDP differentially impact the effects of NUTF2 on the inhibition of the ER signaling pathway. The characterization of endogenous proteoform-proteoform/ligand interactions revealed the molecular diversity of complexoforms and their role in breast cancer growth.
Collapse
Affiliation(s)
| | | | | | - Jerome C Nwachukwu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | - Robin R Kobylski
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Jacqline W Njeri
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Ciaran P Seath
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | | | | | | | | | - Alexandra B Papa
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- Florida Atlantic University, Jupiter, FL, USA
| | - Ianis Ciolacu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- Florida Atlantic University, Jupiter, FL, USA
| | - Neil L Kelleher
- Proteinaceous, Evanston, IL, USA
- Northwestern University, National Resource for Translational and Developmental Proteomics, Evanston, IL, USA
| | - Kendall W Nettles
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA.
| | | |
Collapse
|
48
|
Loomis S, Silva DG, Savopoulos R, Cilia J, Li J, Davis MD, Virley D, Foley A, Loro E, McCreary AC. BEHAVIORAL AND TRANSCRIPTOMIC EFFECTS OF A NOVEL CANNABINOID ON A RAT VALPROIC ACID MODEL OF AUTISM. Neuropharmacology 2025:110450. [PMID: 40187640 DOI: 10.1016/j.neuropharm.2025.110450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by impaired social communication, restricted interests, repetitive behavior and irritability. Exposure to valproic acid (VPA) during pregnancy has been shown to increase the risk of autism in children and has led to the development of the in-utero VPA rat model that elicits neurodevelopmental autistic-like features. Offspring exhibit behavioral and neurobiological alterations modelling ASD symptoms. We performed a behavioral and molecular assessment in a rat in-utero VPA model treated with a novel botanical cannabinoid, JZP541. Male offspring from dams treated with VPA were tested acutely and sub-chronically with JZP541 (10, 30, or 100 mg/kg, intraperitoneally). A behavioral testing battery was performed, and brain frontal cortex and hippocampus used for RNA sequencing. In utero exposure to VPA resulted in progeny showing behavioral phenotypes characteristic of ASD. JZP541 attenuated these deficits in social, stereotypic, hyperactivity and irritability behavior in a dose-dependent fashion. VPA exposure was associated with a substantial transcriptional dysregulation impacting multiple key biological processes in a tissue-dependent manner. The expression profiles were integrated with publicly available datasets of autism-associated genes to support the validity of the model used and to focus on the effects of treatment on known autism-relevant transcriptional targets. This approach indicated a strong and dose-dependent reduction of the autism-associated gene expression signature in brain samples from animals dosed with JZP541. Our findings demonstrate JZP541 was able to ameliorate ASD associated behavioral deficits, and this was supported by improvements in putative transcriptional biomarkers of ASD.
Collapse
Affiliation(s)
- Sally Loomis
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK.
| | - Diogo G Silva
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | | - Jackie Cilia
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | - Jennifer Li
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | - Mat D Davis
- Jazz Pharmaceuticals Inc., Palo Alto, CA, USA
| | - David Virley
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | | - Emanuele Loro
- Jazz Pharmaceuticals Research UK Ltd., Cambridge, UK
| | | |
Collapse
|
49
|
Kianpoor S, Ehsani A, Torshizi RV, Masoudi AA, Bakhtiarizadeh MR. Unlocking the genetic code: a comprehensive Genome-Wide association study and gene set enrichment analysis of cell-mediated immunity in chickens. BMC Genomics 2025; 26:337. [PMID: 40181279 PMCID: PMC11970016 DOI: 10.1186/s12864-025-11538-5] [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: 01/18/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The poultry immune system is essential for protecting against infectious diseases and maintaining health and productivity. Cell-mediated immune responses (CMIs) protect organisms against intracellular pathogens. This study aimed to enrich the findings of genome-wide association studies (GWAS) by including several systematic gene set enrichment analyses (GSEA) related to cell-mediated immune responses in chickens. METHODS To investigate the function of the cellular immune system, phenotypic data were collected based on the differences in skin thickness before and after impregnation with dinitrochlorobenzene (DNCB) solution. Additionally, 312 hybrid birds of the F2 generation of Arian broiler chickens and Urmia native chickens were genotyped using the Illumina 60k SNP BeadChip. A general linear model (GLM) with an FDR < 5% was used for the association analysis. Functional enrichment analysis of the identified candidate genes was performed using the Enrichr database. A protein‒protein interaction (PPI) network was constructed using the STRING database. In addition, colocalization analysis was applied to identify QTLs related to the immune system. RESULTS GWAS revealed 147 SNPs associated with the CMI trait, which were related to 1363 genes. These genes were significantly enriched in eight KEGG pathways, 22 Reactome pathways, and 18 biological processes. PPI network analysis led to the identification of 26 hub genes. The three hub genes PSMA3, PSMC2 and PSMB4 were enriched in almost all pathways related to cellular immunity, including the proteasome, interleukin-1 signaling, and programmed cell death pathways, which makes them important candidates involved in CMI. In addition, the MAP3K8, NLRC5, UBB, CASP6, DAPK2, TNFRSF6B, TNFSF15, and PIK3CD genes were identified as key genes in several functional pathways. A total of 10 SNPs were found in interferon-gamma QTLs, and two SNPs were found in the cell-mediated immune response QTL region, leading to the identification of 12 cellular immune response-related genes that were reported as important candidates in previous studies. CONCLUSION The post-GWAS analysis in this study led to the identification of key genes that regulate the biological processes of cellular immunity in chickens. Therefore, selecting birds that excel in expressing such genes can improve immunity in chickens.
Collapse
Affiliation(s)
- Somayeh Kianpoor
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Alireza Ehsani
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran.
| | - Rasoul Vaez Torshizi
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Ali Akbar Masoudi
- Department of Animal Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
50
|
Kitani A, Matsui Y. Integrative network analysis reveals novel moderators of Aβ-Tau interaction in Alzheimer's disease. Alzheimers Res Ther 2025; 17:70. [PMID: 40176187 PMCID: PMC11967117 DOI: 10.1186/s13195-025-01705-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Although interactions between amyloid-beta and tau proteins have been implicated in Alzheimer's disease (AD), the precise mechanisms by which these interactions contribute to disease progression are not yet fully understood. Moreover, despite the growing application of deep learning in various biomedical fields, its application in integrating networks to analyze disease mechanisms in AD research remains limited. In this study, we employed BIONIC, a deep learning-based network integration method, to integrate proteomics and protein-protein interaction data, with an aim to uncover factors that moderate the effects of the Aβ-tau interaction on mild cognitive impairment (MCI) and early-stage AD. METHODS Proteomic data from the ROSMAP cohort were integrated with protein-protein interaction (PPI) data using a Deep Learning-based model. Linear regression analysis was applied to histopathological and gene expression data, and mutual information was used to detect moderating factors. Statistical significance was determined using the Benjamini-Hochberg correction (p < 0.05). RESULTS Our results suggested that astrocytes and GPNMB + microglia moderate the Aβ-tau interaction. Based on linear regression with histopathological and gene expression data, GFAP and IBA1 levels and GPNMB gene expression positively contributed to the interaction of tau with Aβ in non-dementia cases, replicating the results of the network analysis. CONCLUSIONS These findings suggest that GPNMB + microglia moderate the Aβ-tau interaction in early AD and therefore are a novel therapeutic target. To facilitate further research, we have made the integrated network available as a visualization tool for the scientific community (URL: https://igcore.cloud/GerOmics/AlzPPMap ).
Collapse
Affiliation(s)
- Akihiro Kitani
- Department of Integrated Health Science, Biomedical and Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Matsui
- Department of Integrated Health Science, Biomedical and Health Informatics Unit, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Institute for Glyco-Core Research (Igcore), Nagoya University, Nagoya, Aichi, 461-8673, Japan.
| |
Collapse
|