1
|
Satbhai KM, Marques ES, Ranjan R, Timme-Laragy AR. Single-cell RNA sequencing reveals tissue-specific transcriptomic changes induced by perfluorooctanesulfonic acid (PFOS) in larval zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137515. [PMID: 39947082 PMCID: PMC12038816 DOI: 10.1016/j.jhazmat.2025.137515] [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: 08/22/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 04/16/2025]
Abstract
Perfluorooctanesulfonic acid (PFOS) elicits adverse effects on numerous organs and developmental processes but the mechanisms underlying these effects are not well understood. Here, we use single-cell RNA-sequencing to assess tissue-specific transcriptomic changes in zebrafish (Danio rerio) larvae exposed to 16 µM PFOS or dimethylsulfoxide (0.01 %) from 3-72 h post fertilization (hpf). Data analysis was multi-pronged and included pseudo-bulk, untargeted clustering, informed pathway queries, and a cluster curated for hepatocyte biomarkers (fabp10a, and apoa2). Overall, 8.63 % (2390/27698) genes were significantly differentially expressed. Results from untargeted analysis revealed 22 distinct clusters that were manually annotated to specific tissues using a weight-of-evidence approach. The clusters with the highest number of significant differentially expressed genes (DEGs) were digestive organs, muscle, and otolith. Additionally, we assessed the distribution of pathway-specific genes known to be involved in PFOS toxicity: the PPAR pathway, β-oxidation of fatty acids, the Nfe2l2 pathway, and epigenetic modifications by DNA methylation, across clusters and identified the blood-related tissue to be the most sensitive. The curated hepatocyte cluster showed 220 significant DEGs and was enriched for the Notch signaling pathway. These findings provide insights into both established and novel sensitive target tissues and molecular mechanisms of developmental toxicity of PFOS.
Collapse
Affiliation(s)
- Kruuttika M Satbhai
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Emily S Marques
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ravi Ranjan
- Genomics Resource Laboratory, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
2
|
Pons C. Qarles: a web server for the quick characterization of large sets of genes. NAR Genom Bioinform 2025; 7:lqaf030. [PMID: 40160219 PMCID: PMC11954521 DOI: 10.1093/nargab/lqaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
The characterization of gene sets is a recurring task in computational biology. Identifying specific properties of a hit set compared to a reference set can reveal biological roles and mechanisms, and can lead to the prediction of new hits. However, collecting the features to evaluate can be time consuming, and implementing an informative but compact graphical representation of the multiple comparisons can be challenging, particularly for bench scientists. Here, I present Qarles (quick characterization of large sets of genes), a web server that annotates Saccharomyces cerevisiae gene sets by querying a database of 31 features widely used by the yeast community and that identifies their specific properties, providing publication-ready figures and reliable statistics. Qarles has a deliberately simple user interface with all the functionality in a single web page and a fast response time to facilitate adoption by the scientific community. Qarles provides a rich and compact graphical output, including up to five gene set comparisons across 31 features in a single dotplot, and interactive boxplots to enable the identification of outliers. Qarles can also predict new hit genes by using a random forest trained on the selected features. The web server is freely available at https://qarles.org.
Collapse
Affiliation(s)
- Carles Pons
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
3
|
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
|
4
|
Chokkalla AK, Arruri V, Mehta SL, Vemuganti R. Loss of Epitranscriptomic Modification N 6-Methyladenosine (m 6A) Reader YTHDF1 Exacerbates Ischemic Brain Injury in a Sexually Dimorphic Manner. Transl Stroke Res 2025; 16:831-847. [PMID: 38869772 PMCID: PMC12066170 DOI: 10.1007/s12975-024-01267-4] [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: 03/28/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
N6-Methyladenosine (m6A) is a neuronal-enriched, reversible post-transcriptional modification that regulates RNA metabolism. The m6A-modified RNAs recruit various m6A-binding proteins that act as readers. Differential m6A methylation patterns are implicated in ischemic brain damage, yet the precise role of m6A readers in propagating post-stroke m6A signaling remains unclear. We presently evaluated the functional significance of the brain-enriched m6A reader YTHDF1, in post-stroke pathophysiology. Focal cerebral ischemia significantly increased YTHDF1 mRNA and protein expression in adult mice of both sexes. YTHDF1-/- male, but not female, mice subjected to transient middle cerebral artery occlusion (MCAO) showed worsened motor function recovery and increased infarction compared to sex-matched YTHDF1+/+ mice. YTHDF1-/- male, but not female, mice subjected to transient MCAO also showed significantly perturbed expression of genes related to inflammation, and increased infiltration of peripheral immune cells into the peri-infarct cortex, compared with sex-matched YTHDF1+/+ mice. Thus, this study demonstrates a sexual dimorphism of YTHDF1 in regulating post-ischemic inflammation and pathophysiology. Hence, post-stroke epitranscriptomic regulation might be sex-dependent.
Collapse
Affiliation(s)
- Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
5
|
Škorjanc A, Smrkolj V, Umek N. GOReverseLookup: A gene ontology reverse lookup tool. Comput Biol Med 2025; 191:110185. [PMID: 40239235 DOI: 10.1016/j.compbiomed.2025.110185] [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: 06/15/2024] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND AND OBJECTIVE The Gene Ontology (GO) project has been pivotal in providing a structured framework for characterizing genes and annotating them to specific biological concepts. While traditional gene annotation primarily focuses on mapping genes to GO terms, descriptors of biological concepts, there is a growing need for tools facilitating reverse querying. This paper introduces GOReverseLookup, a novel tool designed to identify over- or underrepresented genes in researcher-defined states of interest (phenotypes), described by sets of GO terms. GOReverseLookup supplements the existing power of Gene Ontology by the possibility of orthologous gene querying across several databases, such as Ensembl and UniProtKB. This combination allows for a more nuanced identification of significant genes across a range of cross-species research contexts. METHODS GOReverseLookup queries genes associated with input GO terms. Bundles of GO terms encapsulate user-defined states of interest, e.g., angiogenesis. In the second stage of the analysis, all GO terms associated with each gene are fetched, and finally, the statistical relevance of the genes being involved in one (or all) of the defined states of interests is computed. RESULTS The two presented use cases illustrate its utility in discovering genes related to rheumatoid arthritis and genes linked with chronic inflammation and tumorigenesis. In both cases, GOReverseLookup discovered a substantial number of genes significantly associated with the aforementioned states of interest. CONCLUSIONS GOReverseLookup proves to be a valuable resource for unraveling the genetic basis of phenotypes, with diverse practical potentials in functional genomics, systems biology, and drug discovery. We anticipate that GOReverseLookup will significantly aid in identifying potential gene targets during the initial research phases.
Collapse
Affiliation(s)
- Aljoša Škorjanc
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia
| | - Vladimir Smrkolj
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia; National Institute of Chemistry, Hajdrihova ulica 19, Ljubljana, Slovenia
| | - Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia.
| |
Collapse
|
6
|
Guzmán-Dinamarca B, Conejeros R, Rivas-Astroza M. Dynamic metabolic regulation of histone modifications during the yeast metabolic cycle. PLoS One 2025; 20:e0323242. [PMID: 40392806 PMCID: PMC12091797 DOI: 10.1371/journal.pone.0323242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/04/2025] [Indexed: 05/22/2025] Open
Abstract
Eukaryotes achieve a wide range of stable phenotypes by virtue of epigenetic modifications. However, what drives epigenetic diversification in the first place remains an open question. Here, we investigated the dynamic interplay between the production fluxes of epigenetic cosubstrates and histone post-translation modifications (PTMs) in Saccharomyces cerevisiae's Yeast Metabolic Cycle (YMC). We developed a novel approach integrating flux analysis with transcriptomic data to investigate the production fluxes of acetyl-CoA and SAM and their influence on histone marks H3K9Ac and H3K4me3. Our results show that acetyl-CoA and SAM flux dynamics are asynchronous during the YMC, suggesting distinct regulatory roles. Gene ontology analysis revealed that genes whose enrichment of H3K9Ac correlates with acetyl-CoA dynamics are associated with metabolic functions, while genes whose enrichment of H3K4me3 correlates with SAM dynamics are associated with translation processes. Finally, we found evidence that chromatin accessibility on genes promoter regions was a precondition for the metabolic fluxes influencing the enrichment of H3K4me3 and H3K9Ac. These findings support the concept that metabolism provides timely cosubstrates essential for histone PTMs.
Collapse
Affiliation(s)
| | - Raúl Conejeros
- Pontificia Universidad Católica de Valparaíso, Escuela de Ingeniería Bioquímica, Valparaíso, Chile
| | - Marcelo Rivas-Astroza
- Universidad Tecnológica Metropolitana, Departamento de Biotecnología, Santiago, Chile
| |
Collapse
|
7
|
Zhang Q, Yang L, Li C, Zhang Y, Li R, Jia F, Wang L, Ma X, Yao K, Tian H, Zhuo C. Exploring the potential antidepressant mechanisms of ibuprofen and celecoxib based on network pharmacology and molecular docking. J Affect Disord 2025; 377:136-147. [PMID: 39986574 DOI: 10.1016/j.jad.2025.02.053] [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: 08/18/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Evidence has shown that ibuprofen and celecoxib are effective in improving depressive symptoms, but their mechanisms of action are unclear. In this study, we aimed to determine the relationship between these two drugs and depressive disorder (DD) and elucidate potential mechanisms of action. METHODS Relevant targets for ibuprofen, celecoxib, and DD were obtained and screened from multiple online drug and disease public databases. A protein-protein interaction network was obtained. The Centiscape and CytoHubba plug-ins were applied to screen for core targets. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed. Molecular docking was performed to predict the binding of ibuprofen and celecoxib to core targets. Examined the differences in core target protein expression between DD patients (DDs, n = 18) and healthy controls (HCs, n = 16) as a further experimental validation of the network pharmacology results. RESULTS In total, 220 potential targets for ibuprofen and 316 potential targets for celecoxib were identified and associated with DD. The antidepressant effects of both drugs involve many key targets in pathways such as "pathways in cancer" and "neuroactive ligand-receptor interaction," including ALB, BCL2, MAPK3, SRC, STAT3, EGFR, and PPARG. The binding affinity of ALB with ibuprofen is the strongest, and it is connected only by hydrophobic interactions. Celecoxib exhibits higher affinity at multiple targets such as SRC, EGFR, and PPARG, with stronger and more specific intermolecular interactions, including salt bridges and halogen bonds. Clinical trials have found that serum ALB expression in DDs is significantly lower than that in HCs (t = 6.653, p < 0.001), further confirming the potential role of ibuprofen in DD. CONCLUSIONS Ibuprofen and celecoxib primarily exert their antidepressant effects through targets and pathways related to inflammation, neural signaling, and cancer, with celecoxib showing a stronger potential antidepressant effect. The expression difference of the core target ALB between depression and healthy individuals further supports the potential effect of the drug on DD. Our findings propose new treatment strategies, support the link between inflammation and depression, and encourage reassessing existing medications for depression.
Collapse
Affiliation(s)
- Qiuyu Zhang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Lei Yang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Chao Li
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ying Zhang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Ranli Li
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Feng Jia
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Lina Wang
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Xiaoyan Ma
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Kaifang Yao
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China
| | - Hongjun Tian
- Animal Imaging Center (AIC) of Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin 300140, China
| | - Chuanjun Zhuo
- Computational Biology and Animal Imaging Center (CBAC), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Anding Hospital, Tianjin Medical University Affiliated Tianjin Mental Health Center, Tianjin 300222, China; Laboratory of Psychiatric-Neuroimaging-Genetic and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin 300222, China.
| |
Collapse
|
8
|
Pekayvaz K, Kilani B, Joppich M, Eivers L, Brambs S, Knottenberg V, Akgöl S, Yue K, Li L, Martinez-Navarro A, Kaiser R, Meißner N, Schulz H, Belz L, Akhalkatsi A, Stockhausen S, Mueller TT, Millonig S, Hartelt L, Gold C, Janjic A, Polewka V, Wendler F, Droste Zu Senden A, Titova A, Leunig A, Voelkl M, Engelmann B, Hernandez Petzsche MR, Boeckh-Behrens T, Liebig T, Winning S, Fandrey J, Dichgans M, Enard W, Zimmer R, Tiedt S, Massberg S, Nicolai L, Stark K. Immunothrombolytic monocyte-neutrophil axes dominate the single-cell landscape of human thrombosis and correlate with thrombus resolution. Immunity 2025; 58:1343-1358.e13. [PMID: 40280129 DOI: 10.1016/j.immuni.2025.03.020] [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: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Thrombotic diseases remain the major cause of death and disability worldwide, and the contribution of inflammation is increasingly recognized. Thromboinflammation has been identified as a key pathomechanism, but an unsupervised map of immune-cell states, trajectories, and intercommunication at a single-cell level has been lacking. Here, we reveal innate leukocyte substates with prominent thrombolytic properties by employing single-cell omics measures on human stroke thrombi. Using in vivo and in vitro thrombosis models, we propose a pro-resolving monocyte-neutrophil axis, combining two properties: (1) NR4A1hi non-classical monocytes acquire a thrombolytic and neutrophil-chemoattractive phenotype, and (2) blood neutrophils are thereby continuously recruited to established thrombi through CXCL8-CXCR1 and CXCR2 and adopt a hypoxia-induced thrombus-resolving urokinase receptor (PLAUR)+ phenotype. This immunothrombolytic axis results in thrombus resolution. Together, with this immune landscape of thrombosis, we provide a valuable resource and introduce the concept of "immunothrombolysis" with broad mechanistic and translational implications at the crossroad of inflammation and thrombosis.
Collapse
Affiliation(s)
- Kami Pekayvaz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Badr Kilani
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sezer Akgöl
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Rainer Kaiser
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nina Meißner
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Larissa Belz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sven Stockhausen
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Simon Millonig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lea Hartelt
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Wendler
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Anna Titova
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Voelkl
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Moritz R Hernandez Petzsche
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boeckh-Behrens
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Liebig
- Institute for Diagnostic and Interventional Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Sandra Winning
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Joachim Fandrey
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Martin Dichgans
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
9
|
Perrella G, Vellutini E, Beveridge A, Hamilton G, Herzyk P, Kaiserli E. TANDEM ZINC-FINGER/PLUS3 integrates light signaling and flowering regulatory pathways at the chromatin level. THE NEW PHYTOLOGIST 2025. [PMID: 40356194 DOI: 10.1111/nph.70213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025]
Abstract
Environmental and endogenous stimuli determine plant developmental transitions including flowering through multiple signaling cascades. Although the key activators and repressors of flowering initiation are defined, the components and mechanisms integrating light signaling and flowering pathways are not fully established. This study investigates the role of TANDEM ZINC-FINGER/PLUS3 (TZP), a light-integrating transcriptional regulator, to elucidate how light cues influence the epigenetic regulation of flowering in Arabidopsis thaliana. To dissect the molecular function of TZP, this study employed a combination of genetics, RNA sequencing, chromatin immunoprecipitation sequencing and phenotypic assays. These approaches were used to determine TZP's genomic binding sites, its downstream gene targets and its influence on flowering time and chromatin modifications. TANDEM ZINC-FINGER/PLUS3 was found to directly associate with the promoter regions of chromatin-modifying genes, including FLOWERING LOCUS D (a histone H3K4 demethylase) and histone deacetylase 6 (a histone deacetylase). This regulation promotes a chromatin environment that represses FLOWERING LOCUS C (FLC) transcription, thereby accelerating flowering. TANDEM ZINC-FINGER/PLUS3 thus functions upstream of multiple pathways integrating photoperiodic and autonomous floral cues. TANDEM ZINC-FINGER/PLUS3 mediates crosstalk between light signaling and flowering pathways by modulating chromatin structure at the FLC locus. This provides a mechanistic framework for understanding how environmental signals dynamically influence epigenetic regulation of developmental transitions.
Collapse
Affiliation(s)
- Giorgio Perrella
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bower Building, Glasgow, G12 8QQ, UK
- Department of Biosciences, University of Milan, Via Giovanni Celoria 26, 20133, Milan, Italy
| | - Elisa Vellutini
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bower Building, Glasgow, G12 8QQ, UK
| | - Allan Beveridge
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bower Building, Glasgow, G12 8QQ, UK
| | - Graham Hamilton
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bower Building, Glasgow, G12 8QQ, UK
| | - Pawel Herzyk
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bower Building, Glasgow, G12 8QQ, UK
| | - Eirini Kaiserli
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bower Building, Glasgow, G12 8QQ, UK
| |
Collapse
|
10
|
Varshney A, Jia Z, Howe MD, Keiler KC, Baughn AD. A trans-Translation Inhibitor is Potentiated by Zinc and Kills Mycobacterium tuberculosis and Nontuberculous Mycobacteria. ACS Infect Dis 2025; 11:1140-1152. [PMID: 40202906 PMCID: PMC12071686 DOI: 10.1021/acsinfecdis.4c00963] [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: 11/29/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Mycobacterium tuberculosis poses a serious challenge for human health, and new antibiotics with novel targets are needed. Here we demonstrate that an acylaminooxadiazole, MBX-4132, specifically inhibits the trans-translation ribosome rescue pathway to kill M. tuberculosis. Our data demonstrate that MBX-4132 is bactericidal against multiple pathogenic mycobacterial species and kills M. tuberculosis in macrophages. We also show that acylaminooxadiazole activity is antagonized by iron but is potentiated by zinc. Our transcriptomic data reveal dysregulation of multiple metal homeostasis pathways after exposure to MBX-4132. Furthermore, we see differential expression of genes related to zinc sensing and efflux when trans-translation is inhibited. Taken together, these data suggest that there is a link between disturbing intracellular metal levels and acylaminooxadiazole-mediated inhibition of trans-translation. These findings provide an important proof-of-concept that trans-translation is a promising antitubercular drug target.
Collapse
Affiliation(s)
- Akanksha Varshney
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Ziyi Jia
- Department
of Microbiology and Immunology, University
of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| | - Michael D. Howe
- Department
of Microbiology and Immunology, University
of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| | - Kenneth C. Keiler
- Department
of Molecular Biosciences, The University
of Texas at Austin, Austin, Texas 78712, United States
| | - Anthony D. Baughn
- Department
of Microbiology and Immunology, University
of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
11
|
Lin ZR, Xia TL, Wang MY, Zhang LJ, Liu YM, Yuan BY, Zhou AJ, Yuan L, Zheng J, Bei JX, Lin DX, Zeng MS, Zhong Q. Inactivation of TACC2 epigenetically represses CDKN1A and confers sensitivity to CDK inhibitors. MED 2025; 6:100568. [PMID: 39793578 DOI: 10.1016/j.medj.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 10/28/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND The genomic landscape of esophageal squamous cell carcinoma (ESCC) has been characterized extensively, but there remains a significant need for actionable targets and effective therapies. METHODS Here, we perform integrative analysis of genome-wide loss of heterozygosity and expression to identify potential tumor suppressor genes. The functions and mechanisms of one of the candidates, TACC2, are then explored both in vitro and in vivo, leading to the proposal of a therapeutic strategy based on the concept of synthetic lethality. FINDINGS We reveal that the inactivation of TACC2, due to copy number loss and promoter hypermethylation, is associated with poor prognosis in ESCC patients. TACC2 depletion enhances ESCC tumorigenesis and progression, as demonstrated in Tacc2 knockout mouse models and by increased growth abilities of ESCC cells. Mechanistically, TACC2 interacts with components of the NuRD and CoREST co-repressor complexes, including MTA1, MBD3, and HMG20B, in the cytoplasm. TACC2 loss leads to the translocation of these proteins into the nucleus, facilitating the formation of functional NuRD and CoREST complexes and the epigenetic repression of CDKN1A. This repression results in elevated CDK1/2 activation. Furthermore, TACC2-deficient cells and ESCC patient-derived organoids with reduced TACC2 expression show increased sensitivity to CDK inhibitors, particularly dinaciclib, which is currently in a phase III trial. Notably, the combination of TACC2-specific RNAi and dinaciclib in subcutaneous ESCC models significantly impairs tumor growth. CONCLUSIONS The findings suggest a strategy for cancer treatment based on synthetic lethality. FUNDING Funded by NKRDP, NSFC, GDIIET, GDBABRF, GDECISTP, and SYSUTP.
Collapse
Affiliation(s)
- Zhi-Rui Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510000, P.R. China
| | - Tian-Liang Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Meng-Yao Wang
- Radiation Oncology Department, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou 510245, P.R. China
| | - Lan-Jun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Yan-Min Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Bo-Yu Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Ai-Jun Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Li Yuan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jian Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China
| | - Dong-Xin Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China; Department of Etiology and Carcinogenesis, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, P.R. China.
| |
Collapse
|
12
|
Lei X, Qiu L, Chen Q, Liao L, Yu P, Wu W, Zhu Z, Li C, Lin G, Zhuang Z, Meng Y, Wang Y, Wang C, Du Y. Exploring the regulatory mechanism of CCNA2 in colorectal cancer: insights from multiomics and experimental analysis. J Biol Chem 2025:110216. [PMID: 40345591 DOI: 10.1016/j.jbc.2025.110216] [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: 05/11/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
Colorectal cancer (CRC) is the third-most common cancer and the second-leading cause of mortality due to cancer worldwide. The underlying regulatory mechanism of CCNA2 in CRC was explored through multiomics and experimental analyses, thus facilitating diagnosis, therapy and prognosis. Two gene expression datasets (i.e., GSE9348 and GSE110223) were extracted from GEO. Differentially expressed genes (DEGs) were identified via GEO2R, which were used for enrichment analyses through DAVID. PPI network of DEGs was constructed by STRING, and the core genes were identified. CCNA2, a prognostic core gene for CRC, was validated in TCGA and HPA via transcriptomics and proteomics. ROC analysis was performed to evaluate the diagnostic value of CCNA2 in CRC. The therapeutic value of CCNA2 was evaluated in DGIdb through pharmacogenomics. The correlation between CCNA2 and immune infiltration was determined in TIMER by immunomics. TF-mRNA and miRNA-mRNA networks for CCNA2 were constructed in miRnet and miRDB via transcriptomics. The role and mechanism of CCNA2 in CRC were investigated both in vitro and in vivo. The miR-548x-3p/CCNA2 regulatory axis in CRC was investigated in vitro. CCNA2 showed excellent diagnostic, therapeutic, and prognostic value in CRC. CCNA2 was closely associated with tumor-infiltrating immunocytes, TFs, and miRNAs. The upregulation of CCNA2 was observed in CRC, and the knockdown of CCNA2 inhibited the proliferation, migration, and invasion while inducing apoptosis of CRC cells. The knockdown of CCNA2 could inhibit epithelial-mesenchymal transition (EMT) pathway. CCNA2 acted as a target of miR-548x-3p in regulating the biological behavior of CRC cells via the EMT-signaling pathway. CCNA2 is a potential biomarker for the diagnosis, treatment, and prognosis of CRC and is associated with immune infiltration, TF, and miRNA. The miR-548x-3p/CCNA2 axis plays a pivotal role in regulating the tumorigenesis of CRC through the EMT-signaling pathway.
Collapse
Affiliation(s)
- Xinyi Lei
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Lanying Qiu
- Department of Chest Radiotherapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Qiang Chen
- Department of Oncology, Cancer Diagnosis and Therapy Research Center, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Lan Liao
- Department of Pathology, the First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
| | - Pengfei Yu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wenjie Wu
- Department of Radiation Physics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhengyang Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chunying Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Gang Lin
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, Zhejiang 310024, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Zirui Zhuang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou, Zhejiang 310024, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuxin Meng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yan Wang
- Collaborative Innovation Center of Yangtza River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Cunchuan Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China.
| | - Yian Du
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
13
|
Yang J, Lin D, Liu D, Zhang D, Wang H. A PANoptosis-Based Signature for Survival and Immune Predication in Glioblastoma Multiforme. Ann Clin Transl Neurol 2025. [PMID: 40333895 DOI: 10.1002/acn3.70066] [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: 11/29/2024] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE PANoptosis is a concept of total cell death characterized by pyroptosis, apoptosis, and necroptosis. We aimed to explore the clinical significance of PANoptosis-related genes (PARGs) in glioblastoma multiforme (GBM). METHODS Expression profiles of GBM were downloaded from the XENA database as a training dataset to construct a differentially expressed PARGs (DE-PARGs)-based risk score (RS) model, and the prognostic prediction role was validated in the CGGA database and GSE108474 using Kaplan-Meier (KM) curve and receiver operating characteristic (ROC) curve. Meanwhile, independent prognostic clinical factors were screened, and their prognosis predictive activity was evaluated by a nomogram model. Furthermore, the relationships between key DE-PARGs and immune cell infiltration, as well as chemotherapy drug sensitivity were analyzed. RESULTS The RS model consisting of five DE-PARGs was constructed, including NOD2, NLRP2, NLRP7, GATA3, and TERT. ROC and KM curves confirmed the good potency of the RS prognostic model both in XENA database and GSE108474. Three clinical prognostic factors, including chemotherapy, pharmaceutical therapy, and RS model, were selected as individual prognostic factors. The nomogram model showed RS contributed most to survival probability, followed by chemotherapy and pharmaceutical therapy. In high- and low-risk groups, B cell memory, NK cell resting, and macrophage M1 had significant differences. As compared with the immune checkpoint therapy non-responder group, the responder involved a higher ratio of patients sub-grouped into the low-risk group. Three drugs between high- and low-risk groups had significant differences, including Cisplatin, Gefitinib, and Vorinostat. INTERPRETATION Our data exhibit the prognostic value of PARGs in GBM and offer new insights for GBM pathogenesis and immune treatment.
Collapse
Affiliation(s)
- Jun Yang
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Da Lin
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Dongyuan Liu
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Dongxu Zhang
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Hao Wang
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Maurice De Sousa D, Perkey E, Le Corre L, Boulet S, Gómez Atria D, Allman A, Duval F, Daudelin JF, Brandstadter JD, Lederer K, Mezrag S, Odagiu L, Ennajimi M, Sarrias M, Decaluwe H, Koch U, Radtke F, Ludewig B, Siebel CW, Maillard I, Labrecque N. Early Notch signals from fibroblastic reticular cells program effector CD8+ T cell differentiation. J Exp Med 2025; 222:e20231758. [PMID: 40111253 PMCID: PMC11925062 DOI: 10.1084/jem.20231758] [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: 09/27/2023] [Revised: 09/06/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
A better understanding of the mechanisms regulating CD8+ T cell differentiation is essential to develop new strategies to fight infections and cancer. Using genetic mouse models and blocking antibodies, we uncovered cellular and molecular mechanisms by which Notch signaling favors the efficient generation of effector CD8+ T cells. Fibroblastic reticular cells from secondary lymphoid organs, but not dendritic cells, were the dominant source of Notch signals in T cells via Delta-like1/4 ligands within the first 3 days of immune responses to vaccination or infection. Using transcriptional and epigenetic studies, we identified a unique Notch-driven T cell-specific signature. Early Notch signals were associated with chromatin opening in regions occupied by bZIP transcription factors, specifically BATF, known to be important for CD8+ T cell differentiation. Overall, we show that fibroblastic reticular cell niches control the ultimate molecular and functional fate of CD8+ T cells after vaccination or infection through the delivery of early Notch signals.
Collapse
Affiliation(s)
- Dave Maurice De Sousa
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Canada
| | - Eric Perkey
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Laure Le Corre
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Canada
| | - Salix Boulet
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
| | - Daniela Gómez Atria
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anneka Allman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frédéric Duval
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
| | | | | | - Katlyn Lederer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah Mezrag
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Canada
| | - Livia Odagiu
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Myriam Ennajimi
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Marion Sarrias
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Hélène Decaluwe
- Cytokines and Adaptive Immunity Laboratory, CHU Sainte-Justine Research Center, Montreal, Canada
| | - Ute Koch
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Freddy Radtke
- École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | | | - Ivan Maillard
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Canada
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Canada
- Département de Médecine, Université de Montréal, Montreal, Canada
| |
Collapse
|
15
|
O’Donoghue S, Earley B, Johnston D, Finnie MS, Cosby SL, Lemon K, McMenamy MJ, Taylor JF, Kim JW, Morris DW, Waters SM. Examination of the lung and lymphoid tissue mRNA transcriptome response in dairy calves following experimental challenge with bovine alphaherpesvirus one (BoHV-1). PLoS One 2025; 20:e0319575. [PMID: 40315186 PMCID: PMC12047826 DOI: 10.1371/journal.pone.0319575] [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: 09/09/2024] [Accepted: 02/04/2025] [Indexed: 05/04/2025] Open
Abstract
Bovine alphaherpesvirus one (BoHV-1) is a primary cause of bovine respiratory disease (BRD), and a leading cause of morbidity and mortality in cattle. The transcriptomic responses of key respiratory and immune associated tissues of dairy calves following experimental challenge with BoHV-1 are unknown. Thus, the study objective was to examine the gene expression profiles of multiple tissue types from dairy calves following an infectious challenge with BoHV-1. Holstein-Friesian bull calves (mean age ± SD 149.2 days ± 23.8; mean weight ± SD 174.6 kg ± 21.3 kg were challenged with either BoHV-1 inoculate (6.3 × 107/mL × 1.35mL) (n = 12) or sterile phosphate buffered saline (n = 6). Animals were euthanised on day 6 post-challenge and tissue samples collected, including bronchial (BLN) and mediastinal lymph nodes (MLN), pharyngeal tonsil (PGT) and healthy (HL) and lesioned right cranial lung (LL). Total RNA was extracted and libraries sequenced on an Illumina NovaSeq 6000. Differential expression analysis was conducted using edgeR and pathways analysed using DAVID. A weighted gene co-expression network analysis (WGCNA) was conducted separately for each tissue type to identify networks significantly associated with BoHV-1 infection. Differentially expressed genes (DEGs) were identified in all tissues (P < 0.05, FDR < 0.1, FC > 2). Thirty-three DEGs were common to all tissues and enriched pathways included Influenza A and Herpes simplex 1 infection (P < 0.05, FDR < 0.05). Modules enriched for antiviral and innate immune processes were identified for each tissue type. Of the 33 DEGs common to all tissues, 26 were also identified as hub genes in the blood (blue) module. Our use of a controlled experimental challenge allowed for improved understanding of the immune response of dairy calves to a BoHV-1 infection. Furthermore, discovering DEGs that are common to all tissues, including whole blood, indicates future focus areas in research surrounding BRD diagnostic biomarkers.
Collapse
Affiliation(s)
- Stephanie O’Donoghue
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Grange, Meath, Ireland
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Bernadette Earley
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Grange, Meath, Ireland
| | - Dayle Johnston
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Grange, Meath, Ireland
| | - Matthew S. Finnie
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Grange, Meath, Ireland
| | - S. Louise Cosby
- Veterinary Sciences Division, Agri-Food and Biosciences Institute, Stormont, Belfast, Ireland,
| | - Ken Lemon
- Veterinary Sciences Division, Agri-Food and Biosciences Institute, Stormont, Belfast, Ireland,
| | - Michael J. McMenamy
- Veterinary Sciences Division, Agri-Food and Biosciences Institute, Stormont, Belfast, Ireland,
| | - Jeremy F. Taylor
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Jae Woo Kim
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Derek W. Morris
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Sinéad M. Waters
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
16
|
Ojiro R, Zou X, Yamagata H, Ebizuka Y, Kobayashi M, Kigata T, Tang Q, Yoshida T, Yoshinari T, Shibutani M. Emerging mycotoxin moniliformin induces renal tubular necrosis after oral exposure in mice. Food Chem Toxicol 2025; 199:115336. [PMID: 39984025 DOI: 10.1016/j.fct.2025.115336] [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: 12/17/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Toxicological information on moniliformin (MON), an emerging mycotoxin, is limited. This study examined the acute and 28-day toxicity of orally administered MON in male ICR mice. Regarding the acute toxicity, among single oral doses of 0, 20, 40, and 80 mg/kg body weight (BW), MON caused proximal tubular necrosis in the kidneys at ≥ 40 mg/kg BW, and the lethal dose 50 value was estimated as 68.1 mg/kg BW. Regarding the 28-day toxicity, among oral doses of 0, 10, 20, and 40 mg/kg BW/day, MON increased absolute heart weight at 40 mg/kg BW, but histopathological changes were not evident in the heart. In contrast, 40 mg/kg BW MON induced centrilobular liver cell hypertrophy accompanied by increased absolute liver weight. Moreover, MON dose-dependently increased the absolute kidney weight at ≥ 20 mg/kg BW and increased the incidence of renal tubular regeneration at 40 mg/kg BW. RNA sequencing analysis in the renal cortex after a single dose of 40 mg/kg BW MON revealed upregulation of metabolic response-related genes, such as Cyp3a13, Cyp26b1, and Cyp4f15, and oxidative stress-related Gpx7. These results suggest that MON targets the kidneys in mice. Orally ingested MON may be metabolized in the kidneys as well as in the liver, and active intermediates or reactive oxygen species may induce renal tubular toxicity, causing proximal tubular necrosis. Based on kidney changes, the no-observed-adverse-effect-level of MON in the 28-day oral toxicity study of male mice was determined to be 10 mg/kg BW/day.
Collapse
Affiliation(s)
- Ryota Ojiro
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Hiroshi Yamagata
- Toxicology Division, Gotemba Laboratory, BoZo Research Center Inc., 1284 Kamado, Gotemba-shi, Shizuoka, 412-0039, Japan.
| | - Yuri Ebizuka
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Tetsuhito Kigata
- Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Laboratory of Veterinary Anatomy, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Qian Tang
- General Toxicology Department, TriApex Laboratories Co., Ltd., No. 9 Xinglong Road, Jiangbei New Area, Nanjing, Jiangsu Province, 211800, China.
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Tomoya Yoshinari
- Division of Microbiology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa, 210-9501, Japan.
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| |
Collapse
|
17
|
Yang H, Zheng Y, Yu T, Wu B, Liu Z, Liu S, Sun X, Zhou L. A functional role for myostatin in muscle hyperplasia and hypertrophy revealed by comparative transcriptomics in Yesso scallop Patinopecten yessoensis. Int J Biol Macromol 2025; 307:142308. [PMID: 40118415 DOI: 10.1016/j.ijbiomac.2025.142308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/03/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Elucidating the molecular regulatory mechanisms underlying muscle growth and development is of profound significance in aquaculture. Yesso scallop is a cold-water bivalve of considerable economic importance, having its primary edible component of adductor muscle. In this study, comparative transcriptomics and histological analysis at different sampling times after Myostatin (MSTN) interference were performed to identify the potential candidate genes potentially involved in muscle growth and development. The comparative transcriptomics revealed that growth factors and cytokines, extracellular matrix proteins and ubiquitin-proteasome system are potentially involved in muscle hypertrophy and hyperplasia. After MSTN interference, striated adductor muscle displays significant muscle hypertrophy (51.77 % increase on day 7 and 59.83 % increase on day 21) and muscle hyperplasia (59.36 % increase on day 7 and 61.83 % increase on day 21). WGCNA identifies the key darkolivegreen module, which may play crucial roles in muscle hyperplasia and hypertrophy within the striated muscle of the scallop. Five key transcription factors (zf-CCCH, zf-C2H2, PPP1R10, LRRFIP2, and Gon4) are identified by analyzing the co-expression patterns of core genes within the module. These findings will aid in understanding the regulatory mechanisms of muscle growth in scallops and provide a basis for genetic improvement in shellfish aquaculture.
Collapse
Affiliation(s)
- Hongsu Yang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China; Fisheries College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yanxin Zheng
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Changdao, China
| | - Tao Yu
- Changdao Enhancement and Experiment Station, Chinese Academy of Fishery Sciences, Changdao, China
| | - Biao Wu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| | - Zhihong Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| | - Shufang Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| | - Xiujun Sun
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China.
| | - Liqing Zhou
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, Shandong 266237, China
| |
Collapse
|
18
|
Chang R, Xue F, Hou Z, Guo H, Cao L, Zhang S, Wang W, Qu C, Yang C, Liu G, Xu Z. Integrative analysis of transcriptome, proteome, and phosphoproteome reveals the complexity of early nitrogen responses in poplar roots. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 222:109703. [PMID: 40054111 DOI: 10.1016/j.plaphy.2025.109703] [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: 11/24/2024] [Revised: 12/28/2024] [Accepted: 02/24/2025] [Indexed: 05/07/2025]
Abstract
Nitrogen (N) availability is a key factor in plant growth, but the molecular mechanisms underlying the early responses of poplar (Populus × xiaohei T. S. Hwang & Liang) roots to nitrogen are not well understood. The primary objective of this study was to elucidate these early molecular responses by integrating transcriptome, proteome, and phosphoproteome under low-nitrogen (LN, 0.2 mM NH4NO3) and high-nitrogen (HN, 2 mM NH4NO3) conditions. Specifically, the objectives of this study were: (i) to identify key metabolic pathways involved in nitrogen responses in poplar roots; (ii) to explore the relationship between differentially expressed genes (DEGs) and transcription factors (TFs) within these pathways; and (iii) to construct co-expression networks to uncover the regulatory mechanisms of nitrogen signaling. KEGG pathway enrichment analysis indicated that nitrogen metabolism and phenylpropanoid metabolism were key pathways in RNA-seq and proteome, while starch and sucrose metabolism were crucial in transcriptome and phosphoproteome. Plant hormone signal transduction was a key pathway in transcriptome, and gluconeogenesis/glycolysis was essential in proteome. WGCNA revealed three key modules (MEgreenyellow, MEblack, and MEblue) significantly associated with physiological indices, including NO3-, soluble sugar, and sucrose contents. Co-expression networks highlighted TFs as central regulators of nitrogen-responsive pathways, with distinct expression patterns between LN and HN treatments. These findings elucidate the complexity of nitrogen-regulated metabolic networks in poplar roots and reveal potential links between nitrogen signaling, carbohydrate metabolism, and secondary metabolism. This study provides a foundation for improving nitrogen-use efficiency in forest trees, with implications for sustainable forestry and ecosystem management.
Collapse
Affiliation(s)
- Ruhui Chang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Fengbo Xue
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Zhaoyin Hou
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Hongye Guo
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Lina Cao
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; School of Forestry, Northeast Forestry University, Harbin, 150040, China
| | - Shuang Zhang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Wenjie Wang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China
| | - Chunpu Qu
- College of Forestry, Guizhou University, Guiyang, 550025, China
| | - Chuanping Yang
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; School of Forestry, Northeast Forestry University, Harbin, 150040, China
| | - Guanjun Liu
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; School of Forestry, Northeast Forestry University, Harbin, 150040, China.
| | - Zhiru Xu
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin, 150040, China; College of Life Science, Northeast Forestry University, Harbin, 150040, China.
| |
Collapse
|
19
|
Valima E, Varis V, Bureiko K, Lempiäinen JK, Schroderus AM, Oksa L, Lohi O, Kinnunen T, Varjosalo M, Niskanen EA, Paakinaho V, Palvimo JJ. SUMOylation inhibition potentiates the glucocorticoid receptor to program growth arrest of acute lymphoblastic leukemia cells. Oncogene 2025; 44:1259-1271. [PMID: 39953147 PMCID: PMC12048349 DOI: 10.1038/s41388-025-03305-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/20/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Glucocorticoids are a mainstay in the treatment of B-cell acute lymphoblastic leukemia (B-ALL). The glucocorticoid receptor (GR), a ligand-activated transcription factor (TF), mediates their actions. Chromatin occupancy, chromatin-protein networks (chromatomes) and gene programmes of GR are regulated by SUMOylation, a post-translational modification with therapeutic implications in other hematomalignancies. To unravel the GR-SUMOylation crosstalk in B-ALL, we induced hypoSUMOylation in NALM6 B-ALL cells with a SUMOylation inhibitor (SUMOi, ML-792). Genome-wide profiling of GR and SUMO chromatin-binding and chromatin accessibility revealed that hypoSUMOylation augmented GR chromatin occupancy and altered chromatin openness. Association with transcriptome data indicated that the hypoSUMOylation-induced GR-binding sites predominantly repressed genes associated with cell cycle and DNA replication. Consistently, hypoSUMOylation potentiated glucocorticoid-induced cell cycle arrest and growth suppression. Moreover, our proteomic analyses revealed that the protein network of chromatin-bound GR is tightly intertwined with SUMO2/3 and that SUMOylation modulates the stability of the network. The chromatome contained several B-cell TFs with cognate binding motifs found on GR-adjacent chromatin sites, indicating their simultaneous occupancy on chromatin. In sum, our data imply potential for targeting SUMOylation to increase sensitivity to glucocorticoids in B-ALL, supported by ex vivo data of glucocorticoid and SUMOi TAK-981 combination-treated B-ALL patient samples.
Collapse
Affiliation(s)
- Emma Valima
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Vera Varis
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Kseniia Bureiko
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | | | | | - Laura Oksa
- Tampere Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
| | - Olli Lohi
- Tampere Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
- Tampere University Hospital, Tays Cancer Center, Tampere, Finland
| | - Tuure Kinnunen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
- ISLAB Laboratory Centre, Kuopio, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- HiLIFE-Proteomics Unit, University of Helsinki, Helsinki, Finland
| | - Einari A Niskanen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Ville Paakinaho
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
20
|
Rani P, Rajak BK, Mahato GK, Rathore RS, Chandra G, Singh DV. Strategic lead compound design and development utilizing computer-aided drug discovery (CADD) to address herbicide-resistant Phalaris minor in wheat fields. PEST MANAGEMENT SCIENCE 2025; 81:2469-2479. [PMID: 39377567 DOI: 10.1002/ps.8455] [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: 05/30/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024]
Abstract
Wheat (Triticum aestivum) is a vital cereal crop and a staple food source worldwide. However, wheat grain productivity has significantly declined as a consequence of infestations by Phalaris minor. Traditional weed control methods have proven inadequate owing to the physiological similarities between P. minor and wheat during early growth stages. Consequently, farmers have turned to herbicides, targeting acetyl-CoA carboxylase (ACCase), acetolactate synthase (ALS) and photosystem II (PSII). Isoproturon targeting PSII was introduced in mid-1970s, to manage P. minor infestations. Despite their effectiveness, the repetitive use of these herbicides has led to the development of herbicide-resistant P. minor biotypes, posing a significant challenge to wheat productivity. To address this issue, there is a pressing need for innovative weed management strategies and the discovery of novel herbicide molecules. The integration of computer-aided drug discovery (CADD) techniques has emerged as a promising approach in herbicide research, that facilitates the identification of herbicide targets and enables the screening of large chemical libraries for potential herbicide-like molecules. By employing techniques such as homology modelling, molecular docking, molecular dynamics simulation and pharmacophore modelling, CADD has become a rapid and cost-effective medium to accelerate the herbicide discovery process significantly. This approach not only reduces the dependency on traditional experimental methods, but also enhances the precision and efficacy of herbicide development. This article underscores the critical role of bioinformatics and CADD in developing next-generation herbicides, offering new hope for sustainable weed management and improved wheat cultivation practices. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Priyanka Rani
- Molecular Modelling and Computer-Aided Drug Discovery Laboratory Department of Bioinformatics, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Bikash Kumar Rajak
- Molecular Modelling and Computer-Aided Drug Discovery Laboratory Department of Bioinformatics, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Gopal Kumar Mahato
- Department of Chemistry, School of Physical and Chemical Sciences, Central University of South Bihar, Gaya, India
| | - Ravindranath Singh Rathore
- Molecular Modelling and Computer-Aided Drug Discovery Laboratory Department of Bioinformatics, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Girish Chandra
- Department of Chemistry, School of Physical and Chemical Sciences, Central University of South Bihar, Gaya, India
| | - Durg Vijay Singh
- Molecular Modelling and Computer-Aided Drug Discovery Laboratory Department of Bioinformatics, School of Earth, Biological and Environmental Sciences, Central University of South Bihar, Gaya, India
| |
Collapse
|
21
|
Nasti A, Inagaki S, Ho TTB, Seki A, Yoshida K, Satomura K, Sakai Y, Kaneko S, Yamashita T. Cystatin A promotes the antitumor activity of T helper type 1 cells and dendritic cells in murine models of pancreatic cancer. Mol Oncol 2025; 19:1452-1470. [PMID: 39792573 PMCID: PMC12077287 DOI: 10.1002/1878-0261.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/21/2024] [Accepted: 12/12/2024] [Indexed: 01/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with poor prognosis due to diagnostic and therapeutic limitations. We previously identified cystatin A (CSTA) as a PDAC biomarker and have conducted the present study to investigate the antitumor effects of CSTA. PDAC murine models were established with genetically modified PAN02 tumor cell lines to evaluate the antitumor immune response. PDAC mouse survival was significantly longer with CSTA, and its antitumor effect was mediated mainly by CD4+ cells and partly by CD8+ cells. We also observed an increased infiltration of CD4+ and CD8+ cells in tumors of mice overexpressing CSTA. Phenotypically, we confirmed higher T helper type 1 (Th1) cell activity and increased frequency and activity of M1 macrophages and dendritic cells (DCs) in CSTA-overexpressing mice. Gene expression analysis highlighted pathways related to interferon gamma (IFN-γ) induction and Th1 lymphocyte activation that were induced by CSTA. Macrophages and DCs shifted toward proinflammatory antitumor phenotypes. Furthermore, activated splenocytes of PDAC model mice expressing CSTA had increased proapoptotic activity. CSTA also promoted the selective migration of CD4+ and CD11c+ immune cells in an in vitro migration assay. In conclusion, CSTA exerts antitumor effects by enhancing Th1-mediated antitumor effects through promotion of DC and M1 macrophage activity, thereby increasing immune cell chemotaxis. CSTA could be a novel therapeutic candidate for PDAC.
Collapse
Affiliation(s)
- Alessandro Nasti
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Shingo Inagaki
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Tuyen Thuy Bich Ho
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
| | - Akihiro Seki
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Keiko Yoshida
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Kosuke Satomura
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Yoshio Sakai
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
| | - Shuichi Kaneko
- Information‐Based Medicine DevelopmentGraduate School of Medical Sciences, Kanazawa UniversityJapan
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| | - Taro Yamashita
- System Biology, Graduate School of Advanced Preventive Medical SciencesKanazawa UniversityJapan
- Department of GastroenterologyKanazawa University HospitalJapan
| |
Collapse
|
22
|
Jing S, Zhenhai C, Xiao H, Chunhan L, Wanlin W, Xuting S, Han Y, Xiaoyu Z, Meiying J. Jiedu Tongluo Tiaogan Formula Modulates Glycolipid Metabolism in Type 2 Diabetes via Pyroptosis: Network Pharmacology and In Vivo Analysis. Biomed Chromatogr 2025; 39:e70077. [PMID: 40219662 DOI: 10.1002/bmc.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by pancreatic β-cell dysfunction and insulin resistance, with pyroptosis emerging as a key contributor to β-cell loss. Jiedu Tongluo Tiaogan Formula (JTTF), a traditional Chinese medicine (TCM), has shown clinical efficacy in T2DM management, but its mechanism linking pyroptosis remains unexplored. This study integrates UPLC-MS/MS, network pharmacology, and in vivo experiments to elucidate JTTF's anti-diabetic mechanisms. UPLC-MS/MS identified 441 compounds in JTTF, predominantly alkaloids, flavonoids, phenols, and terpenoids. Network pharmacology revealed JTTF's multi-target effects on T2DM-associated pyroptosis, particularly via the NLRP3/Caspase-1/GSDMD pathway. In diabetic mice, JTTF dose-dependently reduced fasting blood glucose, insulin resistance, and dyslipidemia, while restoring pancreatic β-cell morphology. Mechanistically, JTTF suppressed NLRP3 inflammasome activation, downregulated Caspase-1 and GSDMD expression, and attenuated IL-1β/IL-18 release. Notably, this study provides the first evidence of JTTF's anti-pyroptotic effects in T2DM, highlighting its unique ability to modulate glycolipid metabolism and inflammatory cell death concurrently. These findings underscore JTTF's translational promise for preserving β-cell function and suggest future exploration of non-classical pyroptosis pathways. Our work bridges traditional medicine and molecular pharmacology, paving the way for clinical trials and integrative T2DM therapies.
Collapse
Affiliation(s)
- Su Jing
- Changchun University of Chinese Medicine, Changchun, China
| | - Cui Zhenhai
- Changchun University of Chinese Medicine, Changchun, China
| | - Han Xiao
- Changchun University of Chinese Medicine, Changchun, China
| | - Li Chunhan
- Jilin Hospital of Integrated Traditional Chinese and Western Medicine, Jilin, China
| | - Wang Wanlin
- Changchun University of Chinese Medicine, Changchun, China
| | - Song Xuting
- Changchun University of Chinese Medicine, Changchun, China
| | - Yu Han
- Changchun University of Chinese Medicine, Changchun, China
| | - Zhuang Xiaoyu
- Changchun University of Chinese Medicine, Changchun, China
| | - Jin Meiying
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
23
|
Holloway K, Neherin K, Song Y, Sato K, Houston A, Chen F, Ding L, Zhang H. Elevated p16Ink4a Expression Enhances Tau Phosphorylation in Neurons Differentiated From Human-Induced Pluripotent Stem Cells. Aging Cell 2025; 24:e14472. [PMID: 39757785 PMCID: PMC12073902 DOI: 10.1111/acel.14472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Increased expression of the cyclin-dependent kinase inhibitor p16Ink4a (p16) is detected in neurons of human Alzheimer's disease (AD) brains and during normal aging. Importantly, selective eliminating p16-expressing cells in AD mouse models attenuates tau pathologies and improves cognition. But whether and how p16 contributes to AD pathogenesis remains unclear. To address this question, we tested whether induction of p16 expression in neurons exacerbates AD pathologies. We created a doxycycline-inducible system to trigger p16 up-regulation in human-induced pluripotent stem cells (iPSCs) and neurons differentiated from iPSCs. We demonstrated that up-regulated p16 expression in iPSCs reduces cell proliferation, down-regulates cell cycle genes, and up-regulates genes involved in focal adhesion, interferon α response and PI3K-Akt signaling. Our approach enables temporal control of p16 induction upon differentiation from iPSCs to neurons. In differentiated cortical neurons, we found that up-regulation of p16 increases tau phosphorylation at Ser202/Thr205 and Thr231 in a cell-autonomous manner, while amyloid beta secretion is not affected. These data suggest a critical role of p16 in regulating tau phosphorylation in neurons, and thereby contributing to pathological progression of AD. As pathological tau tangles have been shown to induce p16 expression, our studies suggest a positive feedback loop between p16 and tau to exacerbate tau pathologies.
Collapse
Affiliation(s)
- Kristopher Holloway
- Department of Pediatrics, 3 NeuroNexus InstituteUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Kashfia Neherin
- Department of Pediatrics, 3 NeuroNexus InstituteUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Yingduo Song
- Department of Medicine, McDonnell Genome InstituteWashington University School of MedicineSt. LouisMissouriUSA
| | - Kazuhito Sato
- Department of Medicine, McDonnell Genome InstituteWashington University School of MedicineSt. LouisMissouriUSA
| | - Andrew Houston
- Department of Medicine, McDonnell Genome InstituteWashington University School of MedicineSt. LouisMissouriUSA
| | - Feng Chen
- Department of Medicine, McDonnell Genome InstituteWashington University School of MedicineSt. LouisMissouriUSA
| | - Li Ding
- Department of Medicine, McDonnell Genome InstituteWashington University School of MedicineSt. LouisMissouriUSA
| | - Hong Zhang
- Department of Pediatrics, 3 NeuroNexus InstituteUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
24
|
Choi H, Lee JY, Yoo H, Jeon K. Identification and Verification of Necroptosis-Related Genes in Patients With Sepsis by Bioinformatic Analysis and Molecular Experiments. J Cell Mol Med 2025; 29:e70582. [PMID: 40318009 PMCID: PMC12049152 DOI: 10.1111/jcmm.70582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
Although necroptosis is an emerging mechanism of multiple organ dysfunction in sepsis, data on the mechanistic link between necroptosis and sepsis are scarce. Bioinformatic analysis was performed to compare the gene profiles between the sepsis (n = 133) and healthy control (n = 12) groups and identify necroptosis-related differentially expressed genes (DEGs). The identified necroptosis-related DEGs were verified by three-step molecular experiments: (1) quantitative real-time PCR and enzyme-linked immunosorbent assay; (2) cell culture, transfection and Western blotting; and (3) cytokine array with apoptosis inhibition. Additionally, receiver-operating characteristic curve analyses were performed to evaluate the performance of the corresponding proteins to the necroptosis-related DEGs in diagnosing sepsis and in predicting in-hospital mortality of patients with sepsis. Eight necroptosis-related DEGs, including five upregulated (PYGL, TNF, CYLD, FADD and TLR3) and three downregulated (TP53, FASLG and NLRP6) DEGs, were identified. Moreover, the levels of the corresponding proteins to necroptosis-related DEGs showed excellent or considerable accuracy in diagnosing sepsis and in predicting the mortality of sepsis patients. In cell culture media transfected with plasma from the sepsis and control groups, Western blotting revealed that the levels of the corresponding proteins were increased in the upregulated DEGs and decreased in the downregulated DEGs. The cytokine array revealed cytokines in cell culture media transfected with plasma from patients with sepsis while preventing apoptosis by inhibiting the caspase-8 activity, wherein the transfected cells potentially underwent necroptosis. Eight necroptosis-related DEGs were identified in patients with sepsis by bioinformatic analysis and verified by molecular experiments, implying that necroptosis may be a key mechanism of sepsis.
Collapse
Affiliation(s)
- Hayoung Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal MedicineHallym University Kangnam Sacred Heart Hospital, Hallym University College of MedicineSeoulRepublic of Korea
| | - Jin Young Lee
- Division of Pulmonary and Critical Care Medicine, Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Hongseok Yoo
- Division of Pulmonary and Critical Care Medicine, Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
- Department of Health Science TechnologySAIHST, Sungkyunkwan UniversitySeoulRepublic of Korea
| |
Collapse
|
25
|
Guo Q, Xie M, Wang X, Han C, Gao G, Wang QN, Li J, Duan L, Bao X. Multi-omic serum analysis reveals ferroptosis pathways and diagnostic molecular signatures associated with Moyamoya diseases. J Neuroinflammation 2025; 22:123. [PMID: 40301939 PMCID: PMC12042548 DOI: 10.1186/s12974-025-03446-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Moyamoya disease (MMD) is a rare cerebrovascular disease in humans. Although early revascularization can improve symptoms, it cannot reverse the progression of the disease. The current diagnosis still relies on traditional a Digital Subtraction Angiography (DSA) examination, which is invasive and expensive, leading to delayed diagnosis and affecting treatment timing and patient prognosis. The ability to diagnose MMD early and develop personalized treatment plans can significantly improve the prognosis of patients. Here, we have introduced the research on MMD biomarkers. By integrating proteomics and metabolomics data, we have successfully identified over 1700 features from more than 60 serum samples collected at the onset of symptoms in MMD patients. We use multiple computational strategies to interpret complex information in serum, providing a comprehensive perspective for early diagnosis of MMD. Diagnostic ability of our biomarker is significantly better than previous studies, especially when used in combination. In the study of molecular mechanisms, we found that the ferroptosis pathway was significant disruption in MMD patients, which was also confirmed by transcriptomics data. Finally, we validated the metabolites and proteins associated with ferroptosis pathways, as well as the biomarkers screened by machine learning, using another independent MMD cohort. Our research provides important clues for the diagnosis of MMD, and this assay can identify MMD early, thereby promoting stronger monitoring and intervention.
Collapse
Affiliation(s)
- Qingbao Guo
- Department of Neurosurgery, XI'AN NO.9 HOSPITAL, Shaanxi, 710054, China.
| | - Manli Xie
- Department of Occupational Diseases, Xi'an Central Hospital, Shaanxi, 710003, China
| | - Xiaopeng Wang
- Medical School of Chinese PLA, Beijing, 100039, China
| | - Cong Han
- Department of Neurosurgery, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China
| | - Gan Gao
- Medical School of Chinese PLA, Beijing, 100039, China
| | - Qian-Nan Wang
- Department of Neurosurgery, the Eighth Medical Centre, Chinese PLA General Hospital, Beijing, 100089, China
| | - Jingjie Li
- Medical School of Chinese PLA, Beijing, 100039, China
| | - Lian Duan
- Department of Neurosurgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiangyang Bao
- Department of Neurosurgery, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
26
|
Morito M, Hata K, Izumi Y, Bamba T, Matsumori N. Comprehensive Identification of Lipid-Membrane Protein Interactions via Advanced Proteomics and Extended Lipid-Immobilized Bead Technology. Anal Chem 2025; 97:8880-8889. [PMID: 40233011 PMCID: PMC12044594 DOI: 10.1021/acs.analchem.5c00074] [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: 01/05/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
In biological membranes, lipids interact with membrane proteins (MPs) and play important roles in allosterically regulating their structure and function. Analyzing lipid-MP interactions is necessary for understanding these regulatory mechanisms; however, there have been few comprehensive and systematic studies to date. To address this, we developed a high-sensitivity, high-throughput platform that integrates lipid-immobilized beads with advanced proteomics to analyze lipid-MP interactions in detail. We prepared six types of lipid-immobilized beads, including sphingomyelin (SM), ceramide (Cer), dihydrosphingomyelin (DHSM), dihydroceramide (DHCer), phosphatidylcholine (PC), and cholesterol (Chol). In addition, we introduced a novel type of beads that immobilized SM and Chol (SM/Chol beads) to mimic lipid rafts. We first demonstrated that SM/Chol beads coprecipitated with Nakanori, a protein that specifically recognizes and binds to SM/Chol complexes, whereas beads immobilized with SM or Chol alone did not coprecipitate. This indicates the effectiveness of SM/Chol beads for the identification of raft-associated proteins. Next, the cell lysates were incubated with the seven types of lipid-immobilized beads and the recovered proteins were analyzed using shotgun proteomics. This approach successfully identified over 7000 lipid-binding proteins. Filtering based on fold-change values and subsequent enrichment analysis revealed distinct binding protein profiles for each lipid, highlighting the functional diversity of lipid-MP interactions and their roles in cellular processes. In summary, our methodology enables an unprecedented large-scale exploration of lipid-MP interactions. This platform provides a versatile tool for examining the lipid-mediated regulation of MPs and offers new insights into the physiological significance of the lipidome and its implications for health and disease.
Collapse
Affiliation(s)
- Masayuki Morito
- Department
of Chemistry, Graduate School of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kosuke Hata
- Division
of Metabolomics, Medical Research Center for High Depth Omics, Medical
Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi,
Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division
of Metabolomics, Medical Research Center for High Depth Omics, Medical
Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi,
Higashi-ku, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division
of Metabolomics, Medical Research Center for High Depth Omics, Medical
Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi,
Higashi-ku, Fukuoka 812-8582, Japan
| | - Nobuaki Matsumori
- Department
of Chemistry, Graduate School of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
27
|
Boukerouis D, Cuadrado I, Benaida ND, Estévez-Braun A, de Las Heras B, Amesty A, Hortelano S. Exploring the anti-inflammatory activity of fupenzic acid using network pharmacology and experimental validation. Sci Rep 2025; 15:14294. [PMID: 40274978 PMCID: PMC12022156 DOI: 10.1038/s41598-025-98901-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Crataegus azarolus L. (Rosaceae), commonly known as Mediterranean hawthorn, has long been valued in Traditional Medicine for treating cardiovascular and inflammation-related diseases, including diabetes, cancer, and rheumatism. Pharmacological benefits of Crataegus azarolus L. are notably linked to its anti-inflammatory properties. Fupenzic acid, a pentacyclic triterpene isolated from its leaves, holds significant pharmacological potential that remains elusive. This study investigates the unexplored capacity of fupenzic acid as a promising anti-inflammatory agent. Using a multidisciplinary approach that integrates network pharmacology, molecular docking, in vitro assays, and predictive in silico analyses of drug-like properties, ADME, and toxicity, the mechanisms and properties of fupenzic acid have been elucidated. Network pharmacology analysis identified the potential targets for fupenzic acid, with enrichment analyses revealing key processes like inflammatory response, cytokine signaling, innate immune system, and MAPK cascade regulation. Transcription factors such as RELA, SP1, and NFKB1 were predicted to play crucial roles in its therapeutic effects. PPI network analysis underscored NF-κB as a central hub, linking these pathways to its anti-inflammatory effects. In vitro experiments demonstrated that fupenzic acid effectively suppressed inflammatory mediators like NOS-2 and COX-2, through the NF-κB pathway. Molecular docking further confirmed its favorable interaction with NF-κB, reinforcing its mechanism of action. Additionally, in silico ADMET profiling revealed favorable drug-like properties including pharmacokinetics and toxicity profiles, emphasizing its suitability as a drug candidate. This study represents a major step forward in understanding the therapeutic potential of fupenzic acid, establishing it as a distinctive and promising anti-inflammatory agent. The findings identified it as a pharmacological agent for clinical development targeting inflammation-driven diseases and also provide a foundation for future translational research.
Collapse
Affiliation(s)
- Djoudi Boukerouis
- Laboratoire de Biochimie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000, Bejaia, Algeria
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de la Laguna, Avda. Astrofísico Francisco Sánchez Nº 2, 38206, La Laguna, Tenerife, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia , Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
- Department of Biology, Faculty of Science, University of Amar Tlidji, 03000, Laghouat, Algeria
| | - Irene Cuadrado
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia , Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Nadjet Debbache Benaida
- Laboratoire de Biochimie Appliquée, Faculté des Sciences de la Nature et de la Vie, Université de Bejaia, 06000, Bejaia, Algeria
| | - Ana Estévez-Braun
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de la Laguna, Avda. Astrofísico Francisco Sánchez Nº 2, 38206, La Laguna, Tenerife, Spain
| | - Beatriz de Las Heras
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia , Universidad Complutense de Madrid (UCM), Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.
| | - Angel Amesty
- Departamento de Química Orgánica, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de la Laguna, Avda. Astrofísico Francisco Sánchez Nº 2, 38206, La Laguna, Tenerife, Spain.
| | - Sonsoles Hortelano
- Unidad de Terapias Farmacológicas, Área de Genética Humana, Instituto de Investigación de Enfermedades Raras (IIER), Instituto de Salud Carlos III, Carretera de Majadahonda-Pozuelo Km 2, 28220, Madrid, Spain.
| |
Collapse
|
28
|
Yeon Chae S, Jang SY, Kim J, Hwang S, Malani D, Kallioniemi O, Yun SG, Kim JS, Kim HI. Mechanisms of chemotherapy failure in refractory/relapsed acute myeloid leukemia: the role of cytarabine resistance and mitochondrial metabolism. Cell Death Dis 2025; 16:331. [PMID: 40268906 PMCID: PMC12019594 DOI: 10.1038/s41419-025-07653-6] [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: 10/18/2024] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy. Patients with wild-type FLT3 relapsed or refractory (R/R) AML face significant therapeutic challenges due to the persistent lack of effective treatments. A comprehensive understanding of the mechanisms underlying chemotherapy resistance is needed to the development of effective treatment strategies. Therefore, we investigated the molecular mechanisms underlying cytarabine (Ara-C) resistance and daunorubicin (DNR) tolerance in Ara-C-resistant RHI-1 cells derived from the wild-type FLT3 AML cell line SHI-1. Quantitative analysis of intracellular drug concentrations, proteomics, and phosphoproteomics showed that DNR resistance in Ara-C-resistant RHI-1 cells is driven by metabolic remodeling toward mitochondrial metabolism, upregulation of DNA repair pathways, and enhanced reactive oxygen species (ROS) detoxification rather than reduced drug uptake. Moreover, targeting these compensatory mechanisms, particularly the OXPHOS complex I proteins, significantly improved the efficacy of both Ara-C and DNR. Conclusively, these findings highlight mitochondrial metabolism and DNA repair as critical factors in chemotherapy resistance and offer valuable insights into potential therapeutic targets for enhancing treatment outcomes in patients with wild-type FLT3 R/R AML.
Collapse
MESH Headings
- Cytarabine/pharmacology
- Cytarabine/therapeutic use
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Mitochondria/metabolism
- Mitochondria/drug effects
- Drug Resistance, Neoplasm/drug effects
- Cell Line, Tumor
- Daunorubicin/pharmacology
- Reactive Oxygen Species/metabolism
- DNA Repair/drug effects
- Treatment Failure
- Oxidative Phosphorylation/drug effects
- fms-Like Tyrosine Kinase 3/metabolism
- fms-Like Tyrosine Kinase 3/genetics
Collapse
Affiliation(s)
- Soo Yeon Chae
- Department of Chemistry, Korea University, Seoul, Republic of Korea
- Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Se-Young Jang
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jinhui Kim
- Department of Chemistry, Korea University, Seoul, Republic of Korea
| | - Sehyun Hwang
- Department of Chemistry, Korea University, Seoul, Republic of Korea
- Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Disha Malani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Olli Kallioniemi
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
- Science for Life Laboratory, Department of Oncology and Pathology, Karolinska Institute, Solna, Sweden
| | - Seung Gyu Yun
- Department of Laboratory Medicine, Korea University College of Medicine, Seoul, Korea.
| | - Jong-Seo Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
- Center for RNA Research, Institute of Basic Science, Seoul National University, Seoul, Korea.
| | - Hugh I Kim
- Department of Chemistry, Korea University, Seoul, Republic of Korea.
- Center for Proteogenome Research, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Wang Q, Qian W, Han Y, Mao Y, Gao Z, Chen Y, Zeng X, Lu H, Jiang L, Li J, Gu N, Qian P. Ferumoxytol promotes haematopoietic stem cell post-injury regeneration as a reactive oxygen species scavenger. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01907-2. [PMID: 40269251 DOI: 10.1038/s41565-025-01907-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 03/17/2025] [Indexed: 04/25/2025]
Abstract
Under stress conditions, such as ex vivo culture, chemotherapy, irradiation and infection, haematopoietic stem cells (HSCs) actively divide to maintain blood cell production. This process leads to production of reactive oxygen species (ROS) that causes HSC exhaustion and haematopoietic failure. Here we show that ferumoxytol (FMT; Feraheme), a Food and Drug Administration-approved nanodrug, is a powerful ROS scavenger capable of relieving ROS in stressed HSCs, facilitating their post-injury regeneration. Mechanistically, the catalase-like activity of FMT reduces intracellular levels of H2O2 and diminishes H2O2-induced cytotoxicity. Moreover, FMT maintains long-term regenerative capacity of transplanted HSCs in pre-conditioned leukaemic mice and shows potential to effectively eliminate leukaemia in vivo while preserving HSCs. Our study highlights FMT as a powerful clinical tool to promote haematopoietic cell recovery in patients undergoing stress-generating treatments.
Collapse
Affiliation(s)
- Qiwei Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Wenchang Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Yingli Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Yu Mao
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhenyue Gao
- Chia-tai Tianqing Pharmaceutical Group (CTTQ Pharma) Co., Ltd., Nanjing, China
| | - Yuxuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xin Zeng
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Huan Lu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Lingli Jiang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Jinxin Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| | - Ning Gu
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
- Medical School of Nanjing University, Nanjing, China.
| | - Pengxu Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, State Key Laboratory of Experimental Hematology, Hangzhou, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China.
| |
Collapse
|
30
|
Machii N, Hatashima R, Niwa T, Taguchi H, Kimirei IA, Mrosso HDJ, Aibara M, Nagasawa T, Nikaido M. Pronounced expression of extracellular matrix proteoglycans regulated by Wnt pathway underlies the parallel evolution of lip hypertrophy in East African cichlids. eLife 2025; 13:RP99160. [PMID: 40259743 PMCID: PMC12014132 DOI: 10.7554/elife.99160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025] Open
Abstract
Cichlid fishes inhabiting the East African Great Lakes, Victoria, Malawi, and Tanganyika, are textbook examples of parallel evolution, as they have acquired similar traits independently in each of the three lakes during the process of adaptive radiation. In particular, 'hypertrophied lip' has been highlighted as a prominent example of parallel evolution. However, the underlying molecular mechanisms remain poorly understood. In this study, we conducted an integrated comparative analysis between the hypertrophied and normal lips of cichlids across three lakes based on histology, proteomics, and transcriptomics. Histological and proteomic analyses revealed that the hypertrophied lips were characterized by enlargement of the proteoglycan-rich layer, in which versican and periostin proteins were abundant. Transcriptome analysis revealed that the expression of extracellular matrix-related genes, including collagens, glycoproteins, and proteoglycans, was higher in hypertrophied lips, regardless of their phylogenetic relationships. In addition, the genes in Wnt signaling pathway, which is involved in promoting proteoglycan expression, was highly expressed in both the juvenile and adult stages of hypertrophied lips. Our comprehensive analyses showed that hypertrophied lips of the three different phylogenetic origins can be explained by similar proteomic and transcriptomic profiles, which may provide important clues into the molecular mechanisms underlying phenotypic parallelisms in East African cichlids.
Collapse
Affiliation(s)
- Nagatoshi Machii
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
| | - Ryo Hatashima
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
| | - Tatsuya Niwa
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of TechnologyYokohamaJapan
| | - Hideki Taguchi
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of TechnologyYokohamaJapan
| | - Ismael A Kimirei
- Tanzania Fisheries Research InstituteDar es SalaamUnited Republic of Tanzania
| | - Hillary DJ Mrosso
- Tanzania Fisheries Research Institute (TAFIRI), Mwanza Fisheries Research CenterMwanzaUnited Republic of Tanzania
| | - Mitsuto Aibara
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
| | - Tatsuki Nagasawa
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
| | - Masato Nikaido
- School of Life Science and Technology, Tokyo Institute of TechnologyTokyoJapan
| |
Collapse
|
31
|
Ogura Y, Sun X, Zhang Z, Kawata K, Wu J, Matsubara R, Ozeki AN, Taniue K, Onoguchi-Mizutani R, Adachi S, Nakayama K, Goda N, Akimitsu N. Fragile X messenger ribonucleoprotein 1 (FMRP) regulates glycolytic gene expression under chronic hypoxia in HCT116 cells. Sci Rep 2025; 15:13273. [PMID: 40246883 PMCID: PMC12006372 DOI: 10.1038/s41598-025-91828-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 02/24/2025] [Indexed: 04/19/2025] Open
Abstract
Oxygen shortage, known as hypoxia, occurs commonly in both physiological and pathological conditions. Transcriptional regulation by hypoxia-inducible factors is a dominant regulatory mechanism controlling hypoxia-responsive genes during acute hypoxia; however, recent studies suggest that post-transcriptional regulation, including RNA degradation, also involves hypoxia-induced gene expression during the chronic hypoxia. In this study, we developed a method to quantify the contributions of RNA synthesis and degradation to differential gene expression, and identified 102 genes mainly regulated via RNA degradation under chronic hypoxia in HCT116 cells. Bioinformatics analysis showed that the genes mainly regulated by RNA degradation were involved in glycolysis. We examined changes in the RNA-binding ability of RNA-binding proteins by RNA interactome capture and statistical analysis using public databases. We identified fragile X messenger ribonucleoprotein 1 (FMRP) as an RNA-binding protein involved in the chronic hypoxia-induced increase in mRNAs encoding rate-limiting enzymes. This study emphasizes the importance of post-transcriptional gene regulation under chronic hypoxia in HCT116 cells.
Collapse
Affiliation(s)
- Yoko Ogura
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Xiaoning Sun
- Advanced Interdisciplinary Studies, Engineering Department, The University of Tokyo, Tokyo, Japan
| | - Zaijun Zhang
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kentaro Kawata
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan.
| | - Jinyu Wu
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ryuma Matsubara
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | | | - Kenzui Taniue
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan
| | | | - Shungo Adachi
- Department of Proteomics, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Koh Nakayama
- Department of Pharmacology, School of Medicine, Asahikawa Medical University, Hokkaido, 078-8510, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, 162-8480, Japan
| | - Nobuyoshi Akimitsu
- Isotope Science Center, The University of Tokyo, Tokyo, 113-0032, Japan.
| |
Collapse
|
32
|
Xu D, Shi J, Jiang S, Meng S, Cheng Z, Wu W, Chang L, Xie Y, Gao Y, Xue Y, Zhang Y. Shotgun and targeted proteomics of Mycolicibacterium smegmatis highlight the role of arginine phosphorylation in the functional adaptation to its environment. J Proteomics 2025; 314:105388. [PMID: 39884554 DOI: 10.1016/j.jprot.2025.105388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/26/2025] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
Although the phosphorylation of serine (S), threonine (T), and tyrosine (Y) is well-established, arginine phosphorylation (pR) has recently garnered significant attention due to its crucial role in bacteria pathogenicity and stress response. Mycolicibacterium smegmatis, a nonpathogenic surrogate of Mycobacterium tuberculosis, serves as a model for studying mycobacterial pathogenesis. A recent proteomics study identified six pR proteins in M. smegmatis. To gain a more comprehensive understanding, we performed pR profiling using mass spectrometry in combination with two distinct phosphopeptide enrichment strategies: titanium-immobilized metal ion affinity chromatography (Ti4+-IMAC) and Fe-NTA cartridge purification. This approach led to the identification of 1192 shared pR peptides with 1553 pR sites in M. smegmatis following both competitive and non-competitive scoring assessments for pR and pS/T/Y. Further stringent filtering through manual verification resulted in 58 high-confident pR sites across 57 proteins. These confirmed pR-proteins are functionally related, particularly in DNA binding and ATP binding. Alterations in the modification of three pR sites during the logarithmic and stationary phases at the phosphorylation level, but not at the total cell protein level, further suggest the role of pR in the bacterium's functional adaptation to its environment. SIGNIFICANCE: Our findings reveal that pR proteins are prevalent and play roles in DNA-binding and ATP-binding activities, providing insights into the broader biological functions of pR peptides in other genetically diverse species. The reliable identification of bacterial pR events in M. smegmatis not only propels the study of pR within the realm of proteomics but also paves the way for exploring its detailed function in bacteria.
Collapse
Affiliation(s)
- Danyang Xu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiahui Shi
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Songhao Jiang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Shuhong Meng
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Zhiyuan Cheng
- School of Mathematical Sciences, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenhui Wu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Lei Chang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Yuping Xie
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China
| | - Yuan Gao
- Central Laboratory of College of Horticulture, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yu Xue
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yao Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Research Unit of Proteomics & Research and Development of New Drug of Chinese Academy of Medical Sciences, Institute of Lifeomics, Beijing 102206, China.
| |
Collapse
|
33
|
Yang F, Ding X, Lv G. Quantitative proteomic analysis based on TMT reveals different responses of Haloxylon ammodendron and Haloxylon persicum to long-term drought. BMC PLANT BIOLOGY 2025; 25:480. [PMID: 40234745 PMCID: PMC11998144 DOI: 10.1186/s12870-025-06513-x] [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: 11/24/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
The essence of the plant drought tolerance mechanism lies in determining protein expression patterns, identifying key drought-tolerant proteins, and elucidating their association with specific functions within metabolic pathways. So far, there is limited information on the long-term drought tolerance of Haloxylon ammodendron and Haloxylon persicum grown in natural environments, as analyzed through proteomics. Therefore, this study conducted proteomic research on H. ammodendron and H. persicum grown in natural environments to identify their long-term drought-tolerant protein expression patterns. Totals of 71 and 348 differentially expressed proteins (DEPs) were identified in H. ammodendron and H. persicum, respectively. Bioinformatics analysis of DEPs reveals that H. ammodendron primarily generates a large amount of energy by overexpressing proteins related to carbohydrate metabolism pathways (pyruvate kinase, purple acid phosphatases and chitinase), and simultaneously encodes proteins capable of degrading misfolded/damaged proteins (tam3-transposase, enhancer of mRNA-decapping protein 4, and proteinase inhibitor I3), thus adapting to long-term drought environments. For H. persicum, most DEPs (enolase and UDP-xylose/xylose synthase) involved in metabolic pathways are up-regulated, indicating that it mainly adapts to long-term drought environments through mechanisms related to positive regulation of protein expression. These results offer crucial insights into how desert plants adapt to arid environments over the long term to maintain internal balance. In addition, the identified key drought-tolerant proteins can serve as candidate proteins for molecular breeding in the genus Haloxylon, aiming to develop new germplasm for desert ecosystem restoration.
Collapse
Affiliation(s)
- Fang Yang
- School of Ecology and Environment, Xinjiang University, Urumqi Xinjiang, 830017, China
- Key Laboratory of Oasis Ecology, Ministry of Education, Urumqi, 830017, China
- Xinjiang Jinghe Observation and Research Station of Temperate Desert Ecosystem, Ministry of Education, Jinghe, 833300, China
| | - Xuelian Ding
- School of Ecology and Environment, Xinjiang University, Urumqi Xinjiang, 830017, China
- Key Laboratory of Oasis Ecology, Ministry of Education, Urumqi, 830017, China
- Xinjiang Jinghe Observation and Research Station of Temperate Desert Ecosystem, Ministry of Education, Jinghe, 833300, China
| | - Guanghui Lv
- School of Ecology and Environment, Xinjiang University, Urumqi Xinjiang, 830017, China.
- Key Laboratory of Oasis Ecology, Ministry of Education, Urumqi, 830017, China.
- Xinjiang Jinghe Observation and Research Station of Temperate Desert Ecosystem, Ministry of Education, Jinghe, 833300, China.
| |
Collapse
|
34
|
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
|
35
|
Custode BM, Annunziata F, Dos Santos Matos F, Schiano V, Maffia V, Lillo M, Colonna R, De Cegli R, Ballabio A, Pastore N. Folliculin depletion results in liver cell damage and cholangiocarcinoma through MiT/TFE activation. Cell Death Differ 2025:10.1038/s41418-025-01486-8. [PMID: 40189703 DOI: 10.1038/s41418-025-01486-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 05/11/2025] Open
Abstract
Mutations in the tumor suppressor gene Folliculin (FLCN) are responsible for Birt-Hogg-Dube' (BHD) syndrome, a rare inherited condition that predisposes affected individuals to skin tumors, pulmonary cysts, and kidney tumors. FLCN regulates key cellular pathways, including TFEB, TFE3, and mTORC1, which are critical for maintaining cell homeostasis. Loss of FLCN leads to both hyperactivation of mTORC1 and constitutive activation of TFEB and TFE3, contributing to tumorigenesis. While previous studies showed that Flcn liver-specific conditional knockout (FlcnLiKO) mice are protected from developing liver fibrosis and damage upon high-fat diet exposure, the potential role of FLCN loss in liver carcinogenesis remained unexplored. Here, we demonstrate that hepatic loss of FLCN in mice results in cancer associated with inflammation and fibrosis with features of cholangiocarcinoma (CCA). This phenotype emerges in mice over 90-week-old, with a male predominance. Moreover, FlcnLiKO mice are more prone to develop diethylnitrosamine (DEN)- or 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)- induced liver tumors with heterogenous histological features. Notably, depletion of TFE3, but not TFEB, in the liver of FlcnLiKO mice fully rescues the cancer phenotype and normalized mTORC1 signaling, highlighting TFE3 as the primary driver of liver cancer and mTORC1 hyperactivity in the absence of FLCN.
Collapse
Affiliation(s)
| | | | | | - Valentina Schiano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Veronica Maffia
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Milena Lillo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Rita Colonna
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, Italy
| | - Nunzia Pastore
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy.
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, Italy.
| |
Collapse
|
36
|
Pérez Malla CU, Kalla J, Tiefenbacher A, Wasinger G, Kluge K, Egger G, Sheibani-Tezerji R. Goistrat: gene-of-interest-based sample stratification for the evaluation of functional differences. BMC Bioinformatics 2025; 26:97. [PMID: 40188042 PMCID: PMC11971790 DOI: 10.1186/s12859-025-06109-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
PURPOSE Understanding the impact of gene expression in pathological processes, such as carcinogenesis, is crucial for understanding the biology of cancer and advancing personalised medicine. Yet, current methods lack biologically-informed-omics approaches to stratify cancer patients effectively, limiting our ability to dissect the underlying molecular mechanisms. RESULTS To address this gap, we present a novel workflow for the stratification and further analysis of multi-omics samples with matched RNA-Seq data that relies on MSigDB curated gene sets, graph machine learning and ensemble clustering. We compared the performance of our workflow in the top 8 TCGA datasets and showed its clear superiority in separating samples for the study of biological differences. We also applied our workflow to analyse nearly a thousand prostate cancer samples, focusing on the varying expression of the FOLH1 gene, and identified specific pathways such as the PI3K-AKT-mTOR gene sets as well as signatures linked to prostate tumour aggressiveness. CONCLUSION Our comprehensive approach provides a novel tool to identify disease-relevant functions of genes of interest (GOI) in large datasets. This integrated approach offers a valuable framework for understanding the role of the expression variation of a GOI in complex diseases and for informing on targeted therapeutic strategies.
Collapse
Affiliation(s)
- Carlos Uziel Pérez Malla
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Jessica Kalla
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Andreas Tiefenbacher
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Gabriel Wasinger
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Kilian Kluge
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Raheleh Sheibani-Tezerji
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.
| |
Collapse
|
37
|
Allard JB, Sharma S, Patel R, Sanderford M, Tamura K, Vucetic S, Gerhard GS, Kumar S. Evolutionary sparse learning reveals the shared genetic basis of convergent traits. Nat Commun 2025; 16:3217. [PMID: 40185716 PMCID: PMC11971283 DOI: 10.1038/s41467-025-58428-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Cases abound in which nearly identical traits have appeared in distant species facing similar environments. These unmistakable examples of adaptive evolution offer opportunities to gain insight into their genetic origins and mechanisms through comparative analyses. Here, we present an approach to build genetic models that underlie the independent origins of convergent traits using evolutionary sparse learning with paired species contrast (ESL-PSC). We tested the hypothesis that common genes and sites are involved in the convergent evolution of two key traits: C4 photosynthesis in grasses and echolocation in mammals. Genetic models were highly predictive of independent cases of convergent evolution of C4 photosynthesis. Genes contributing to genetic models for echolocation were highly enriched for functional categories related to hearing, sound perception, and deafness, a pattern that has eluded previous efforts applying standard molecular evolutionary approaches. These results support the involvement of sequence substitutions at common genetic loci in the evolution of convergent traits. Benchmarking on empirical and simulated datasets showed that ESL-PSC could be more sensitive in proteome-scale analyses to detect genes with convergent molecular evolution associated with the acquisition of convergent traits. We conclude that phylogeny-informed machine learning naturally excludes apparent molecular convergences due to shared species history, enhances the signal-to-noise ratio for detecting molecular convergence, and empowers the discovery of common genetic bases of trait convergences.
Collapse
Affiliation(s)
- John B Allard
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA
- Department of Biology, Temple University, Philadelphia, PA, USA
| | - Sudip Sharma
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA
- Department of Biology, Temple University, Philadelphia, PA, USA
| | - Ravi Patel
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA
- Department of Biology, Temple University, Philadelphia, PA, USA
| | - Maxwell Sanderford
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA
- Department of Biology, Temple University, Philadelphia, PA, USA
| | - Koichiro Tamura
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo, Japan
- Research Center for Genomics and Bioinformatics, Tokyo Metropolitan University, Tokyo, Japan
| | - Slobodan Vucetic
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA
| | - Glenn S Gerhard
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA.
| | - Sudhir Kumar
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.
- Department of Biology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
38
|
Song Y, Liu Q, Ma J, Mabrouk I, Zhou Y, Li X, Xue G, Ma X, Xu J, Wang J, Pan H, Hua G, Cao H, Hu J, Sun Y. Integrated omics reveals the regulatory role of PKCα in Sertoli cell proliferation and apoptosis through the MAPK/ERK signaling pathway in goose testis. Poult Sci 2025; 104:105123. [PMID: 40209467 PMCID: PMC12008624 DOI: 10.1016/j.psj.2025.105123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Testicular development is essential for reproductive performance in geese, as the testes are the primary organs for sperm production and play a pivotal role in egg-laying physiology. Despite their importance, genes, proteins, and pathways regulating goose testicular development are poorly understood. This study employed integrative transcriptomic and proteomic analysis methods to identify critical regulators of testicular development in geese across three reproductive periods. Additionally, the role of PKCα in Sertoli cell proliferation via the MAPK/ERK pathway was evaluated at the cellular level. A total of 8,921 differentially expressed genes and 1,866 differentially expressed proteins were identified, revealing key pathways such as FOXO, MAPK, PPAR, and Hedgehog that regulate testicular development. Both omics correlation analysis and signal pathway regulation network results show the importance of MAPK in this process, while cellular experiment revealed that PKCα affects proliferation and apoptosis of Sertoli cells through the MAPK/ERK signaling pathway. The findings revealed that PKCα downregulation reduced the expression of genes associated with both cell proliferation and apoptosis, resulting in a diminished activity of Sertoli cells. This study compared testicular transcriptomes and proteomes of Hungarian and Jilin white geese, identifying key genes, proteins, and pathways critical for reproduction. These findings advance our understanding of molecular mechanisms underlying testicular development and provide insights to enhance gander reproductive performance.
Collapse
Affiliation(s)
- Yupu Song
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiuyuan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingyun Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Ichraf Mabrouk
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yuxuan Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xinyue Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Guizhen Xue
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xiaoming Ma
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jing Xu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingbo Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Hongxiao Pan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Guoqing Hua
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Heng Cao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jingtao Hu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Yongfeng Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Jilin Agricultural University, Ministry of Education, Changchun, 130118, China; Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
39
|
Yao T, Kobayashi H, Hirai T, Tokuoka Y, Tokoro M, Asayama Y, Suzuki Y, Hatano Y, Ikeda H, Sugimura S, Yamamoto T, Yamada TG, Hosoi Y, Funahashi A, Fukunaga N, Asada Y, Kurimoto K, Yamagata K. Zinc eluted from glassware is a risk factor for embryo development in human and animal assisted reproduction†. Biol Reprod 2025:ioaf050. [PMID: 40169168 DOI: 10.1093/biolre/ioaf050] [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: 09/23/2024] [Revised: 01/16/2025] [Indexed: 04/03/2025] Open
Abstract
In assisted reproduction, many factors in the culture environment, including light, temperature, pH, and culture media, can reduce preimplantation embryo viability. Laboratory glassware is also a known risk factor for in vitro embryos; however, the underlying mechanisms that disrupt embryonic development remain unclear. We identified Zn eluted from glassware as an embryotoxic substance. In mouse embryos, Zn induced delayed development, abnormalities in chromosome segregation, cytokinesis, zygotic gene activation (e.g. Zscan4a and murine endogenous retrovirus with leucine, also known as MERVL), and aberrantly upregulated developmental gene expression (e.g. Hoxa1, Hoxb9, T, and Fgf8) that could be mediated through metal regulatory transcription factors (e.g. Mtf1). Subsequently, Zn exposure led to significantly reduced blastocyst formation. Post-implantation, Zn-exposed embryos were associated with normal birth rates, however, the birth weight increased by an average of 18% compared with embryos cultured without Zn. Furthermore, Zn exposure affected the development of bovine and human embryos, with species-based variation in the strength and timing of these effects. To mitigate these embryotoxic effects, we identified a method to prevent glass toxicity using chelating agents. This research not only highlights the importance of risk control in embryo culture but also facilitates the development of safe and effective methods for assisted reproduction.
Collapse
Affiliation(s)
- Tatsuma Yao
- Research and Development Center, Fuso Pharmaceutical Industries, Ltd., Joto-ku, Osaka 536-8523, Japan
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
| | - Hisato Kobayashi
- Department of Embryology, Nara Medical University, Kashihara, Nara 634-0813, Japan
| | - Tatsuki Hirai
- Research and Development Center, Fuso Pharmaceutical Industries, Ltd., Joto-ku, Osaka 536-8523, Japan
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
| | - Yuta Tokuoka
- Center for Biosciences and Informatics, Graduate School of Fundamental Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Mikiko Tokoro
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
- Asada Institute for Reproductive Medicine, Asada Ladies Clinic, Nagoya, Aichi 450-0002, Japan
| | - Yuta Asayama
- Research and Development Center, Fuso Pharmaceutical Industries, Ltd., Joto-ku, Osaka 536-8523, Japan
| | - Yuka Suzuki
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
| | - Yu Hatano
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
| | - Hiroki Ikeda
- Department of Embryology, Nara Medical University, Kashihara, Nara 634-0813, Japan
| | - Satoshi Sugimura
- Department of Biological Production, Tokyo University of Agriculture and Technology, Fuchu, Tokyo 183-8509, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takahiro G Yamada
- Center for Biosciences and Informatics, Graduate School of Fundamental Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
- Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Yoshihiko Hosoi
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
| | - Akira Funahashi
- Center for Biosciences and Informatics, Graduate School of Fundamental Science and Technology, Keio University, Yokohama, Kanagawa 223-8522, Japan
- Department of Biosciences and Informatics, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Noritaka Fukunaga
- Asada Institute for Reproductive Medicine, Asada Ladies Clinic, Nagoya, Aichi 450-0002, Japan
| | - Yoshimasa Asada
- Asada Institute for Reproductive Medicine, Asada Ladies Clinic, Nagoya, Aichi 450-0002, Japan
| | - Kazuki Kurimoto
- Department of Embryology, Nara Medical University, Kashihara, Nara 634-0813, Japan
- Advanced Medical Research Center, Nara Medical University, Kashihara, Nara 634-0813, Japan
| | - Kazuo Yamagata
- Graduate School of Biology-Oriented Science and Technology, Kindai University, Kinokawa, Wakayama 649-6493, Japan
| |
Collapse
|
40
|
Gao F, Li J. Identification of ferroptosis-related gene signatures in temporal lobe epilepsy with hippocampal sclerosis. Front Neurosci 2025; 19:1530182. [PMID: 40242460 PMCID: PMC11999976 DOI: 10.3389/fnins.2025.1530182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025] Open
Abstract
Background Ferroptosis is a form of regulated cell death that damages neurons in the central nervous system. In this study, we aimed to construct ferroptosis-related gene signatures in temporal lobe epilepsy with hippocampal sclerosis (TLE-HS) and explore their diagnostic role in TLE-HS. Methods The GSE205661 dataset was acquired for training purposes, while the GSE71058 was obtained to serve as the validation dataset. Subsequently, ferroptosis-related differentially expressed genes (FR-DEGs) in TLE-HS were further analyzed. We used weighed gene co-expression network analysis (WGCNA) algorithm, single-factor logistic regression analysis, and LASSO algorithm to screen characteristic FR-DEGs. Then, the receiver operating characteristic (ROC) was used to evaluate the value of these characteristic genes in disease diagnosis. Finally, a long non-coding RNA (lncRNA)-microRNA (miRNA)-messenger RNA (mRNA) network was constructed. Results We identified 141 FR-DEGs in TLE-HS, and these genes were enriched in T-cell activation involved in immune response and signaling pathways related to lipids and atherosclerosis. Further WGCNA was performed to select 47 overlapping FR-DEGs, which were significantly enriched in 13 biological processes and 14 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including the negative regulation of apoptotic process and ferroptosis. Four genes, namely PDK4, SMPD1, GPT2, and METTL14, were identified as signature genes in TLE-HS. Moreover, the ROC derived from the four genes in GSE205661 and GSE71058 for predicting TLE-HS had an area under the curve (AUC) of 0.988 and 0.929, respectively. Furthermore, the lncRNA-miRNA-mRNA network constructed from the 4 FR-DEGs consisted of 5 lncRNAs and 14 miRNAs. The signatures based on four FR-DEGs were found to be a strong predictor of TLE-HS, and they may represent valuable therapeutic targets for TLE-HS.
Collapse
Affiliation(s)
| | - Jinzi Li
- Department of Pediatrics, Yanbian University Hospital, Yanji, China
| |
Collapse
|
41
|
Coombes JD, Manka PP, Swiderska-Syn M, Vannan DT, Riva A, Claridge LC, Moylan C, Suzuki A, Briones-Orta MA, Younis R, Kitamura N, Sydor S, Bittencourt S, Mi Z, Kuo PC, Diehl AM, van Grunsven LA, Chokshi S, Canbay A, Abdelmalek MF, Aspichueta P, Papa S, Eksteen B, Syn WK. Osteopontin Promotes Cholangiocyte Secretion of Chemokines to Support Macrophage Recruitment and Fibrosis in MASH. Liver Int 2025; 45:e16131. [PMID: 39422353 PMCID: PMC11893260 DOI: 10.1111/liv.16131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND AND AIMS Osteopontin (OPN) promotes the ductular reaction and is a major driver of chronic liver disease (CLD) progression. Although CLD is characterised by the accumulation of inflammatory cells including macrophages around the peri-portal regions, the influence of OPN on recruitment is unclear. We investigated the role of OPN in cholangiocyte chemokine production and macrophage recruitment by combining in vivo, in vitro, and in silico approaches. METHODS The effects of OPN on cholangiocyte chemokine production and macrophage migration were assessed in culture, alongside RNA-sequencing to identify genes and pathways affected by OPN depletion. Murine liver injury models were used to assess liver chemokine expression and liver macrophage/monocyte recruitment. OPN and chemokine expression were analysed in liver tissue and plasma from biopsy-proven metabolic dysfunction-associated alcoholic steatohepatitis (MASH) patients. RESULTS OPN-knockdown in cholangiocytes reduced chemokine secretion. RNA-sequencing showed OPN-related effects clustered around immunity, chemotaxis and chemokine production. Macrophage exposure to cholangiocyte-conditioned media showed OPN-supported migration via chemokines chemokine (C-C motif) ligand (CCL)2, CCL5 and chemokine (C-X-C motif) ligand (CXCL)1. These effects were related to NF-κB signalling. Murine liver fibrosis was accompanied by upregulated liver OPN, CCL2, CCL5 and CXCL1 mRNA, and accumulation of liver cluster of differentiation (CD)11b/F4/80+CC chemokine receptors (CCR2)high macrophages but treatment with OPN-specific neutralising aptamers reduced fibrosis, chemokine mRNAs and accumulation of liver CD11b/F4/80+CCR2high/lymphocyte antigen 6 complexhigh inflammatory monocytes. In human MASH, liver OPN correlated with chemokines CCL2 and IL8 in association with portal injury and fibrosis. Plasma OPN, serum CCL2 and IL8 also increased with fibrosis stage. CONCLUSIONS OPN promotes cholangiocyte chemokine secretion and the accumulation of pro-inflammatory monocytes. These data support neutralisation of OPN as an anti-inflammatory and anti-fibrotic strategy.
Collapse
Affiliation(s)
- Jason D. Coombes
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Division of Gastroenterology and Hepatology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Paul P Manka
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Gastroenterology and Hepatology, University Clinic Bochum, Bochum, Germany
| | - Marzena Swiderska-Syn
- Division of Gastroenterology and Hepatology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Danielle T Vannan
- Snyder Institute for Chronic Diseases, University of Calgary, Alberta, Canada
- Aspen Woods Clinic, Calgary, Alberta, Canada
| | - Antonio Riva
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Viral Hepatitis and Alcohol Research Group, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
| | - Lee C Claridge
- Department of Hepatology, Leeds Teaching Hospital NHS Trust, UK
| | - Cynthia Moylan
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Ayako Suzuki
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Marco A Briones-Orta
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Rasha Younis
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
| | - Naoto Kitamura
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
| | - Svenja Sydor
- Gastroenterology and Hepatology, University Clinic Bochum, Bochum, Germany
| | | | - Zhiyong Mi
- Department of Surgery, University of South Florida, Tampa, Florida
| | - Paul C. Kuo
- Department of Surgery, University of South Florida, Tampa, Florida
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | | | - Shilpa Chokshi
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Viral Hepatitis and Alcohol Research Group, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
| | - Ali Canbay
- Gastroenterology and Hepatology, University Clinic Bochum, Bochum, Germany
| | - Manal F. Abdelmalek
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, EPV/EHU, Leioa
| | - Salvatore Papa
- Leeds Institute of Medical Research, St. James’s University Hospital, University of Leeds, Leeds, UK
| | - Bertus Eksteen
- Snyder Institute for Chronic Diseases, University of Calgary, Alberta, Canada
- Aspen Woods Clinic, Calgary, Alberta, Canada
| | - Wing-Kin Syn
- Regeneration and Repair, Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, EPV/EHU, Leioa
- Division of Gastroenterology and Hepatology, School of Medicine, Saint Louis University, Saint Louis, Missouri, USA
| |
Collapse
|
42
|
Lee T, Kim CJ, Lim DH, Lee YS. microRNA miR-315-5p regulates developmental growth in Drosophila wings by targeting S6k. INSECT SCIENCE 2025. [PMID: 40166978 DOI: 10.1111/1744-7917.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 04/02/2025]
Abstract
Tissue growth in Drosophila is regulated by various factors, with microRNAs (miRNAs) emerging as key players over the past decade. However, the precise roles of miRNAs in growth regulation remain incompletely understood. In this study, we explored the biological role of miR-315 in wing growth regulation. Inhibition of miR-315-5p activity using a miR-315 sponge led to an increase in wing size, whereas its overexpression resulted in reduced wing size, primarily through a decrease in wing cell size. We identified ribosomal protein kinase p-70-S6k (S6k) as a target of miR-315-5p in relation to wing growth control. Overexpression of miR-315 reduced both total S6k and phosphorylated S6k protein levels in Drosophila S2 cells and wing discs. Additionally, a luciferase reporter assay confirmed that miR-315-5p directly binds to the 3'-untranslated region of S6k. Consistently, RNAi-mediated depletion of S6k led to smaller wings, primarily due to a reduction in cell size. Notably, co-overexpression of active S6k rescued the wing defects caused by miR-315 overexpression. Overall, these findings demonstrate that miR-315 regulates wing growth by suppressing S6k expression.
Collapse
Affiliation(s)
- Taeheon Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Chae Jeong Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Do-Hwan Lim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Young Sik Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Mattson E, Warner GR. Single-cell RNA-seq reveals that granulosa cells are a target of phthalate toxicity in the ovary. Toxicol Sci 2025; 204:169-180. [PMID: 39752319 PMCID: PMC11939077 DOI: 10.1093/toxsci/kfaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Phthalates are known endocrine-disrupting chemicals and ovarian toxicants that are used widely in consumer products. Phthalates have been shown to exert ovarian toxicity on multiple endpoints, altering transcription of genes responsible for normal ovarian function. However, the molecular mechanisms by which phthalates act on the ovary are not well understood. In this study, we hypothesized that phthalates specifically target granulosa cells within the ovarian follicle. To test our hypothesis, we cultured whole mouse antral follicles for 96 h in the presence of vehicle or 10 µg/ml of a phthalate metabolite mixture. At the end of the culture period, follicles were dissociated into single-cell suspensions and subjected to single-cell RNA-sequencing. We used markers from published studies to identify cell-type clusters, the largest of which were granulosa and theca/stroma cells. We further identified subpopulations of granulosa, theca, and stromal cells and analyzed differentially expressed genes between the phthalate treatment and control. Granulosa cells, specifically mural granulosa cells, had the most differentially expressed genes. Pathway analysis of differentially expressed genes from the overall granulosa cell cluster revealed disruption of cell cycle and mitosis, whereas pathway analysis of the mural granulosa cell subcluster identified terms related to translation, ribosome, and endoplasmic reticulum. Our findings suggest that phthalates have both broad impacts on cell types and specific impacts on cellular subtypes, emphasizing the complexity of phthalate toxicity and highlighting how bulk sequencing can mask effects on vulnerable cell types.
Collapse
Affiliation(s)
- Erik Mattson
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| | - Genoa R Warner
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, Newark, NJ 07103, United States
| |
Collapse
|
44
|
Nowak AL, Saadat N, Sun J, Forsman AM, Liang X, Joyce C, Woo J, Engeland CG, Misra DP, Giurgescu C, Zhang W, Anderson CM. Preterm Birth in African American Women: A Multi-Omic Pilot Study in Early Pregnancy. Biol Res Nurs 2025; 27:205-215. [PMID: 39440846 DOI: 10.1177/10998004241275049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Preterm birth (PTB; <37 weeks completed gestation) is a devastating problem affecting over 13 million live births worldwide. In the U.S., African Americans experience significantly higher rates of PTB compared to non-Hispanic Whites. PTB disparities have been linked to social determinants of health (e.g., socioeconomic status, discrimination). However, the biological underpinnings related to these associations are unclear. DNA methylation (DNAm) is subject to environmental influences, and DNAm modifications are known to affect gene expression. Using a multi-omic approach, we examined differences in combined DNA methylation (DNAm) and messenger RNA (mRNA) transcriptomic data from 20 pregnant African American women (12 PTB; 8 term birth) early in pregnancy (8-18 weeks gestation). We found that the HLA-DQB2 gene was both differentially methylated (cg12296550; p = .02) and differentially expressed (p = .014; log2FC = 2.5) between women with PTB and term birth. Gene expression analysis showed HLA-DQB2 and HLA-DRB4 (p = .028; log2FC = -3.6) were the two most highly expressed genes. HLA-DQB2 expressed higher in PTB and HLA-DRB4 expressed higher in term birth. However, no genes remained significant (p < .05) after Bonferroni correction. HLA-DRB4 and AKR1C1 were identified as a potential biomarkers in dimensionality reduction models and are also important to immune function and allogenic breakdown. Altered gene expression may lead to inflammatory imbalances or allogenic intolerance resulting in PTB. This study provides proof-of-concept evidence for the feasibility and importance of future multi-omics studies with larger populations to further explore the genes and pathways identified here.
Collapse
Affiliation(s)
- Alexandra L Nowak
- Marcella Niehoff School of Nursing, Loyola University at Chicago, Maywood, IL, USA
| | - Nadia Saadat
- Department of Paediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Jiao Sun
- Department of Computer Science, University of Central Florida, Orlando, FL, USA
| | - Anna M Forsman
- Department of Biology, Colby College, Waterville, ME, USA
| | - Xiaoyu Liang
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Cara Joyce
- Biostatistics Core, Department of Medicine, Center for Translational Research and Education, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Jennifer Woo
- University of Texas at Arlington, Arlington, TX, USA
| | - Christopher G Engeland
- Biobehavioral Health, College of Health and Human Development, Ross and Carol Ness College of Nursing, The Pennsylvania State University, University Park, PA, USA
| | - Dawn P Misra
- Department of Epidemiology and Biostatistics, MSU College of Human Medicine, East Lansing, MI, USA
| | - Carmen Giurgescu
- Chatlos Foundation Endowed Chair in Nursing, University of Central Florida College of Nursing, Orlando, FL, USA
| | - Wei Zhang
- Department of Computer Science, University of Central Florida, Orlando, FL, USA
| | - Cindy M Anderson
- Maternal Infant Health, Martha S. Pitzer Center for Women, Children and Youth, The Ohio State University College of Nursing, Columbus, OH, USA
| |
Collapse
|
45
|
Yin A, Xu Y, Su X, Wang R, Zhang Z, Chen Y, Han L, Fu G, Wang W, Wang J. EFTUD2 is a promising diagnostic and prognostic indicator involved in the tumor immune microenvironment and glycolysis of lung adenocarcinoma. Front Oncol 2025; 15:1499217. [PMID: 40236649 PMCID: PMC11996642 DOI: 10.3389/fonc.2025.1499217] [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: 09/20/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Background Elongation Factor Tu GTP Binding Domain Containing 2 (EFTUD2), a conserved spliceosomal GTPase, is involved in craniofacial development and various cancers, but its role in lung adenocarcinoma (LUAD) remains unclear. Methods EFTUD2 expression in LUAD tissues was analyzed using data from TCGA and GEO, and validated by immunohistochemistry, RT-qPCR, and Western blotting. The relationship between EFTUD2 expression and clinical features was examined using Fisher's exact test. Diagnostic and prognostic analyses were performed in R. Hub genes related to EFTUD2 were identified through topological algorithms, and immune infiltration was assessed using CIBERSORT. The cGAS-STING pathway and m6A modification were also analyzed in the TCGA LUAD cohort. Functional assays were conducted to assess EFTUD2's impact on LUAD cell proliferation, cell cycle, invasion, and metastasis, while glycolytic enzyme levels were measured by Western blotting. Results EFTUD2 was upregulated in LUAD tissues and cells, correlating with N classification, visceral pleural invasion, intravascular tumor embolism, and cytokeratin-19 fragment antigen 21-1. Sixteen EFTUD2-related hub genes were identified. Higher EFTUD2 expression was linked to altered immune cell infiltration, with increased TumorPurity scores and decreased StromalScore, ImmuneScore, and ESTIMATEScore values. Gene enrichment analyses highlighted EFTUD2's involvement in cell adhesion, immune response. EFTUD2 was strongly associated with the cGAS-STING pathway and m6A modification. EFTUD2 knockdown inhibited LUAD cell proliferation, migration, and tumorigenicity, causing G0/G1 phase cell cycle arrest, and altered glycolytic enzyme expression. These findings may suggest that EFTUD2 positively regulates the progression of LUAD and modulates the glycolytic activity of tumor cells, making it valuable for LUAD treatment and prognosis. Conclusions EFTUD2 is a potential diagnostic and prognostic marker for LUAD, associated with immune infiltration, the tumor microenvironment, the cGAS-STING pathway, m6A modification, and glycolysis.
Collapse
Affiliation(s)
- Ankang Yin
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yufan Xu
- Department of Pathology, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiyang Su
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Runan Wang
- Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zebin Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yi Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lu Han
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Guoxiang Fu
- Department of Pathology, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Juan Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
46
|
Wang Z, Kim S, Farrell BD, de Medeiros BAS. Customizable PCR-based target enrichment probes for sequencing fungi-parasitized insects. INSECT SCIENCE 2025; 32:719-724. [PMID: 39034422 DOI: 10.1111/1744-7917.13413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024]
Affiliation(s)
- Zhengyang Wang
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
- College of Life Sciences, Sichuan University, Chengdu, China
| | - Sangil Kim
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
- Research Institute of Basic Sciences, Seoul National University, Seoul, Republic of Korea
| | - Brian D Farrell
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
| | - Bruno A S de Medeiros
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts
- Field Museum of Natural History, Chicago, Illinois, United States
| |
Collapse
|
47
|
Nickchi P, Vadadokhau U, Mirzaie M, Baumann M, Saei AA, Jafari M. Monitoring Functional Posttranslational Modifications Using a Data-Driven Proteome Informatic Pipeline. Proteomics 2025; 25:e202400238. [PMID: 40100226 PMCID: PMC12019915 DOI: 10.1002/pmic.202400238] [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: 07/10/2024] [Revised: 01/20/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
Posttranslational modifications (PTMs) are of significant interest in molecular biomedicine due to their crucial role in signal transduction across various cellular and organismal processes. Characterizing PTMs, distinguishing between functional and inert modifications, quantifying their occupancies, and understanding PTM crosstalk are challenging tasks in any biosystem. Studying each PTM often requires a specific, labor-intensive experimental design. Here, we present a PTM-centric proteome informatic pipeline for predicting relevant PTMs in mass spectrometry-based proteomics data without prior information. Once predicted, these in silico identified PTMs can be incorporated into a refined database search and compared to measured data. As a practical application, we demonstrate how this pipeline can be used to study glycoproteomics in oral squamous cell carcinoma based on the proteome profile of primary tumors. Subsequently, we experimentally identified cellular proteins that are differentially expressed in cells treated with multikinase inhibitors dasatinib and staurosporine using mass spectrometry-based proteomics. Computational enrichment analysis was then employed to determine the potential PTMs of differentially expressed proteins induced by both drugs. Finally, we conducted an additional round of database search with the predicted PTMs. Our pipeline successfully analyzed the enriched PTMs, and detected proteins not identified in the initial search. Our findings support the effectiveness of PTM-centric searching of MS data in proteomics based on computational enrichment analysis, and we propose integrating this approach into future proteomics search engines.
Collapse
Affiliation(s)
- Payman Nickchi
- Department of StatisticsUniversity of British ColumbiaVancouver, Biritish ColumbiaCanada
| | - Uladzislau Vadadokhau
- MedicumDepartment of Biochemistry and Developmental BiologyMeilahti Clinical Proteomics Core FacilityUniversity of HelsinkiHelsinkiFinland
| | - Mehdi Mirzaie
- Department of PharmacologyFaculty of Medicine & Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | - Marc Baumann
- MedicumDepartment of Biochemistry and Developmental BiologyMeilahti Clinical Proteomics Core FacilityUniversity of HelsinkiHelsinkiFinland
| | - Amir A. Saei
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Mohieddin Jafari
- MedicumDepartment of Biochemistry and Developmental BiologyMeilahti Clinical Proteomics Core FacilityUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
48
|
Javed MJ, Howard RM, Li H, Carrasco L, Dirain ML, Su G, Cai G, Upchurch GR, Jiang Z. GSDMD Deficiency Attenuates the Development of Ascending Aortic Dissections in a Novel Mouse Model. Arterioscler Thromb Vasc Biol 2025; 45:541-556. [PMID: 39945067 PMCID: PMC11945581 DOI: 10.1161/atvbaha.124.321740] [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] [Accepted: 01/27/2025] [Indexed: 02/19/2025]
Abstract
BACKGROUND Mechanisms driving the development of type A aortic dissection (TAD) are currently poorly understood, and animal models of spontaneous TAD are limited. In the present study, we developed a novel mouse TAD model and evaluated the role of GSDMD (gasdermin D) in TAD development. METHODS TADs were created by treating the ascending aorta of adult C57BL/6J mice with Act E (active elastase) and β-aminopropionitrile. The temporal progress of the TAD pathology was rigorously characterized by histological evaluation and scanning electron microscopy, while potential mechanisms were explored using bulk RNA sequencing of specimens collected at multiple time points. With this novel TAD model, we conducted additional experiments to investigate the impact of GSDMD deficiency on TAD formation. RESULTS Ascending aortas challenged with Act E and β-aminopropionitrile developed pathology featuring the early onset of intimomedial tears (complete penetration) and intramural hematomas, followed by progressive medial loss and aortic dilation. Ingenuity pathway analysis and functional annotation of differentially expressed genes suggested that a unique inflammatory microenvironment, rather than general inflammation, promotes the onset of TADs by specifically recruiting neutrophils to the aortic wall. At later stages, T cell-mediated immune injury emerged as the primary driver of pathology. Gsdmd deficiency attenuated medial loss, adventitial fibrosis, and dilation of TADs. This protective effect correlated with a reduced cell death and decreased T-cell infiltration in TADs. Notably, cleaved GSDMD was detected in human TADs but was absent in healthy aortas. CONCLUSIONS A novel mouse TAD model was developed, specifically targeting the ascending aorta. This model generates a unique microenvironment that activates specific immune cell subsets, driving the onset and subsequent remodeling of TADs. Consistently, Gsdmd deficiency mitigates TAD development, likely by modulating cell death and T-cell responses. This model provides a valuable tool for studying immune injury mechanisms in TAD pathogenesis.
Collapse
Affiliation(s)
- Muhammad J. Javed
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Rachael M. Howard
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Hua Li
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Laura Carrasco
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Marvin L.S Dirain
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Gang Su
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Guoshuai Cai
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Gilbert R. Upchurch
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
| | - Zhihua Jiang
- Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, FL 32601, United States; Division of Thoracic and Cardiovascular Surgery, University of Florida College of Medicine, Gainesville, FL 32601, United States
- Malcom Randll VA Medical Center, Gainesville, FL 32608
| |
Collapse
|
49
|
Fernández-Pisonero I, Lorenzo-Martín LF, Drosten M, Santos E, Barbacid M, Alarcón B, Bustelo XR. Active R-RAS2/TC21 prevents cell cycle arrest and morphological alterations in mouse embryonic fibroblasts lacking RAS proteins. Oncogene 2025:10.1038/s41388-025-03367-3. [PMID: 40164870 DOI: 10.1038/s41388-025-03367-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
R-RAS2/TC21, a member of the R-RAS subfamily of GTP-binding proteins, shares structural and signaling properties with the RAS subfamily proteins H-, K-, and N-RAS. However, little information is available regarding its role in normal cells and the level of functional redundancy with R-RAS and classical RAS proteins. In this work, we used loss and gain-of-function approaches to assess these issues in mouse embryonic fibroblasts (MEFs). Using primary MEFs from Rras2-/-, Rras-/- or Rras-/-; Rras2-/- embryos, we show here that endogenous R-RAS2/TC21 is required for activation of the phosphatidylinositol 3 kinase (PI3K)-AKT axis, the proliferation, and the adhesion properties of these cells. Endogenous R-RAS does not influence any of these cell parameters. We also show that the depletion of R-RAS2/TC21 worsens the proliferative and morphological defects elicited by the combined loss of H-, K- and N-RAS proteins in MEFs. Conversely, the ectopic expression of an active version of R-RAS2/TC21, but not of R-RAS, overcomes such defects. This rescue activity involves the inhibition of the tumor suppressor TP53 and is PI3K-, mTORC-, and MEK/ERK-dependent. These results indicate that R-RAS2/TC21, R-RAS, and RAS subfamily GTPases play different roles in MEFs. They also show that R-RAS2 provides subsidiary signals that are essential for the short-term proliferation and long-term viability of MEFs lacking RAS signaling.
Collapse
Affiliation(s)
- Isabel Fernández-Pisonero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mattias Drosten
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Eugenio Santos
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Mariano Barbacid
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Balbino Alarcón
- Centro de Biología Molecular Severo Ochoa, CSIC and Universidad Autónoma de Madrid, Madrid, Spain
| | - Xosé R Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and Universidad de Salamanca, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
50
|
Slone S, Anthony SR, Green LC, Parkins S, Acharya P, Kasprovic DA, Reynolds K, Jaggers RM, Nieman ML, Alam P, Wu X, Roy S, Aubé J, Xu L, Li Z, Lorenz JN, Owens AP, Kanisicak O, Tranter M. HuR inhibition reduces post-ischemic cardiac remodeling by dampening myocyte-dependent inflammatory gene expression and the innate immune response. FASEB J 2025; 39:e70433. [PMID: 40085190 PMCID: PMC11908633 DOI: 10.1096/fj.202400532rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/16/2025]
Abstract
The RNA-binding protein human antigen R (HuR) has been shown to reduce cardiac remodeling following both myocardial infarction and cardiac pressure overload, but the full extent of the HuR-dependent mechanisms within cells of the myocardium has yet to be elucidated. Wild-type mice were subjected to 30 min of cardiac ischemia (via LAD occlusion) and treated with a novel small molecule inhibitor of HuR at the time of reperfusion, followed by direct in vivo assessment of cardiac structure and function. Direct assessment of HuR-dependent mechanisms was done in vitro using neonatal rat ventricular myocytes (NRVMs) and bone marrow-derived macrophages (BMDMs). HuR activity is increased within 2 h after ischemia/reperfusion (I/R) and is necessary for early post-I/R inflammatory gene expression in the myocardium. Despite an early reduction in inflammatory gene expression, HuR inhibition has no effect on initial infarct size at 24 h post-I/R. However, pathological remodeling is reduced with preserved cardiac function at 2 weeks post-I/R upon HuR inhibition. RNA sequencing analysis of gene expression in NRVMs treated with LPS to model damage-associated molecular pattern (DAMP)-mediated activation of toll-like receptors (TLRs) demonstrates a HuR-dependent regulation of pro-inflammatory chemokine and cytokine gene expression in cardiomyocytes. Importantly, we show that conditioned media transfer from NRVMs pre-treated with HuR inhibitor loses the ability to induce inflammatory gene expression and M1-like polarization in bone marrow-derived macrophages (BMDMs) compared to NRVMs treated with LPS alone. Functionally, HuR inhibition reduces macrophage infiltration to the post-ischemic myocardium in vivo. Additionally, we show that LPS-treated NRVMs induce the migration of peripheral blood monocytes in a HuR-dependent endocrine manner. These studies demonstrate that HuR is necessary for early pro-inflammatory gene expression in cardiomyocytes following I/R injury that subsequently mediates monocyte recruitment and macrophage activation in the post-ischemic myocardium.
Collapse
Grants
- HL166326 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 20POST35200267 American Heart Association Postdoctoral Fellowship
- PRE35210795 American Heart Association (AHA)
- HL125204 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL132111 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 1029875 American Heart Association (AHA)
- R01 HL166326 NHLBI NIH HHS
- CA252158 HHS | NIH | National Cancer Institute (NCI)
- HL148598 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- CDA34110117 American Heart Association (AHA)
- CA243445 HHS | NIH | National Cancer Institute (NCI)
- R01 HL132111 NHLBI NIH HHS
- R01 CA243445 NCI NIH HHS
- CA191785 HHS | NIH | National Cancer Institute (NCI)
- 23CDA1052132 American Heart Association Career Development Grant
- F31-HL170636 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- PRE35230020 American Heart Association (AHA)
- R33 CA252158 NCI NIH HHS
- HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HHS | NIH | National Cancer Institute (NCI)
- American Heart Association (AHA)
Collapse
Affiliation(s)
- Samuel Slone
- Division of Cardiovascular Health and DiseaseUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Department of Pharmacology and Systems PhysiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Sarah R. Anthony
- Department of Molecular Medicine and TherapeuticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Lisa C. Green
- Division of Cardiovascular Health and DiseaseUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Department of Pharmacology and Systems PhysiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Sharon Parkins
- Division of Cardiovascular Health and DiseaseUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Department of Molecular Medicine and TherapeuticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Pooja Acharya
- Department of Molecular Medicine and TherapeuticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Daniel A. Kasprovic
- Department of Molecular Medicine and TherapeuticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Kelsi Reynolds
- Pelotonia Institute for Immuno‐OncologyThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Robert M. Jaggers
- Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Division of Basic and Translational Science, Department of Emergency MedicineThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Michelle L. Nieman
- Department of Pharmacology and Systems PhysiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Perwez Alam
- Department of Biomedical Sciences, College of Veterinary MedicineUniversity of MissouriColumbiaMissouriUSA
| | - Xiaoqing Wu
- Department of Molecular BiosciencesUniversity of KansasLawrenceKansasUSA
| | - Sudeshna Roy
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of PharmacyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of PharmacyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Liang Xu
- Department of Molecular BiosciencesUniversity of KansasLawrenceKansasUSA
| | - Zihai Li
- Pelotonia Institute for Immuno‐OncologyThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - John N. Lorenz
- Department of Pharmacology and Systems PhysiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - A. Phillip Owens
- Division of Cardiovascular Health and DiseaseUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Onur Kanisicak
- Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Division of Basic and Translational Science, Department of Emergency MedicineThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Michael Tranter
- Department of Molecular Medicine and TherapeuticsThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| |
Collapse
|