1
|
Zeng S, Liu N, Zhang A, Duan N, Xu B, Ai C. Molecular basis identification and hypnotic drug interactions for cognitive impairment related to sleep deprivation. BMC Psychiatry 2025; 25:371. [PMID: 40229714 PMCID: PMC11995581 DOI: 10.1186/s12888-024-06395-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/09/2024] [Indexed: 04/16/2025] Open
Abstract
Chronic sleep deprivation can lead to cognitive impairment which makes it difficult to think, focus, and make comprehensive decisions. This in turn leads to the progression and increased risk of several diseases. This study aimed to explore potential drug targets and biomarkers underlying the increased disease risk due to sleep deprivation, including stress responses, immune dysfunction, and metabolic dysregulation. Four datasets namely GSE40562, GSE98566, GSE98582 for sleep deprivation, and GSE26576 normal brain cells were utilized to understand the molecular basis and potential drug targets associated with sleep deprivation. The GEO2R tool, Robust rank aggregations, and Venny were used to retrieve the common DEGs. Functional gene and pathway analyses were carried out via GO and the KEGG analyses. The STRING and CytoHuba plugins were utilized to identify the protein-protein interactions (PPIs) as well as the hub genes in the main PPI subnetworks following the drug interaction of the hub genes and GEPIA-based survival analysis of the DEGs. A total of 160 common DEGs were retrieved from all four datasets. Among them, 65 were down-regulated and 95 were up-regulated. TOP2A, AURKB, NEFL, CDC42, ASPM, GAP43, PVALB, NUF2, CALM1, TPR, KIF5B, KIF15, TROAP, NDC80, PBK, MKI67, SST, AHSP, ALAS2, and NEFH were retrieved as hub genes. While based on drug interaction, survival analysis and gene expression profile eight hub gene named TOP2A, AURKB, PVALB, CALM1, KIF5B, PBK, MKI67, and SST were found to be potential drug candidates and significantly correlated with infiltration levels of CD8 + T cells, B cells, macrophages, CD4 + T cells, neutrophils, and dendritic cells. These genes might play a role in sleep disorders via various pathways associated with neurodegeneration and diseases, potentially serving as biomarkers to support treatment and diagnosis.
Collapse
Affiliation(s)
- Shun Zeng
- Department of Mental Health Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Nannan Liu
- Department of Sleep Disorders Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Andong Zhang
- College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Na Duan
- Obstetrical Department, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Bo Xu
- College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China
| | - Chunqi Ai
- Department of Mental Health Center, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China.
- College of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, Hubei Province, China.
| |
Collapse
|
2
|
Yang B, Wang C, Chen X, Zhai H, Wu Y, Cui M, Wu J, Li W, Hua B. In silico and animal studies on the anti-cancer mechanisms of Shaoyao Decoction against colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119444. [PMID: 39929402 DOI: 10.1016/j.jep.2025.119444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE It is well known that Shaoyao Decoction (SYD), as a commonly used formula of traditional Chinese medicine (TCM), has a beneficial effect on the treatment of ulcerative colitis (UC). It is found that SYD can also prevent colitis-associated colorectal cancer (CAC). However, its potential anti-cancer mechanism is still waiting to be revealed. AIM OF THE STUDY The aim of this study is to investigate the underlying mechanisms of SYD in inhibiting CAC through silico analysis as well as animal experiment validation. MATERIALS AND METHODS The primary active compounds, potential therapeutic targets and intervening signaling pathways, which SYD might inhibit the CAC process were predicted by network pharmacology analysis combined with our previous research result of high performance liquid chromatography (HPLC). We attempted to validate the acquired hub targets from molecular docking combined with the Gene Expression Profiling Interactive Analysis (GEPIA), the Human Protein Atlas (HPA), and the cBioPortal database comprehensively. Subsequently, an animal model of CAC mice induced by azoxymethane (AOM) and dextran sulfate sodium (DSS) was constructed and treated with SYD for 14 weeks, and tumor-related physical indicators were evaluated after sacrificed. In addition, samples of colon tissues were obtained for histologic and protein level studies to verify the predicted mechanism. RESULTS We obtained 166 active ingredients of SYD and predicted 148 potential targets through network pharmacology analysis, among which quercetin, berberine, kaempferol, wogonin and naringenin were selected as core drug ingredients, and TP53, AKT1, CASP3, PTGS2 and CCND1 were identified and included into the range of core targets. GO and KEGG analyses suggested that the PI3K-Akt signaling pathway might hold a crucial role in CAC prevention and treatment by promoting apoptosis and inhibiting tumor proliferation. In the animal experiment, both SYD and SASP treatments improved the inflammatory condition and pathological damage of the colon tissues in mice. After treatments with SYD and SASP, it was found that decreases of Cyclin D1 and Survivin expression levels and increases of p53 and Cleaved caspase-3 expression levels could be mediated by decreasing the phosphorylation levels of PI3K and Akt proteins in the colon tissues of mice. CONCLUSION The results of our study provide supports that SYD effectively inhibits CAC based on modulating PI3K-Akt signaling pathway to suppress tumor proliferation process as well as to promote tumor apoptosis process.
Collapse
Affiliation(s)
- Bingwei Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chenglei Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xue Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haoyu Zhai
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ying Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Muyao Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiahe Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Weidong Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
3
|
Hernandez I, Botana L, Diez-Mata J, Tesoro L, Jimenez-Guirado B, Gonzalez-Cucharero C, Alcharani N, Zamorano JL, Saura M, Zaragoza C. CAP1: a novel extracellular vesicle marker linked to endothelial senescence in atherosclerosis. Biol Direct 2025; 20:46. [PMID: 40189560 PMCID: PMC11974053 DOI: 10.1186/s13062-025-00646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/29/2025] [Indexed: 04/09/2025] Open
Abstract
Endothelial senescence (ES) contributes to aging-related disorders and triggers a senescence-associated secretory-pattern (SASP), releasing Extracellular Vesicles (EVs), potentially impacting atherosclerosis. We used EVs from young (8 weeks) and aged (24 months) ApoE-knockout mice to detect ES in human aortic (HAEC) and coronary (CAEC) endothelial cells. Age-related atherosclerosis was confirmed by increased atheroma plaque formation in aged compared to young ApoE-knockout mice fed a high-fat diet, and the contribution of EVs from aged ApoE-knockout mice on ES was evidenced by a replicative senescence assay in cultured HAEC and CAEC, starting with the promotion of ES. A proteomic analysis depicted the recently PCSK9-associated CAP1 protein as a cargo component in EVs from aged animals and highly expressed in mouse and human endarterectomy plaques. Gene silencing of CAP1 inhibited HAEC and CAEC ES while overexpressing CAP1 in these cells restored the senescent-phenotype. The in vivo contribution of CAP1 was assessed by injecting CAP1-containing EVs isolated from aged ApoE-knockout mice into wild-type (WT) mice fed either a regular or high-fat diet. Compared to the EVs from young mice, the CAP1-containing EVs led to a pronounced ES along with the formation of intraluminal atheroma plaques. Similarly, young ApoE-knockout mice developed thickened and calcified atheroma plaques, along with increased ß-Gal-positive aortic staining when injected with EVs isolated from aged ApoE-knockout mice, like the atheroma plaques observed in aged ApoE-knockout animals. In conclusion, early molecular targets of ES may contribute to better management of atherosclerosis, in which here we unveiled CAP1 as a new molecular target.
Collapse
Affiliation(s)
- Ignacio Hernandez
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, Madrid, Spain
| | - Laura Botana
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Madrid, Spain
| | - Javier Diez-Mata
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain
| | - Laura Tesoro
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain
- Facultad de Medicina, Universidad Francisco de Vitoria, Madrid, Spain
| | - Beatriz Jimenez-Guirado
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain
| | - Claudia Gonzalez-Cucharero
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain
| | - Nunzio Alcharani
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain
| | - Jose Luis Zamorano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Departamento de Cardiología, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain
| | - Marta Saura
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Unidad de Fisiología, Departamento de Biologia de Sistemas, Universidad de Alcalá (IRYCIS), Alcala de Henares, Madrid, Spain
| | - Carlos Zaragoza
- Unidad Mixta de Investigación Cardiovascular Universidad Francisco de Vitoria Hospital Ramon y Cajal (IRYCIS), Madrid, Spain.
- Facultad de Medicina, Universidad Francisco de Vitoria, Madrid, Spain.
| |
Collapse
|
4
|
Boulaki V, Efthimiopoulos S, Moschonas NK, Spyrou GΜ. Exploring potential key genes and disease mechanisms in early-onset genetic epilepsy via integrated bioinformatics analysis. Neurobiol Dis 2025; 210:106888. [PMID: 40180227 DOI: 10.1016/j.nbd.2025.106888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/22/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
Epilepsy is a severe common neurological disease affecting all ages. Epilepsy with onset before the age of 5 years, designated early-onset epilepsy (EOE), is of special importance. According to previous studies, genetic factors contribute significantly to the pathogenesis of EOE that remains unclear and must be explored. So, a list of 229 well-selected EOE-associated genes expressed in the brain was created for the investigation of genetic factors and molecular mechanisms involved in its pathogenesis. Enrichment analysis showed that among significant pathways were nicotine addiction, GABAergic synapse, synaptic vesicle cycle, regulation of membrane potential, cholinergic synapse, dopaminergic synapse, and morphine addiction. Performing an integrated analysis as well as protein-protein interaction network-based approaches with the use of GO, KEGG, ClueGO, cytoHubba and 3 network metrics, 12 hub genes were identified, seven of which, CDKL5, GABRA1, KCNQ2, KCNQ3, SCN1A, SCN8A and STXBP1, were identified as key genes (via Venn diagram analysis). These key genes are mostly enriched in SNARE interactions in vesicular transport, regulation of membrane potential and synaptic vesicle exocytosis. Clustering analysis of the PPI network via MCODE showed significant functional modules, indicating also other pathways such as N-Glycan biosynthesis and protein N-linked glycosylation, retrograde endocannabinoid signaling, mTOR signaling and aminoacyl-tRNA biosynthesis. Drug-gene interaction analysis identified a number of drugs as potential medications for EOE, among which the non-FDA approved drugs azetukalner (under clinical development), indiplon and ICA-105665 and the FDA approved drugs retigabine, ganaxolone and methohexital.
Collapse
Affiliation(s)
- Vasiliki Boulaki
- Division of Animal and Human Physiology, Department of Biology, National & Kapodistrian University of Athens, Panepistimiopolis, Ilisia 15784, Greece
| | - Spiros Efthimiopoulos
- Division of Animal and Human Physiology, Department of Biology, National & Kapodistrian University of Athens, Panepistimiopolis, Ilisia 15784, Greece
| | - Nicholas K Moschonas
- Department of General Biology, School of Medicine, University of Patras, Patras 26500, Greece; Metabolic Engineering &Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology Hellas (FORTH/ICE-HT), Patras, Greece
| | - George Μ Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology & Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus.
| |
Collapse
|
5
|
Cortés-Hernández LE, Eslami-S Z, Attina A, Batista S, Cayrefourcq L, Vialeret J, Di Vizio D, Hirtz C, Costa-Silva B, Alix-Panabières C. Proteomic profiling and functional analysis of extracellular vesicles from metastasis-competent circulating tumor cells in colon cancer. J Exp Clin Cancer Res 2025; 44:102. [PMID: 40119417 PMCID: PMC11929255 DOI: 10.1186/s13046-025-03360-4] [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: 01/23/2025] [Accepted: 03/09/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are pivotal in cancer progression, and in vitro CTC models are crucial for understanding their biological mechanisms. This study focused on the characterization of extracellular vesicles (EVs) from CTC lines derived from a patient with metastatic colorectal cancer (mCRC) at different stages of progression who progressed despite therapy (thus mirroring the clonal evolution of cancer). METHODS AND RESULTS Morphological and size analyses revealed variations among EVs derived from different CTC lines. Compared with the Vesiclepedia database, proteomic profiling of these EVs revealed enrichment of proteins related to stemness, endosomal biogenesis, and mCRC prognosis. Integrin family proteins were significantly enriched in EVs from CTC lines derived after therapy failure. The role of these EVs in cancer progression was analyzed by assessing their in vivo distribution, particularly in the liver, lungs, kidneys, and bones. EVs accumulate significantly in the liver, followed by the lungs, kidneys and femurs. CONCLUSIONS This study is a pioneering effort in highlighting therapy progression-associated changes in EVs from mCRC patients via an in vitro CTC model. The results offer insights into the role of metastasis initiator CTC-derived EVs in cancer spread, suggesting their utility for studying cancer tissue distribution mechanisms. However, these findings must be confirmed and extended to patients with mCRC. This work underscores the potential of CTC-derived EVs as tools for understanding cancer dissemination.
Collapse
Affiliation(s)
- Luis Enrique Cortés-Hernández
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France.
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Aurore Attina
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Silvia Batista
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Jérôme Vialeret
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Dolores Di Vizio
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christophe Hirtz
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
| |
Collapse
|
6
|
Siddiqui AJ, Elkahoui S, Alshammari AM, Patel M, Ghoniem AEM, Abdalla RAH, Dwivedi-Agnihotri H, Badraoui R, Adnan M. Mechanistic Insights into the Anticancer Potential of Asparagus racemosus Willd. Against Triple-Negative Breast Cancer: A Network Pharmacology and Experimental Validation Study. Pharmaceuticals (Basel) 2025; 18:433. [PMID: 40143209 PMCID: PMC11944961 DOI: 10.3390/ph18030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: The present study investigated the anticancer potential of Asparagus racemosus Willd. against triple-negative breast cancer (TNBC) using a combined in silico and in vitro approach. Methods: Network pharmacology identified 115 potential targets shared between A. racemosus phytochemicals and TNBC, highlighting key cancer-related pathways. Molecular docking predicted strong binding affinities between specific phytochemicals (beta-sitosterol, quercetin, and others) and crucial TNBC targets, including AKT1 and ERBB2. Results: Molecular dynamics simulations validated these interactions, demonstrating stable complex formation. In vitro, A. racemosus crude extracts exhibited potent anticancer activity against MDA-MB-231 TNBC cells, showing a dose-dependent reduction in viability (IC50 = 90.44 μg/mL), induction of G1 phase cell cycle arrest, and significant early apoptosis. Conclusions: These integrated findings provide compelling evidence for the anticancer potential of A. racemosus against TNBC, suggesting its promise for further development as a therapeutic strategy.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (S.E.); (A.M.A.); (A.E.M.G.); (R.B.); (M.A.)
| | - Salem Elkahoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (S.E.); (A.M.A.); (A.E.M.G.); (R.B.); (M.A.)
| | - Ahmed Mohajja Alshammari
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (S.E.); (A.M.A.); (A.E.M.G.); (R.B.); (M.A.)
| | - Mitesh Patel
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India;
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| | - Ahmed Eisa Mahmoud Ghoniem
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (S.E.); (A.M.A.); (A.E.M.G.); (R.B.); (M.A.)
| | - Randa Abdeen Husien Abdalla
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia;
| | | | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (S.E.); (A.M.A.); (A.E.M.G.); (R.B.); (M.A.)
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (S.E.); (A.M.A.); (A.E.M.G.); (R.B.); (M.A.)
| |
Collapse
|
7
|
Sima N, Ayllon-Hermida A, Fernández-Becerra C, del Portillo HA. Extracellular vesicles in malaria: proteomics insights, in vitro and in vivo studies indicate the need for transitioning to natural human infections. mBio 2025; 16:e0230424. [PMID: 39868784 PMCID: PMC11898581 DOI: 10.1128/mbio.02304-24] [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: 01/28/2025] Open
Abstract
Globally, an estimated 2.1 billion malaria cases and 11.7 million malaria deaths were averted in the period 2000-2022. Noticeably, despite effective control measurements, in 2022 there were an estimated 249 million malaria cases in 85 malaria-endemic countries and an increase of 5 million cases compared with 2021. Further understanding the biology, epidemiology, and pathogenesis of human malaria is therefore essential for achieving malaria elimination. Extracellular vesicles (EVs) are membrane-enclosed nanoparticles pivotal in intercellular communication and secreted by all cell types. Here, we will review what is currently known about EVs in malaria, from biogenesis and cargo to molecular insights of pathophysiology. Of relevance, a meta-analysis of proteomics cargo, and comparisons between in vitro and in vivo human studies revealed striking differences with those few studies reported from patients. Thus, indicating the need for rigor standardization of methodologies and for transitioning to human infections to elucidate their physiological role. We conclude with a focus on translational aspects in diagnosis and vaccine development and highlight key gaps in the knowledge of EVs in malaria research.
Collapse
Affiliation(s)
- Núria Sima
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
| | - Alberto Ayllon-Hermida
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
- School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Carmen Fernández-Becerra
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
- CIBERINFEC, ISCIII-CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Hernando A. del Portillo
- ISGlobal, Barcelona, Spain
- IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
8
|
Galande S, Ravikanth VV, Tokala RK, Satyanarayana Singh S, Rao GV, Talukdar R, Peddapulla C, Reddy DN, Sasikala M. Transcriptomic analysis of pancreatic tissue from humans and mice identifies potential gene signatures and unexplored pathways during progression from acute to chronic pancreatitis. Gene 2025; 940:149200. [PMID: 39732348 DOI: 10.1016/j.gene.2024.149200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND A comprehensive understanding of the molecular pathogenesis of chronic pancreatitis (CP), a fibroinflammatory disorder of the pancreas, is warranted for the development of targeted therapies. The current study focused on comparing the transcriptomes of pancreatic tissues obtained from patients with CP with those of two rodent models of chemically induced CP to identify dysregulated genes/signaling pathways. METHODS Pancreatitis was induced in mice using cerulein and L-arginine. Pancreatic tissues were obtained from humans and mice. The RNA was isolated, and the transcriptomes were generated using the GeneChip Human Transcriptome Array 2.0 and Clariom D Mouse Array respectively. Differentially expressed genes with log2-fold changes ≥ +2 and ≤ -2 were considered for functional and signaling pathway enrichment analysis. The expression of NUCB2, which plays a role in β-cell function, was validated by ELISA in acute pancreatitis (AP) and immune cell responses in AP and CP using flow cytometry. RESULTS The current study identifies L-arginine-induced CP as a better model for investigating the pathogenesis of human CP, with greater similarity in dysregulated genes (22%), transcription factors (34%) and enriched pathways (58%) compared to cerulein model (2%, 11% and 9%) respectively. Nesfatin-1, encoded by NUCB2, was decreased in patients with AP (12% nondiabetic, 41% post pancreatitis diabetes). The Th1 immune cell response was greater in the patients with AP (44%), whereas Th17 immune response was greater in patients with CP (18%). CONCLUSION Our study highlights potential novel and unexplored pathways involved in inflammation, fibrosis, and pain in CP and paves the way for testing them as putative drug targets using a severe disease model.
Collapse
Affiliation(s)
- Sheethal Galande
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | - V V Ravikanth
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | - Ranjeet K Tokala
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India
| | | | - G V Rao
- Department of Surgical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Rupjyoti Talukdar
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Chandan Peddapulla
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - D Nageshwar Reddy
- Department of Medical Gastroenterology, AIG Hospitals, Gachibowli, Hyderabad, India
| | - Mitnala Sasikala
- Translational Research Centre, Asian Healthcare Foundation, AIG Hospitals, Hyderabad, India.
| |
Collapse
|
9
|
Parsons M, O'Connell K, Szklanna P, Weiss L, Kenny M, Donnelly A, Norris J, Babyuk Y, O'Donoghue L, Ní Áinle F, McGuigan C, Maguire PB. Characterisation of Platelet Releasate Proteome in Relapsing-Remitting Multiple Sclerosis Reveals Dysregulation of Inflammatory Signalling and Extracellular Vesicle Dynamics. Proteomics Clin Appl 2025; 19:e202400019. [PMID: 39831369 DOI: 10.1002/prca.202400019] [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: 01/22/2025]
Abstract
PURPOSE Multiple Sclerosis is an inflammatory neurodegenerative disease characterised by blood-brain barrier dysfunction and leukocyte infiltration into the CNS. Platelets are best known for their contributions to haemostasis, however, upon activation, platelets release an abundance of soluble and vesicular-associated proteins, termed the platelet releasate (PR). This milieu contains numerous inflammatory and vasoactive proteins, that can attract leukocytes and alter endothelial permeability. EXPERIMENTAL DESIGN We aimed to characterise the PR of Relapsing-Remitting multiple sclerosis (RRMS) patients, previously characterized regarding thrombin generation dynamics compared to healthy controls. We carried out LFQ proteomic profiling of the PR from 15 RRMS and 19 aged-matched healthy controls. RESULTS We identified 9 proteins increased and 16 proteins decreased in the PR of RRMS patients. Platelet/endothelial cell-adhesion molecule-1 (PECAM-1) was uniquely found in healthy control PR and circulating levels of PECAM-1 were significantly lower in RRMS patient samples. GO analysis revealed a strong link between altered proteins and extracellular vesicles (EVs). Small EV levels were significantly reduced in RRMS PR compared to healthy PR and showed a negative correlation with PECAM-1 levels in RRMS plasma. CONCLUSIONS AND CLINICAL RELEVANCE Our findings suggest that platelet reactivity may be linked to disease activity, even in periods of disease remission.
Collapse
Affiliation(s)
- Martin Parsons
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Karen O'Connell
- Department of Neurology, Tallaght, University Hospital, Dublin, Ireland
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Paulina Szklanna
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Luisa Weiss
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Martin Kenny
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Aisling Donnelly
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Jessica Norris
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Yuri Babyuk
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Lorna O'Donoghue
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Fionnuala Ní Áinle
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- Department of Haematology, Rotunda Hospital, Dublin, Ireland
- Department of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Christopher McGuigan
- School of Medicine, University College Dublin, Dublin, Ireland
- Department of Neurology, St. Vincent's University Hospital, Dublin, Ireland
| | - Patricia B Maguire
- SPHERE Research Group, Conway Institute, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Institute for Discovery, O'Brien Centre for Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Sultana A, Alam MS, Khanam A, Liang H. Unraveling the molecular landscape of non-small cell lung cancer: Integrating bioinformatics and statistical approaches to identify biomarkers and drug repurposing. Comput Biol Med 2025; 187:109744. [PMID: 39914199 DOI: 10.1016/j.compbiomed.2025.109744] [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: 08/28/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/21/2025]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most malignant tumors and the leading cause of death from cancer worldwide and is increasing at a massive rate every year. Most NSCLC patients are diagnosed at advanced stages, which is associated with a poor prognosis and a very low 5-year survival rate. Therefore, the purpose of this study is to investigate molecular markers for early diagnosis, prognosis and therapy of NSCLC through the integration of bioinformatics and statistical methods. A total of 93 overlapping differentially expressed genes (oDEGs) were identified between NSCLC and normal samples through Linear Models for Microarray (LIMMA) and Significance Analysis of Microarrays (SAM) methods. Six top-degree oDEGs (CCNA2, CDC6, AURKA, CCNB1, MKI67, and PRC1) were identified as key genes (KGs) through the protein-protein interaction (PPI) network. The predictive accuracy analysis of the identified KGs revealed an accuracy of 96.92 %, with a sensitivity of 91.73 % and a specificity of 98.15 %. KGs associated with 3 molecular functions (MFs), 5 cellular components (CCs), 3 biological processes (BPs), and 4 pathways were identified through FunRich software. Analysis of expression levels using the UALCAN database revealed that KGs are significantly associated with potential early diagnostic biomarkers. Survival analysis using the GEPIA database demonstrated that the KGs possessed strong prognostic power for NSCLC. Finally, seven repurposed candidate drugs ENTRECTINIB, SORAFENIB, CHEMBL1765740, TOZASERTIB, NERVIANO, AZD-1152-HQPA, and SELICICLIB were proposed through molecular docking analysis. In conclusion, the findings of this study have the potential to significantly impact the early diagnosis, prognosis, and treatment of NSCLC.
Collapse
Affiliation(s)
- Adiba Sultana
- Medical Big Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510080, China
| | - Md Shahin Alam
- Medical Big Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510080, China; Laboratory of Molecular Neuropathology, Department of Pharmacology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, China.
| | - Alima Khanam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Huiying Liang
- Medical Big Data Center, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, 510080, China.
| |
Collapse
|
11
|
Khanal S, Kumar P, da Silva MB, Singh R, Suassuna N, Jones DC, Davis RF, Chee PW. Time-course RNA-seq analysis of upland cotton (Gossypium hirsutum L.) responses to Southern root-knot nematode (Meloidogyne incognita) during compatible and incompatible interactions. BMC Genomics 2025; 26:183. [PMID: 39994510 PMCID: PMC11849305 DOI: 10.1186/s12864-025-11339-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND The Southern root-knot nematode (Meloidogyne incognita) poses a substantial threat to cotton (Gossypium hirsutum L.) by causing significant agricultural losses. Host plant resistance is the most plausible approach for minimizing these losses. QTL mapping and early transcriptomic studies have identified candidate genes within the QTL regions on chromosome 11 (qMi-C11) and chromosome 14 (qMi-C14). Although these QTL regions have been fine-mapped and candidate genes identified, expression profiling of Meloidogyne-Gossypium interactions across different stages of infection could further refine the list of candidate genes. This study advances our understanding of the molecular mechanisms underlying the resistance conferred by qMi-C11 and qMi-C14 against Southern root-knot nematode. RESULTS Using time-course RNA-seq analyses across nematode developmental phases, we uncovered transcriptomic events-both genome-wide and within QTL intervals-underlying defense responses during compatible interactions (with Cocker 201, a susceptible line) and incompatible interactions (with M-120 RNR, a resistant line). Basal defense responses were observed in both compatible and incompatible interactions, with stronger expression in the incompatible interaction. Nematode-responsive genes associated with defense pathways showed distinct dynamics, characterized by repression during compatible interactions and early induction, greater diversity, and heightened upregulation during incompatible interactions. This study uncovers a broad repertoire of disease resistance and putative resistance genes, as well as pathogenesis-related genes, ligands, and receptors, that are differentially expressed in response to nematode parasitism. Mapping of these genes across the cotton genome identified promising candidates, including Gh_A11G3090 (PUB21) and Gh_A11G2836 (RPPL1) within the chromosome 11 QTL region, andGh_D02G0257 (RLP12) and Gh_D02G0259 (RLP12) within the chromosome 14 QTL region. CONCLUSIONS The findings of this study deepen our understanding of host-nematode interactions, identify candidate genes for downstream applications, and contribute to advancements in resistance breeding and sustainable nematode management strategies.
Collapse
Affiliation(s)
- Sameer Khanal
- Department of Crop and Soil Sciences and Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, 2356 Rainwater Road, Tifton, GA, 31793, USA.
| | - Pawan Kumar
- Department of Crop and Soil Sciences and Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, 2356 Rainwater Road, Tifton, GA, 31793, USA
- Bayer Crop Science Division, 700 W. Chesterfield Pkwy W, Chesterfield, MO, 63017, USA
| | - Mychele B da Silva
- Department of Crop and Soil Sciences and Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, 2356 Rainwater Road, Tifton, GA, 31793, USA
- Tidewater AREC, Virginia Tech, 6321 Holland Road, Suffolk, VA, 23437, USA
| | - Rippy Singh
- Department of Crop and Soil Sciences and Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, 2356 Rainwater Road, Tifton, GA, 31793, USA
- Bayer Crop Science Division, 700 W. Chesterfield Pkwy W, Chesterfield, MO, 63017, USA
| | - Nelson Suassuna
- Department of Crop and Soil Sciences and Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, 2356 Rainwater Road, Tifton, GA, 31793, USA
- Empresa Brasileira de Pesquisa Agropecuária (Embrapa), Santo Antônio de Goiás, GO, Brazil
| | - Don C Jones
- Cotton Incorporated, 6399 Weston Parkway, Cary, NC, 27513, USA
| | - Richard F Davis
- U.S. Department of Agriculture - Agricultural Research Service, 115 Coastal Way, Tifton, GA, 31793, USA
| | - Peng W Chee
- Department of Crop and Soil Sciences and Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, 2356 Rainwater Road, Tifton, GA, 31793, USA.
| |
Collapse
|
12
|
Lago-Baameiro N, Camino T, Vazquez-Durán A, Sueiro A, Couto I, Santos F, Baltar J, Falcón-Pérez JM, Pardo M. Intra and inter-organ communication through extracellular vesicles in obesity: functional role of obesesomes and steatosomes. J Transl Med 2025; 23:207. [PMID: 39979938 PMCID: PMC11844161 DOI: 10.1186/s12967-024-06024-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/22/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) represent a sophisticated mechanism of intercellular communication that is implicated in health and disease. Specifically, the role of EVs in metabolic regulation and their implications in metabolic pathologies, such as obesity and its comorbidities, remain unclear. METHODS Extracellular vesicles (EVs) were isolated through serial ultracentrifugation from murine adipocytes treated with palmitate or oleic acid, whole visceral and subcutaneous adipose tissue (obesesomes) of bariatric surgery obese donors, and human hepatocytes under steatosis (steatosomes) for functional in vitro experiments. Functional effects on inflammation and glucose and lipid metabolism of target cells (human and murine macrophages and hepatocytes) were assessed using ELISA, RT-PCR, and immunodetection. Isolated EVs from human steatotic (steatosomes) and control hepatocytes (hepatosomes) were characterized for quantity, size, and tetraspanin profile by NTA and Single Particle Interferometric Reflectance Imaging Sensor (SP-IRIS), and their protein cargo analyzed by qualitative (DDA) and quantitative (DIA-SWATH) proteomics using LC-MS/MS. Proteins identified by proteomics were validated by capturing EVs on functionalized chips by SP-IRIS. RESULTS AND CONCLUSIONS In this study, we investigated the role of EVs in the local communication between obese adipocytes and immune cells within adipose tissue, and the interaction of steatotic and healthy hepatocytes in the context of fatty liver disease progression. Furthermore, we analyzed obese adipose tissue-to-liver interactions through EV-obesesomes to elucidate their role in obesity-associated hepatic metabolic dysregulation. Our findings reveal that obesesomes promote inflammation and the secretion of pro-inflammatory cytokines upon interaction with macrophages, exerting a significant impact on reducing insulin resistance and altering lipid and glucose metabolism upon interaction with hepatocytes; in both cases, EVs from palmitate-loaded adipocytes and obesesomes from human visceral adipose depots demonstrated the most deleterious effect. Additionally, EVs secreted by steatotic hepatocytes (steatosomes) induced insulin resistance and altered lipid and glucose metabolism in healthy hepatocytes, suggesting their involvement in MASLD development. Proteomic analysis of steatosomes revealed that these vesicles contain liver disease-associated proteins, rendering them significant repositories of real-time biomarkers for the early stages and progression of MASLD.
Collapse
Affiliation(s)
- N Lago-Baameiro
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| | - T Camino
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| | - A Vazquez-Durán
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
| | - A Sueiro
- Grupo Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - I Couto
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
- Servicio de Cirugía Plástica y Reparadora, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - F Santos
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
- Servicio de Cirugía General, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - J Baltar
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain
- Servicio de Cirugía General, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Spain
| | - J M Falcón-Pérez
- Exosomes Laboratory and Metabolomics Platform, CIC bioGUNE-BRTA, CIBERehd, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - M Pardo
- Grupo Obesidómica, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, A Coruña, Spain.
- CIBER Fisiopatología Obesidad y Nutrición, Instituto de Salud Carlos III, Santiago de Compostela, Spain.
| |
Collapse
|
13
|
Woo HK, Ahn HS, Park J, Bae J, Kim B, Yu J, Seo JK, Kim K, Cho YK. Size-Dependent Separation of Extracellular Vesicle Subtypes with Exodisc Enabling Proteomic Analysis in Prostate Cancer. J Proteome Res 2025; 24:861-870. [PMID: 39873726 DOI: 10.1021/acs.jproteome.4c00945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Extracellular vesicles (EVs) are emerging as crucial biomarkers in cancer diagnostics and therapeutics with their heterogeneity presenting both challenges and opportunities in prostate cancer research. However, existing methods for isolating and characterizing EV subtypes have been limited by inefficient separation and inadequate proteomic analysis. Here we show an optimized centrifugal microfluidic device, Exodisc, that efficiently isolates large quantities of EV subtypes from particle-enriched medium, enabling comprehensive proteomic analysis of small (EV-S, 20-200 nm) and large (EV-L, >200 nm) EVs. Using this device, we successfully separated EV-S and EV-L from prostate cancer cell lines LNCaP and PC3. Mass spectrometry-based proteomics revealed that EV proteins reflect parental cell characteristics more than EV size, with EV-L demonstrating increased expression of PSMA-correlated proteins. Our optimized protocol addresses challenges in EV isolation and characterization, providing a more effective method for studying cellular and molecular mechanisms of specific EV subtypes. This study extends the potential use of EVs as a liquid biopsy for cancer theranostics, paving the way for more precise isolation of EV subtypes and potentially leading to improved biomarker discovery and the development of personalized treatments.
Collapse
Affiliation(s)
- Hyun-Kyung Woo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Hee-Sung Ahn
- AMC Sciences, Asan Medical Center, Seoul 05505, Republic of Korea
- Clinical Proteomics Core Lab, Convergence Research Center, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Juhee Park
- Center for Algorithmic and Robotic Synthesis, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Jonghoon Bae
- UNIST Central Research Facility (UCRF), UNIST, Ulsan 44919, Republic of Korea
| | - Bokyung Kim
- Department of Digital Medicine, BK21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Jiyoung Yu
- Clinical Proteomics Core Lab, Convergence Research Center, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Jeong Kon Seo
- UNIST Central Research Facility (UCRF), UNIST, Ulsan 44919, Republic of Korea
| | - Kyunggon Kim
- Clinical Proteomics Core Lab, Convergence Research Center, Asan Medical Center, Seoul 05505, Republic of Korea
- Department of Digital Medicine, BK21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| |
Collapse
|
14
|
Desiderio A, Goppa L, Santambrogio C, Brocca S, Buratti S, Girometta CE, Sarkar M, Venuti MT, Savino E, Rossi P, Ferrari E. Improving the Proteome-Mining of Schizophyllum commune to Enhance Medicinal Mushroom Applications. J Fungi (Basel) 2025; 11:120. [PMID: 39997414 PMCID: PMC11856175 DOI: 10.3390/jof11020120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
This study presents the first comprehensive proteomic profile of an Italian strain of Schizophyllum commune, a highly heterogeneous white-rot fungal species with significant potential for industrial, nutraceutical, cosmeceutical, and clinical applications. Three protein extraction methods and their impact on yield and resulting protein composition have been compared. Results revealed that the combination of Tris-Cl and urea increases the total protein yield and the variety of enzymatic species related to pivotal pathways. Notably, over 2000 proteins were identified, including enzymes involved in the growth and development of mycelium, trehalose biosynthesis, and different types of carbohydrate-active enzymes (CAZymes). These enzymes are crucial for nutraceutical and agro-industrial applications of S. commune. The multiple-step proteomic approach used could be a model for investigating other fungal species.
Collapse
Affiliation(s)
- Anthea Desiderio
- Department of Earth and Environmental Sciences (DSTA), University of Pavia, 27100 Pavia, Italy; (A.D.); (L.G.); (S.B.); (C.E.G.); (E.S.)
| | - Lorenzo Goppa
- Department of Earth and Environmental Sciences (DSTA), University of Pavia, 27100 Pavia, Italy; (A.D.); (L.G.); (S.B.); (C.E.G.); (E.S.)
| | - Carlo Santambrogio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (C.S.); (S.B.)
| | - Stefania Brocca
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (C.S.); (S.B.)
| | - Simone Buratti
- Department of Earth and Environmental Sciences (DSTA), University of Pavia, 27100 Pavia, Italy; (A.D.); (L.G.); (S.B.); (C.E.G.); (E.S.)
| | - Carolina Elena Girometta
- Department of Earth and Environmental Sciences (DSTA), University of Pavia, 27100 Pavia, Italy; (A.D.); (L.G.); (S.B.); (C.E.G.); (E.S.)
| | - Meghma Sarkar
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (M.S.); (M.T.V.)
| | - Maria Teresa Venuti
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (M.S.); (M.T.V.)
| | - Elena Savino
- Department of Earth and Environmental Sciences (DSTA), University of Pavia, 27100 Pavia, Italy; (A.D.); (L.G.); (S.B.); (C.E.G.); (E.S.)
| | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (M.S.); (M.T.V.)
| | - Emanuele Ferrari
- Molecular Ecology Group (MEG), Water Research Institute (CNR-IRSA), National Research Council of Italy, 28922 Verbania, Italy
| |
Collapse
|
15
|
Heidari R, Assadollahi V, Marashi SN, Elahian F, Mirzaei SA. Identification of Novel lncRNAs Related to Colorectal Cancer Through Bioinformatics Analysis. BIOMED RESEARCH INTERNATIONAL 2025; 2025:5538575. [PMID: 39949372 PMCID: PMC11824705 DOI: 10.1155/bmri/5538575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/15/2024] [Indexed: 02/16/2025]
Abstract
Long noncoding RNA (lncRNA) plays a critical role in cancer cell proliferation, invasion, metastasis, and chemoresistance. The current study introduces novel lncRNAs in colorectal cancer (CRC) through bioinformatics analysis. GSE134834 CRC-related microarray of Gene Expression Omnibus (GEO) was analyzed to identify differentially expressed genes (DEGs) in CRC samples against normal samples. Analysis revealed 6763 DEGs (p < 0.05 and |log fold change (FC)| ≥ 0.5) that include differentially expressed mRNA (DEmRNA) and differentially expressed long noncoding RNA (DElncRNA). Novel lncRNAs were identified, and to better understand the biological function of the identified lncRNAs, gene modules were constructed using weighted gene coexpression network analysis (WGCNA), and finally, two modules for lncRNAs were obtained. The coexpression modules with these lncRNAs were subjected to enrichment analysis in FunRich software to predict their functions through their coexpressed genes. Gene ontology results of modules related to novel lncRNA revealed they significantly enriched the cellular pathways regulation in cancer. The protein-protein interaction (PPI) network of novel lncRNAs-related modules was constructed using Search Tool for the Retrieval of Interacting Genes (STRING) and visualized using the Cytoscape software. Hub genes were screened from the PPI network by the CytoHubba plug-in of Cytoscape. The hub genes were MRTO4, CDK1, CDC20, RPF2, NOP58, NIFK, GTPBP4, BUB1, BUB1B, and BOP1 for the lightpink4 module and BYSL, RPS23 (ribosomal protein S23), RSL1D1 (ribosomal L1 domain containing 1), NAT10, NOP14, GNL2, MRPS12, NOL6 (nucleolar protein 6), IMP4, and RRP12 (ribosomal RNA processing 12 homolog) for the pink module. The expression levels of the top DEmRNA and module hub genes in CRC were validated using the Gene Expression Profiling Interactive Analysis (GEPIA) database. Generally, our findings offer crucial insight into the hub genes and novel lncRNAs in the development of CRC by bioinformatics analysis, information that may prove useful in the identification of new biomarkers and treatment targets in CRC; however, more experimental investigation is required to validate the findings of the present study.
Collapse
Affiliation(s)
- Razieh Heidari
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vahideh Assadollahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyedeh Negar Marashi
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Elahian
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Advanced Technologies Core, Baylor College of Medicine, Houston, Texas, USA
| | - Seyed Abbas Mirzaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
16
|
Sun J, Dong W, Zhao Y, Sun G, Chen L, Huo J, Wang W. Study based on metabolomics and network pharmacology to explore the mechanism of Ginseng-Douch compound fermentation products in the treatment of hyperlipidemia. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:611-625. [PMID: 39264139 DOI: 10.1002/jsfa.13858] [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: 02/02/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Ginseng-Douchi (GD) is a complex fermented product of ginseng and soybean, similar to natto, and is effective in the treatment of hyperlipidemia, but the mechanism of action involved needs to be further explored. RESULTS The present study combines a comprehensive strategy of network pharmacology and metabolomics to explore the lipid-lowering mechanism of GD. First, a hyperlipidemia rats model induced by a high-fat diet was established to evaluate the therapeutic effects of GD. Second, potential biomarkers were identified using serum metabolomics and metabolic pathway analysis was performed with MetaboAnalyst. Third, network pharmacology is used to find potential therapeutic targets based on the blood-influencing components of GD. Finally, core targets were obtained through a target-metabolite and the enrichment analysis of biomarkers-genes. Biochemistry analysis showed that GD exerted hypolipidemic effects on hyperlipidemic rats. Nineteen potential biomarkers for the GD treatment of hyperlipidemia were identified by metabolomics, which was mainly involved in linoleic acid metabolism, glycerophospholipid metabolism, ether lipid metabolism, alpha-linolenic acid metabolism and glycosylphosphatidylinositol-anchor biosynthesis. GD had a callback function for ether lipid metabolism and glycerophospholipid metabolism pathways. Eighteen blood components were identified in serum, associated with 85 potential therapeutic targets. The joint analysis showed that three core therapeutic targets were regulated by GD, including PIK3CA, AKT1 and EGFR. CONCLUSION This study combines serum medicinal chemistry of traditional Chinese medicine, network pharmacology and metabolomics to reveal the regulatory mechanism of GD on hyperlipidemia. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jianfeng Sun
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Wenting Dong
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Yuanyuan Zhao
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Guodong Sun
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Liyan Chen
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Jinhai Huo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Weiming Wang
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| |
Collapse
|
17
|
Sahin F, Gunel A, Atasoy BT, Guler U, Salih B, Kuzu I, Taspinar M, Cinar O, Kahveci S. Enhancing proteasome activity by NMDAR antagonists explains their therapeutic effect in neurodegenerative and mental diseases. Sci Rep 2025; 15:1165. [PMID: 39805913 PMCID: PMC11729902 DOI: 10.1038/s41598-024-84479-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
NMDAR antagonists, such as memantine and ketamine, have shown efficacy in treating neurodegenerative diseases and major depression. The mechanism by which these drugs correct the aforementioned diseases is still unknown. Our study reveals that these antagonists significantly enhance 20S proteasome activity, crucial for degrading intrinsically disordered, oxidatively damaged, or misfolded proteins, factors pivotal in neurodegenerative diseases like Alzheimer's and Parkinson's. In our mouse model experiment, ketamine administration notably altered brain synaptic protein profiles within two hours, significantly downregulating proteins strongly associated with Alzheimer's and Parkinson's diseases. Furthermore, the altered proteins exhibited enrichment in terms related to plasticity and potentiation, including retrograde endocannabinoid signaling-a pivotal pathway in both short- and long-term plasticity that may elucidate the long-lasting effects of ketamine in major depression. Via the ubiquitin-independent 20S proteasome pathway (UIPS), these drugs maintain cellular protein homeostasis, which is crucial as proteasome activity declines with age, leading to protein aggregation and disease symptoms. Therefore, these findings hold promise for new treatment options not only for brain diseases but also for other systemic conditions associated with unfolded or misfolded proteins.
Collapse
Affiliation(s)
- Fikret Sahin
- Department of Medical Microbiology, Ankara University School of Medicine, Ankara, Turkey.
| | - Aslihan Gunel
- Faculty of Arts and Science Department of Chemistry-Biochemistry, Kırşehir Ahi Evran University, Kırşehir, Turkey
| | - Buse Turegun Atasoy
- Department of Medical Microbiology, Ankara University School of Medicine, Ankara, Turkey
| | - Ulku Guler
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Bekir Salih
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Isinsu Kuzu
- Department of Medical Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Mehmet Taspinar
- Department of Medical Biology, Aksaray University School of Medicine, Aksaray, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Selda Kahveci
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
- Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| |
Collapse
|
18
|
Meng T, Wu W, Wang B, Li C, Li J, Liu J, Wang J, Qie R. Treating chronic pulmonary heart disease with traditional Chinese medicine: Systematic evaluation and mechanistic insights into the resolving phlegm and activating blood approach. Heart Lung 2025; 69:111-126. [PMID: 39378530 DOI: 10.1016/j.hrtlng.2024.09.017] [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: 04/02/2024] [Revised: 09/14/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Chronic Pulmonary Heart Disease (CPHD) significantly impacts global health, especially among middle-aged and older adults. In China, the Traditional Chinese Medicine (TCM) technique of Resolving Phlegm and Activating Blood (RPAB) is widely used to treat CPHD, although high-quality evidence supporting its efficacy remains limited. OBJECTIVES The purpose of this study was to rigorously assess the clinical efficacy of RPAB for CPHD and elucidate the mechanisms underlying its primary herbal components. METHODS Through a detailed search of literature in both Chinese and English and strict inclusion and exclusion criteria, 18 randomized controlled trials (RCTs) were selected for meta-analysis. We identified RPAB's core herbal combinations using association rule analysis. This method statistically analyzes the frequency and correlation of herbal medicine usage. We then analyzed the chemical components of these combinations and investigated their potential intervention mechanisms on CPHD through network pharmacology. RESULTS The combination of RPAB with Western medicine was superior to Western medicine alone in improving blood gas analysis and pulmonary function and reducing plasma viscosity in CPHD patients. The core herbal combination identified was Astragalus membranaceus (Fisch.) Bunge, Ligusticum chuanxiong Hort. ex S. H. Qiu & al., and Stellaria alsine Grimm (ALS). This combination targeted 588 therapeutic and 27 core targets. It influenced ten core compounds across 34 pathways, primarily through the chemokine signaling pathway and the JAK-STAT signaling pathway. CONCLUSION RPAB with Western medicine significantly improves CPHD treatment outcomes. The study highlights the therapeutic potential of the ALS combination, which operates through multiple pathways to remodel pulmonary arteries, decrease inflammation, and lessen oxidative stress. These insights support the clinical application of RPAB in CPHD treatment and open new avenues for research and therapeutic development.
Collapse
Affiliation(s)
- Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Weidong Wu
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Jiarui Li
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Jiawen Liu
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Jianying Wang
- The Second Cardiology Department of the Affiliated Second Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150001, China
| | - Rui Qie
- Geriatrics, the first affiliated hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
19
|
Ozel B, Kipcak S, Caglar HO, Kayabasi C, Goker Bagca B, Gunduz C, Selvi Gunel N, Biray Avci C. PI3K/mTOR Inhibitor VS-5584 Alters Expression of WNT Signaling Genes and Induces Apoptosis in Lung Adenocarcinoma Cells: In Vitro and In Silico Insight. Cell Biochem Biophys 2024:10.1007/s12013-024-01643-9. [PMID: 39690396 DOI: 10.1007/s12013-024-01643-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
Lung cancer (LC) accounts for approximately 25% of all cancer cases, with 80-85% of these being non-small cell lung cancer (NSCLC). VS-5584 is a novel anti-cancer agent that specifically inhibits mTORC1/2 and class I PI3K isoforms. There is cross-talk between the PI3K-Akt-mTOR and WNT signaling pathways that are abnormally activated in NSCLC. In this study, we aimed to evaluate the anti-cancer effects of VS-5584 on A549 lung adenocarcinoma cells and changes in WNT signaling gene expression in vitro, while also correlating differentially expressed genes in silico. The effect of VS-5584 on A549 cell viability was assessed by the MTT assay. Apoptosis and cell cycle profiles were analyzed by flow cytometry, while WNT signaling gene expression was measured by quantitative RT-PCR. Differentially expressed genes (DEGs) in the TCGA LUAD and LUSC datasets were identified using the GEPIA2 platform. VS-5584 treatment induced apoptosis and caused cell cycle arrest at the G0/G1 phase in A549 cells. The mRNA expression levels of WNT signaling genes significantly decreased in treated cells. The expression of some upregulated DEGs in the datasets decreased in A549 cells treated with VS-5584. VS-5584 shows promise as an anti-cancer agent in the treatment of NSCLC by downregulating the expression of WNT signaling genes.
Collapse
Affiliation(s)
- Buket Ozel
- Ege University, Faculty of Medicine, Medical Biology Department, Izmir, Turkey
| | - Sezgi Kipcak
- Ege University, Faculty of Medicine, Medical Biology Department, Izmir, Turkey
| | - Hasan Onur Caglar
- Department of Molecular Biology and Genetics, Science Faculty, Erzurum Technical University, Erzurum, Turkey
| | - Cagla Kayabasi
- Balıkesir University, Faculty of Medicine, Medical Biology Department, Balıkesir, Turkey
| | - Bakiye Goker Bagca
- Aydın Adnan Menderes University, Faculty of Medicine, Medical Biology Department, Aydın, Turkey
| | - Cumhur Gunduz
- Ege University, Faculty of Medicine, Medical Biology Department, Izmir, Turkey
| | - Nur Selvi Gunel
- Ege University, Faculty of Medicine, Medical Biology Department, Izmir, Turkey
| | - Cigir Biray Avci
- Ege University, Faculty of Medicine, Medical Biology Department, Izmir, Turkey.
| |
Collapse
|
20
|
Ferrao Blanco MN, Lesage R, Kops N, Fahy N, Bekedam FT, Chavli A, Bastiaansen-Jenniskens YM, Geris L, Chambers MG, Pitsillides AA, Narcisi R, van Osch GJ. A multi-model approach identifies ALW-II-41-27 as a promising therapy for osteoarthritis-associated inflammation and endochondral ossification. Heliyon 2024; 10:e40871. [PMID: 39717596 PMCID: PMC11664402 DOI: 10.1016/j.heliyon.2024.e40871] [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: 06/24/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
Low-grade inflammation and pathological endochondral ossification are key processes underlying the progression of osteoarthritis, the most prevalent joint disease worldwide. In this study, we employed a multi-faceted approach, integrating publicly available datasets, in silico analyses, in vitro experiments and in vivo models to identify new therapeutic candidates targeting these processes. Data mining of transcriptomic datasets identified EPHA2, a receptor tyrosine kinase associated with cancer, as being linked to both inflammation and endochondral ossification in osteoarthritis. A computational model of cellular signaling networks in chondrocytes predicted that in silico activation of EPHA2 in healthy chondrocytes increases inflammatory mediators and induces hypertrophic differentiation, a hallmark of endochondral ossification. We then evaluated the effect of EPHA2 inhibition using the tyrosine kinase inhibitor ALW-II-41-27 in cultured human chondrocytes from individuals with osteoarthritis, demonstrating significant reductions in both inflammation and hypertrophy. Additionally, systemic subcutaneous administration of ALW-II-41-27 in a mouse osteoarthritic model attenuated joint degeneration by reducing local inflammation and pathological endochondral ossification. Collectively, this study demonstrates a novel drug discovery pipeline that integrates computational, experimental, and animal models, paving the way for the development of disease-modifying treatments for osteoarthritis.
Collapse
Affiliation(s)
- Mauricio N. Ferrao Blanco
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raphaelle Lesage
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
- Biomechanics Section, KU Leuven, Belgium
| | - Nicole Kops
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Niamh Fahy
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Applied Science, Technological University of the Shannon: Midlands Midwest, Limerick, Ireland
| | - Fjodor T. Bekedam
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Athina Chavli
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Belgium
- Biomechanics Section, KU Leuven, Belgium
- GIGA In Silico Medicine, University of Liège, Belgium
| | - Mark G. Chambers
- Lilly Research Laboratories, Eli Lilly Pharmaceuticals, Indianapolis, USA
| | | | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, University of Technology Delft, Delft, the Netherlands
| |
Collapse
|
21
|
Patel H, Nagani A, Patel M, Patel M, Yadav MR. Design, synthesis and biological evaluation of some imidazo[1,2- a]pyridine derivatives as anti-tubercular agents: an in silico - in vitro approach. J Biomol Struct Dyn 2024:1-18. [PMID: 39663643 DOI: 10.1080/07391102.2024.2436554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/14/2024] [Indexed: 12/13/2024]
Abstract
In this study, we designed, synthesized and evaluated some novel imidazo[1,2-a]pyridine derivatives as potential anti-TB agents. Preliminary in vitro screening for anti-TB activity of the synthesized compounds was performed against H37Rv strain using the microplate Alamar Blue assay (MABA). Network pharmacology was used to identify the possible targets and pathways of these compounds against Mtb infection. Molecular docking and molecular dynamics simulations were also performed to investigate the binding modes and stability of these compounds with the selected targets. The results showed that some of the synthesized compounds (6b, 6c, 6e, 6f, 6h, 6i, 6j, 6n and 6o) exhibited potent anti-TB activity, with minimum inhibitory concentrations (MICs) ranging from 1.6 to 6.25 μg/mL. The network pharmacology analysis revealed that among the 455 putative targets of imidazo[1,2-a]pyridine derivatives, 24 targets are the potential targets for treatment of Mtb infection. Among these 24 targets, 10 hub-targets were identified (TLR4, ICAM1, TLR9, STAT3, TNFRSF1A, ERBB2, CXCR3, ACE, IKBKG and NOS2) which were significantly involved in GO processes such as positive regulation of DNA-binding transcription factor activity, peptidyl-tyrosine phosphorylation, positive regulation of inflammatory response, mononuclear cell proliferation, regulation of hemopoiesis and cytokine production involved in inflammatory response and KEGG pathways such as pathways in Tuberculosis, NF-kappa B signalling, HIF-1 signalling PD-L1 expression, and PD-1 checkpoint pathway in cancer. Molecular docking and dynamics simulations confirmed the stable interactions of imidazo[1,2-a]pyridine derivatives with core target active sites, highlighting their potential as novel anti-TB drug candidates.
Collapse
Affiliation(s)
- Harnisha Patel
- Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Afzal Nagani
- Parul Institute of Pharmacy, Faculty of Pharmacy, Parul University, Vadodara, Gujarat, India
| | - Mirav Patel
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Research & Development Cell, Parul University, Vadodara, Gujarat, India
| | - Mitesh Patel
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Research & Development Cell, Parul University, Vadodara, Gujarat, India
| | - Mange Ram Yadav
- Research & Development Cell, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
22
|
Giannopoulos-Dimitriou A, Saiti A, Malousi A, Anagnostopoulos AK, Vatsellas G, Al-Maghrabi PM, Müllertz A, Fatouros DG, Vizirianakis IS. Molecular Profiling of A549 Cell-Derived Exosomes: Proteomic, miRNA, and Interactome Analysis for Identifying Potential Key Regulators in Lung Cancer. Cancers (Basel) 2024; 16:4123. [PMID: 39766023 PMCID: PMC11674491 DOI: 10.3390/cancers16244123] [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: 11/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Exosomes, nano-sized extracellular vesicles released by all cells, play a key role in intercellular communication and carry tumorigenic properties that impact surrounding or distant cells. The complexity of the exosomal molecular interactome and its effects on recipient cells still remain unclear. This study aims to decipher the molecular profile and interactome of lung adenocarcinoma A549 cell-derived exosomes using multi-omics and bioinformatics approaches. METHODS We performed comprehensive morphological and physicochemical characterization of exosomes isolated from cell culture supernatant of A549 cells in vitro, using DLS, cryo-TEM, Western blot, and flow cytometry. Proteomic and miRNA high-throughput profiling, coupled with bioinformatics network analysis, were applied to elucidate the exosome molecular cargo. A comparative miRNA analysis was also conducted with exosomes derived from normal lung fibroblast MRC-5 cells. RESULTS Exosomes exhibited an average size of ~40 nm and disk-shaped lipid bilayer structures, with tetraspanins CD9 and CD63 validated as exosomal markers. Proteomic analysis identified 68 proteins, primarily linked to the extracellular matrix organization and metabolic processes. miRNA sequencing revealed 72 miRNAs, notably hsa-miR-619-5p, hsa-miR-122-5p, hsa-miR-9901, hsa-miR-7704, and hsa-miR-151a-3p, which are involved in regulating metabolic processes, gene expression, and tumorigenic pathways. Th integration of proteomic and miRNA data through a proteogenomics approach identified dually affected genes including ERBB2, CD44, and APOE, impacted by both exosomal miRNA targeting and protein interactions through synergistic or antagonistic interactions. Differential analysis revealed a distinct miRNA profile in A549 exosomes, associated with cancer-related biological processes, compared to MRC-5 exosomes; notably, hsa-miR-619-5p emerged as a promising candidate for future clinical biomarker studies. The network analysis also revealed genes targeted by multiple upregulated tumor-associated miRNAs in potential exosome-recipient cells. CONCLUSIONS This integrative study provides insights into the molecular interactome of lung adenocarcinoma A549 cell-derived exosomes, providing a foundation for future research on exosomal cargo and its role in tumor cell communication, growth, and progression.
Collapse
Affiliation(s)
| | - Aikaterini Saiti
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.G.-D.); (A.S.)
| | - Andigoni Malousi
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios K. Anagnostopoulos
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Giannis Vatsellas
- Greek Genome Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece;
| | - Passant M. Al-Maghrabi
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Dimitrios G. Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.G.-D.); (A.S.)
- Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| |
Collapse
|
23
|
Ferrara B, Bourgoin-Voillard S, Habert D, Vallée B, Nicolas-Boluda A, Simanic I, Seve M, Vingert B, Gazeau F, Castellano F, Cohen J, Courty J, Cascone I. Matrix stiffness regulates the protein profile of extracellular vesicles of pancreatic cancer cell lines. Proteomics 2024; 24:e2400058. [PMID: 39279557 DOI: 10.1002/pmic.202400058] [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: 02/20/2024] [Revised: 07/31/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024]
Abstract
The fibrotic stroma characterizing pancreatic ductal adenocarcinoma (PDAC) derives from a progressive tissue rigidification, which induces epithelial mesenchymal transition and metastatic dissemination. The aim of this study was to investigate the influence of matrix stiffness on PDAC progression by analyzing the proteome of PDAC-derived extracellular vesicles (EVs). PDAC cell lines (mPDAC and KPC) were grown on synthetic supports with a stiffness close to non-tumor (NT) or tumor tissue (T), and the protein expression levels in cell-derived EVs were analyzed by a quantitative MSE label-free mass spectrometry approach. Our analysis figured out 15 differentially expressed proteins (DEPs) in mPDAC-EVs and 20 DEPs in KPC-EVs in response to matrix rigidification. Up-regulated proteins participate to the processes of metabolism, matrix remodeling, and immune response, altogether hallmarks of PDAC progression. A multimodal network analysis revealed that the majority of DEPs are strongly related to pancreatic cancer. Interestingly, among DEPs, 11 related genes (ACTB/ANXA7/C3/IGSF8/LAMC1/LGALS3/PCD6IP/SFN/TPM3/VARS/YWHAZ) for mPDAC-EVs and 9 (ACTB/ALDH2/GAPDH/HNRNPA2B/ITGA2/NEXN/PKM/RPN1/S100A6) for KPC-EVs were significantly overexpressed in tumor tissues according to gene expression profiling interaction analysis (GEPIA). Concerning the potential clinical relevance of these data, the cluster of ACTB, ITGA2, GAPDH and PKM genes displayed an adverse effect (p < 0.05) on the overall survival of PDAC patients.
Collapse
Affiliation(s)
- Benedetta Ferrara
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sandrine Bourgoin-Voillard
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, CHU Grenoble Alpes, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, LBFA et BEeSy, Inserm, U1055, CHU Grenoble Alpes, PROMETHEE Proteomic Platform, Grenoble, France
| | - Damien Habert
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - Benoit Vallée
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - Alba Nicolas-Boluda
- Matière et Systèmes Complexes MSC, CNRS, Université Paris Cité, Paris, France
| | - Isidora Simanic
- Modèles de cellules souches malignes et therapeutiques, INSERM UMR-S 935, Université Paris-Saclay, Villejuif, France
| | - Michel Seve
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, CNRS UMR 5525, Grenoble INP, CHU Grenoble Alpes, TIMC, EPSP, Grenoble, France
- Université Grenoble Alpes, LBFA et BEeSy, Inserm, U1055, CHU Grenoble Alpes, PROMETHEE Proteomic Platform, Grenoble, France
| | - Benoit Vingert
- Etablissement Français du Sang, Créteil, France
- Inserm, U955, Equipe 2, Créteil, France
| | - Florence Gazeau
- Matière et Systèmes Complexes MSC, CNRS, Université Paris Cité, Paris, France
| | - Flavia Castellano
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
| | - José Cohen
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Centre d'investigation clinique Biotherapie, Créteil, France
| | - José Courty
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Centre d'investigation clinique Biotherapie, Créteil, France
| | - Ilaria Cascone
- Immunorégulation et Biothérapie, INSERM U955, Hôpital Henri Mondor, Université Paris-Est, Créteil, France
- AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Centre d'investigation clinique Biotherapie, Créteil, France
| |
Collapse
|
24
|
An Z, Sun Y, Yang X, Zhou J, Yu Y, Zhang B, Xu Z, Zhu Y, Wang G. Enhanced expression of miR-20a driven by nanog exacerbated the degradation of extracellular matrix in thoracic aortic dissection. Noncoding RNA Res 2024; 9:1040-1049. [PMID: 39022686 PMCID: PMC11254500 DOI: 10.1016/j.ncrna.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a life-threatening vascular disease manifested as intramural bleeding in the medial layers of the thoracic aorta. The key histopathologic feature of TAD is medial degeneration, characterized by depletion of vascular smooth muscle cells (VSMCs) and degradation of extracellular matrix (ECM). MicroRNA, as essential epigenetic regulators, can inhibit the protein expression of target genes without modifying the sequences. This study aimed to elucidate the role and underlying mechanism of miR-20a, a member of the miR-17-92 cluster, in regulating ECM degradation during the pathogenesis of TAD. The expression of the miR-17-92 cluster was significantly increased in synthetic VSMCs derived from TAD lesions compared to contractile VSMCs isolated from normal thoracic aortas. Notably, the expression of miR-20a was increased in VSMCs in response to serum exposure and various stimuli. In TAD lesions, the expression of miR-20a was significantly negatively correlated with that of elastin. Elevated expression of miR-20a was also observed in thoracic aortas of TAD mice induced by β-aminopropionitrile fumarate and angiotensin II. Overexpression of miR-20a via mimic transfection enhanced the growth and invasive capabilities of VSMCs, with no significant impact on their migratory activity or the expression of phenotypic markers (α-SMA, SM22, and OPN). Silencing of miR-20a with inhibitor transfection mitigated the hyperactivation of MMP2 in VSMCs stimulated by PDGF-bb, as evidenced by reduced levels of active-MMP2 and increased levels of pro-MMP2. Subsequently, TIMP2 was identified as a novel target gene of miR-20a. The role of miR-20a in promoting the activation of MMP2 was mediated by the suppression of TIMP2 expression in VSMCs. In addition, the elevated expression of miR-20a was found to be directly driven by Nanog in VSMCs. Collectively, these findings indicate that miR-20a plays a crucial role in maintaining the homeostasis of the thoracic aortic wall during TAD pathogenesis and may represent a potential therapeutic target for TAD.
Collapse
Affiliation(s)
- Zhao An
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yangyong Sun
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaodong Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingwen Zhou
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yongchao Yu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Boyao Zhang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuming Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guokun Wang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
25
|
Nguyen KN, Graner AN, Fringuello AR, Zizzo Z, Valenzuela L, Anyanwu K, Lillehei KO, Youssef AS, Guzman S, Coughlan C, Graner MW. Extracellular Vesicles from a Novel Chordoma Cell Line, ARF-8, Promote Tumorigenic Microenvironmental Changes When Incubated with the Parental Cells and with Human Osteoblasts. Int J Mol Sci 2024; 25:12731. [PMID: 39684443 DOI: 10.3390/ijms252312731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Chordomas are rare, generally slow-growing spinal tumors that nonetheless exhibit progressive characteristics over time, leading to malignant phenotypes and high recurrence rates, despite maximal therapeutic interventions. The tumors are notoriously resistant to therapies and are often located in regions that complicate achieving gross total resections. Cell lines from these tumors are rare as well. We cultured a new chordoma cell line (ARF-8) derived from an extensive clival chordoma that extended back to the cervical spine. We characterized the ARF-8 cellular and extracellular vesicle (EV) proteomes, as well as the impacts of ARF-8 EVs on the proteomes and secretomes of recipient cells (both ARF-8 and human osteoblasts) in autocrine and paracrine settings. Our proteomic analyses suggested roles for transforming growth factor beta (TGFB/TGFβ), cell-matrix interactions involving the epithelial-to-mesenchymal transition (EMT), and cell-extracellular matrix interactions in cell migration, consistent with a migratory/metastatic tumor phenotype. We demonstrated that ARF-8 tumor cell migration was dependent on general (arginine-glycine-aspartic acid [RGD]-based) integrin activity and that ARF-8 EVs could promote such migration. ARF-8 EVs also prompted proteomic/secretomic changes in human osteoblast cells, again with indications that cell-cell and cell-extracellular matrix interactions would be activated. All the characteristics typically associated with chordomas as cancers-migration and invasion, therapeutic resistance, metastatic potential-can be driven by tumor EVs. Overall, ARF-8 EVs promoted predicted tumorigenic phenotypes in recipient cells and suggested novel therapeutic targets for chordomas.
Collapse
Affiliation(s)
- Khoa N Nguyen
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Arin N Graner
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anthony R Fringuello
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zoe Zizzo
- Department of Biochemistry, Colorado College, 14 E Cache La Poudre St., Colorado Springs, CO 80903, USA
| | - Lorena Valenzuela
- Department of Biomedical Sciences, Regis University, 3333 Regis Blvd., Denver, CO 80221, USA
| | - Kamara Anyanwu
- Department of Biomedical Sciences, Claremont McKenna College, 888 N Columbia Ave., Claremont, CA 91711, USA
| | - Kevin O Lillehei
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - A Samy Youssef
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Samuel Guzman
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Christina Coughlan
- Department of Neurology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael W Graner
- Department of Neurosurgery, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
26
|
Pérez-Mato M, Dopico-López A, Akkoc Y, López-Amoedo S, Correa-Paz C, Candamo-Lourido M, Iglesias-Rey R, López-Arias E, Bugallo-Casal A, da Silva-Candal A, Bravo SB, Chantada-Vázquez MDP, Arias S, Santamaría-Cadavid M, Estany-Gestal A, Zaghmi A, Gauthier MA, Gutiérrez-Fernández M, Martin A, Llop J, Rodríguez C, Almeida Á, Migliavacca M, Polo E, Pelaz B, Gozuacik D, El Yamani N, SenGupta T, Rundén-Pran E, Vivancos J, Castellanos M, Díez-Tejedor E, Sobrino T, Rabinkov A, Mirelman D, Castillo J, Campos F. Preclinical validation of human recombinant glutamate-oxaloacetate transaminase for the treatment of acute ischemic stroke. iScience 2024; 27:111108. [PMID: 39524351 PMCID: PMC11543921 DOI: 10.1016/j.isci.2024.111108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/21/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The blood enzyme glutamate-oxaloacetate transaminase (GOT) has been postulated as an effective therapeutic to protect the brain during stroke. To demonstrate its potential clinical utility, a new human recombinant form of GOT (rGOT) was produced for medical use. We tested the pharmacokinetics and evaluated the protective efficacy of rGOT in rodent and non-human primate models that reflected clinical stroke conditions. We found that continuous intravenous administration of rGOT within the first 8 h after ischemic onset significantly reduced the infarct size in both severe (30%) and mild lesions (48%). Cerebrospinal fluid and proteomics analysis, in combination with positron emission tomography imaging, indicated that rGOT can reach the brain and induce cytoprotective autophagy and induce local protection by alleviating neuronal apoptosis. Our results suggest that rGOT can be safely used immediately in patients suspected of having a stroke. This study requires further validation in clinical stroke populations.
Collapse
Affiliation(s)
- María Pérez-Mato
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Antonio Dopico-López
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul 34450, Turkey
- Department of Medical Biology, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Sonia López-Amoedo
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María Candamo-Lourido
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ana Bugallo-Casal
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Andrés da Silva-Candal
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, 15006 A Coruña, Spain
| | - Susana B. Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María del Pilar Chantada-Vázquez
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Research Unit, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain
| | - Susana Arias
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - María Santamaría-Cadavid
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unit of Methodology of the Research, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ahlem Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC J3X 1S2, Canada
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Marc A. Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC J3X 1S2, Canada
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Abraham Martin
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Basque Foundation for Science, 48009 Bilbao, Spain
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 San Sebastian, Spain
| | - Cristina Rodríguez
- Institute of Functional Biology and Genomics (IBFG), CSIC, University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, CSIC, University of Salamanca, 37007 Salamanca, Spain
| | - Ángeles Almeida
- Institute of Functional Biology and Genomics (IBFG), CSIC, University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, CSIC, University of Salamanca, 37007 Salamanca, Spain
| | - Martina Migliavacca
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Ester Polo
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul 34450, Turkey
- Department of Medical Biology, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Naouale El Yamani
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - Tanima SenGupta
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - Elise Rundén-Pran
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - José Vivancos
- Stroke Unit, Department of Neurology, Hospital Universitario de La Princesa & Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Mar Castellanos
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, 15006 A Coruña, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Aharon Rabinkov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - David Mirelman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
27
|
Gera R, Arora R, Chhabra P, Sharma U, Parsad R, Ahlawat S, Mir MA, Singh MK, Kumar R. Exploring transcriptomic mechanisms underlying pulmonary adaptation to diverse environments in Indian rams. Mol Biol Rep 2024; 51:1111. [PMID: 39485559 DOI: 10.1007/s11033-024-10067-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024]
Abstract
BACKGROUND The Changthangi sheep thrive at high altitudes in the cold desert regions of Ladakh, India while Muzaffarnagri sheep are well-suited to the low altitude plains of northern India. This study investigates the molecular mechanisms of pulmonary adaptation to diverse environments by analyzing gene expression profiles of lung tissues through RNA sequencing. METHODS AND RESULTS Four biological replicates of lung tissue from each breed were utilized to generate the transcriptomic data. Differences in gene expression analysis revealed discrete expression profiles in lungs of each breed. In Changthangi sheep, genes related to immune responses, particularly cytokine signaling, were significantly enriched. Pathway analysis highlighted the activation of NF-kB signaling, a key mediator of inflammation and immune response. Additionally, the gene network analysis indicated a strong association between cytokine signaling, hypoxia-inducible factor (HIF) and NF-kB activation, suggesting a coordinated response to hypoxic stress in lungs of Changthangi sheep. In Muzaffarnagri sheep, the gene expression profiles were enriched for pathways related to energy metabolism, homeostasis and lung physiology. Key pathways identified include collagen formation and carbohydrate metabolism, both of which are crucial for maintaining lung function and structural integrity. Gene network analysis further reinforced this by revealing a strong connection between genes associated with lung structure and function. CONCLUSIONS Our findings shed light on the valuable insights into gene expression mechanisms that enable these sheep breeds to adapt to their respective environments and contribute to a better understanding of high altitude adaptation in livestock.
Collapse
Affiliation(s)
- Ritika Gera
- ICAR-National Bureau of Animal Genetic Resources, Karnal, 132001, Haryana, India
- UIET, Kurukshetra University, Kurukshetra, 136119, Haryana, India
| | - Reena Arora
- ICAR-National Bureau of Animal Genetic Resources, Karnal, 132001, Haryana, India.
| | - Pooja Chhabra
- ICAR-National Bureau of Animal Genetic Resources, Karnal, 132001, Haryana, India
| | - Upasna Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, 132001, Haryana, India
| | - Ram Parsad
- ICAR-National Bureau of Animal Genetic Resources, Karnal, 132001, Haryana, India
| | - Sonika Ahlawat
- ICAR-National Bureau of Animal Genetic Resources, Karnal, 132001, Haryana, India
| | - Mohsin Ayoub Mir
- Mountain Research Centre for Sheep and Goat, SKUAST, Aulestang, 190006, Shuhama, Kashmir, India
| | - Manoj Kumar Singh
- ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura, 281122, Uttar Pradesh, India
| | - Rajesh Kumar
- UIET, Kurukshetra University, Kurukshetra, 136119, Haryana, India
| |
Collapse
|
28
|
Hallal SM, Sida LA, Tűzesi Á, Shivalingam B, Sim H, Buckland ME, Satgunaseelan L, Alexander KL. Size matters: Biomolecular compositions of small and large extracellular vesicles in the urine of glioblastoma patients. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70021. [PMID: 39554867 PMCID: PMC11565258 DOI: 10.1002/jex2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/10/2024] [Accepted: 10/24/2024] [Indexed: 11/19/2024]
Abstract
The promise of urinary extracellular vesicles (uEVs) in biomarker discovery is emerging. However, the characteristics and compositions of different uEV subpopulations across normal physiological and pathological states require rigorous explication. We recently reported proteomic signatures of small (s)-uEVs (<200 nm membranous nanoparticles) and described putative biomarkers corresponding to the diagnosis, tumour burden and recurrence of the lethal adult primary brain tumour, glioblastoma. Here, we comprehensively characterise uEV populations with significantly different mean and mode particle sizes obtained by differential centrifugation at 100,000 × g (100K-uEVs; smaller) and 17,000 × g (17K-uEVs; larger) using Fourier-transform infrared spectroscopy and quantitative data-independent acquisition mass spectrometry. We show distinct differences in protein and lipid content, prominent protein secondary structures, and proteome distributions between uEV populations that can distinguish glioblastoma patients from healthy controls and correspond to clinically relevant tumour changes (i.e., recurrence and treatment resistance). Among the key findings is a putative seven-protein biomarker panel associated with 17K-uEVs that could distinguish all glioblastoma patients from healthy controls and accurately classify 98.2% of glioblastoma samples. These novel, significant findings demonstrate that both uEV populations offer individual and combined biomarker potential. Further research is warranted to elucidate the complete diagnostic, prognostic, and predictive capabilities of often-neglected 17K-uEV populations.
Collapse
Affiliation(s)
- Susannah M. Hallal
- Neurosurgery DepartmentChris O'Brien LifehouseCamperdownNSWAustralia
- Department of NeuropathologyRoyal Prince Alfred HospitalCamperdownNSWAustralia
- School of Medical SciencesThe University of SydneyCamperdownNSWAustralia
| | - Liam A. Sida
- School of Medical SciencesThe University of SydneyCamperdownNSWAustralia
| | - Ágota Tűzesi
- Department of NeuropathologyRoyal Prince Alfred HospitalCamperdownNSWAustralia
- School of Medical SciencesThe University of SydneyCamperdownNSWAustralia
| | - Brindha Shivalingam
- Neurosurgery DepartmentChris O'Brien LifehouseCamperdownNSWAustralia
- Neurosurgery DepartmentRoyal Prince Alfred HospitalCamperdownNSWAustralia
- Sydney Medical School, Faculty of Medicine and Health SciencesThe University of SydneyCamperdownNSWAustralia
| | - Hao‐Wen Sim
- Department of Medical OncologyChris O'Brien LifehouseCamperdownNSWAustralia
- NHMRC Clinical Trials CentreThe University of SydneyCamperdownNSWAustralia
- Faculty of Medicine and HealthUniversity of New South WalesKensingtonNSWAustralia
| | - Michael E. Buckland
- Department of NeuropathologyRoyal Prince Alfred HospitalCamperdownNSWAustralia
- School of Medical SciencesThe University of SydneyCamperdownNSWAustralia
| | - Laveniya Satgunaseelan
- Neurosurgery DepartmentChris O'Brien LifehouseCamperdownNSWAustralia
- Department of NeuropathologyRoyal Prince Alfred HospitalCamperdownNSWAustralia
- Sydney Medical School, Faculty of Medicine and Health SciencesThe University of SydneyCamperdownNSWAustralia
| | - Kimberley L. Alexander
- Neurosurgery DepartmentChris O'Brien LifehouseCamperdownNSWAustralia
- Department of NeuropathologyRoyal Prince Alfred HospitalCamperdownNSWAustralia
- School of Medical SciencesThe University of SydneyCamperdownNSWAustralia
| |
Collapse
|
29
|
Matamoros‐Angles A, Karadjuzovic E, Mohammadi B, Song F, Brenna S, Meister SC, Siebels B, Voß H, Seuring C, Ferrer I, Schlüter H, Kneussel M, Altmeppen HC, Schweizer M, Puig B, Shafiq M, Glatzel M. Efficient enzyme-free isolation of brain-derived extracellular vesicles. J Extracell Vesicles 2024; 13:e70011. [PMID: 39508423 PMCID: PMC11541858 DOI: 10.1002/jev2.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024] Open
Abstract
Extracellular vesicles (EVs) have gained significant attention as pathology mediators and potential diagnostic tools for neurodegenerative diseases. However, isolation of brain-derived EVs (BDEVs) from tissue remains challenging, often involving enzymatic digestion steps that may compromise the integrity of EV proteins and overall functionality. Here, we describe that collagenase digestion, commonly used for BDEV isolation, produces undesired protein cleavage of EV-associated proteins in brain tissue homogenates and cell-derived EVs. In order to avoid this effect, we studied the possibility of isolating BDEVs with a reduced amount of collagenase or without any protease. Characterization of the isolated BDEVs from mouse and human samples (both female and male) revealed their characteristic morphology and size distribution with both approaches. However, we show that even minor enzymatic digestion induces 'artificial' proteolytic processing in key BDEV markers, such as Flotillin-1, CD81, and the cellular prion protein (PrPC), whereas avoiding enzymatic treatment completely preserves their integrity. We found no major differences in mRNA and protein content between non-enzymatically and enzymatically isolated BDEVs, suggesting that the same BDEV populations are purified with both approaches. Intriguingly, the lack of Golgi marker GM130 signal, often referred to as contamination indicator (or negative marker) in EV preparations, seems to result from enzymatic digestion rather than from its actual absence in BDEV samples. Overall, we show that non-enzymatic isolation of EVs from brain tissue is possible and avoids artificial pruning of proteins while achieving an overall high BDEV yield and purity. This protocol will help to understand the functions of BDEV and their associated proteins in a near-physiological setting, thus opening new research approaches.
Collapse
Affiliation(s)
| | - Emina Karadjuzovic
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Behnam Mohammadi
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Feizhi Song
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Santra Brenna
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | | | - Bente Siebels
- Section Mass Spectrometry and ProteomicsUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Hannah Voß
- Section Mass Spectrometry and ProteomicsUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Carolin Seuring
- Multi‐User‐CryoEM‐FacilityCentre for Structural Systems Biology (CSSB)HamburgGermany
- Department of ChemistryUniversität HamburgHamburgGermany
- Leibniz Institute of Virology (LIV)HamburgGermany
| | - Isidre Ferrer
- IDIBELLUniversity of BarcelonaL'Hospitalet de LlobregatSpain
| | - Hartmut Schlüter
- Section Mass Spectrometry and ProteomicsUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Matthias Kneussel
- Institute for Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | | | - Michaela Schweizer
- Electron Microscopy Core Facility, Center for Molecular Neurobiology (ZMNH)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Berta Puig
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Mohsin Shafiq
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| | - Markus Glatzel
- Institute of NeuropathologyUniversity Medical Center Hamburg‐Eppendorf (UKE)HamburgGermany
| |
Collapse
|
30
|
Kokesh KJ, Bala N, Dogan YE, Nguyen VAL, Costa M, Alli A. Mycobacterium avium inhibits protein kinase C and MARCKS phosphorylation in human cystic fibrosis and non-cystic fibrosis cells. PLoS One 2024; 19:e0308299. [PMID: 39413095 PMCID: PMC11482691 DOI: 10.1371/journal.pone.0308299] [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: 11/21/2023] [Accepted: 07/03/2024] [Indexed: 10/18/2024] Open
Abstract
In cystic fibrosis (CF), there is abnormal translocation and function of the cystic fibrosis transmembrane conductance regulator (CFTR) and an upregulation of the epithelial sodium channel (ENaC). This leads to hyperabsorption of sodium and fluid from the airway, dehydrated mucus, and an increased risk of respiratory infections. In this study, we performed a proteomic assessment of differentially regulated proteins from CF and non-CF small airway epithelial cells (SAEC) that are sensitive to Mycobacterium avium. CF SAEC and normal non-CF SAEC were infected with M. avium before the cells were harvested for protein. Protein kinase C (PKC) activity was greater in the CF cells compared to the non-CF cells, but the activity was significantly attenuated in both cell types after infection with M. avium compared to vehicle. Western blot and densitometric analysis showed a significant increase in cathepsin B protein expression in M. avium infected CF cells. Myristoylated alanine rich C-kinase substrate (MARCKS) protein was one of several differentially expressed proteins between the groups that was identified by mass spectrometry-based proteomics. Total MARCKS protein expression was greater in CF cells compared to non-CF cells. Phosphorylation of MARCKS at serine 163 was also greater in CF cells compared to non-CF cells after treating both groups of cells with M. avium. Taken together, MARCKS protein is upregulated in CF cells and there is decreased phosphorylation of the protein due to a decrease in PKC activity and presumably increased cathepsin B mediated proteolysis of the protein after M. avium infection.
Collapse
Affiliation(s)
- Kevin J. Kokesh
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Niharika Bala
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Yunus E. Dogan
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Pediatrics, Erciyes University of Medicine, Kayseri, Turkey
| | - Van-Anh L. Nguyen
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Marcus Costa
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Abdel Alli
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Medicine, Division of Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, Florida, United States of America
| |
Collapse
|
31
|
Awadelkareem AM, Patel M, Banu H, Adnan M. Integrating computational methods and i n vitro experimental validation reveals the pharmacological mechanism of Selaginella bryopteris (L.) Baker targeting major proteins in breast cancer. Heliyon 2024; 10:e38801. [PMID: 39430520 PMCID: PMC11489316 DOI: 10.1016/j.heliyon.2024.e38801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024] Open
Abstract
Breast cancer remains a significant global health challenge, necessitating the exploration of novel therapeutic options. The present study employs an integrated approach encompassing network pharmacology, molecular docking, molecular dynamics simulations, and in-vitro validation to investigate the potential of Selaginella bryopteris in breast cancer treatment. Initial network pharmacology analysis revealed different potential targets and pathways associated with breast cancer that could be modulated by S. bryopteris phytochemical constituents. Molecular docking and dynamics simulations further elucidated the stability and dynamics of protein-ligand complexes (lanaroflavone-EGFR and sequoiaflavone-CTNNB1). The in-vitro assays demonstrated the ability of S. bryopteris crude extract to inhibit cancer cell growth (IC50 - 78.34 μg/mL) migration and invasion, supporting the computational predictions. The integrated approach employed in the present study offers a robust framework for the systematic exploration of S. bryopteris in drug discovery as a promising candidate for breast cancer treatment.
Collapse
Affiliation(s)
- Amir Mahgoub Awadelkareem
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Mitesh Patel
- Research and Development Cell, Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Humera Banu
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| |
Collapse
|
32
|
Shanbhag S, Al-Sharabi N, Fritz-Wallace K, Kristoffersen EK, Bunæs DF, Romandini M, Mustafa K, Sanz M, Gruber R. Proteomic Analysis of Human Serum Proteins Adsorbed onto Collagen Barrier Membranes. J Funct Biomater 2024; 15:302. [PMID: 39452600 PMCID: PMC11508515 DOI: 10.3390/jfb15100302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
Collagen barrier membranes are frequently used in guided tissue and bone regeneration. The aim of this study was to analyze the signature of human serum proteins adsorbed onto collagen membranes using a novel protein extraction method combined with mass spectrometry. Native porcine-derived collagen membranes (Geistlich Bio-Gide®, Wolhusen, Switzerland) were exposed to pooled human serum in vitro and, after thorough washing, subjected to protein extraction either in conjunction with protein enrichment or via a conventional surfactant-based method. The extracted proteins were analyzed via liquid chromatography with tandem mass spectrometry. Bioinformatic analysis of global profiling, gene ontology, and functional enrichment of the identified proteins was performed. Overall, a total of 326 adsorbed serum proteins were identified. The enrichment and conventional methods yielded similar numbers of total (315 vs. 309), exclusive (17 vs. 11), and major bone-related proteins (18 vs. 14). Most of the adsorbed proteins (n = 298) were common to both extraction groups and included several growth factors, extracellular matrix (ECM) proteins, cell adhesion molecules, and angiogenesis mediators involved in bone regeneration. Functional analyses revealed significant enrichment of ECM, exosomes, immune response, and cell growth components. Key proteins [transforming growth factor-beta 1 (TGFβ1), insulin-like growth factor binding proteins (IGFBP-5, -6, -7)] were exclusively detected with the enrichment-based method. In summary, native collagen membranes exhibited a high protein adsorption capacity in vitro. While both extraction methods were effective, the enrichment-based method showed distinct advantages in detecting specific bone-related proteins. Therefore, the use of multiple extraction methods is advisable in studies investigating protein adsorption on biomaterials.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5009 Bergen, Norway
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0455 Oslo, Norway
| | - Niyaz Al-Sharabi
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| | - Katarina Fritz-Wallace
- Proteomics Unit of University of Bergen (PROBE), University of Bergen, 5009 Bergen, Norway
| | - Einar K. Kristoffersen
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, 5009 Bergen, Norway
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway
| | - Dagmar Fosså Bunæs
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| | - Mario Romandini
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0455 Oslo, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| | - Mariano Sanz
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0455 Oslo, Norway
- ETEP Research Group, University Complutense of Madrid, 28040 Madrid, Spain
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
33
|
Torres Iglesias G, López-Molina M, Botella L, Laso-García F, Chamorro B, Fernández-Fournier M, Puertas I, Bravo SB, Alonso-López E, Díez-Tejedor E, Gutiérrez-Fernández M, Otero-Ortega L. Differential Protein Expression in Extracellular Vesicles Defines Treatment Responders and Non-Responders in Multiple Sclerosis. Int J Mol Sci 2024; 25:10761. [PMID: 39409091 PMCID: PMC11477160 DOI: 10.3390/ijms251910761] [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: 09/13/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Multiple sclerosis (MS) remains the leading cause of neurological disability among young adults worldwide, underscoring the urgent need to define the best therapeutic strategy. Recent advances in proteomics have deepened our understanding of treatment mechanisms and revealed promising biomarkers for predicting therapeutic outcomes. This study focuses on the identification of a protein profile of circulating extracellular vesicles (EVs) derived from neurons, oligodendrocytes, and B and T cells able to differentiate treatment responders and non-responders in 80 patients with MS. In the patients who responded to treatment, T cell-derived EVs were enriched in LV151, a protein involved in the promotion of anti-inflammatory cytokines, whereas Bcell-derived EVs showed elevated PSMD6 and PTPRC, related to immunoproteasome function. Oligodendrocyte- and neuron-derived EVs showed upregulated CO6A1 and COEA1, involved in extracellular matrix reorganisation, as well as LAMA5, NonO, SPNT, and NCAM, which are critical for brain repair. In contrast, non-responders showed higher levels of PSMD7 and PRS10 from B cell-derived EVs, associated with DNA damage, and increased levels of PERM and PERL from T cell-derived EVs, linked to nuclear factor kappa B activation and drug-resistant proteins such as HS90A and RASK. These findings highlight a distinct panel of proteins in EVs that could serve as an early indicator of treatment efficacy in MS.
Collapse
Affiliation(s)
- Gabriel Torres Iglesias
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - MariPaz López-Molina
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Lucía Botella
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Fernando Laso-García
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Beatriz Chamorro
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Mireya Fernández-Fournier
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Inmaculada Puertas
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Susana B. Bravo
- Proteomics Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
| | - Elisa Alonso-López
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| | - Laura Otero-Ortega
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research—IdiPAZ (La Paz University Hospital—Universidad Autónoma de Madrid), 28046 Madrid, Spain; (G.T.I.); (M.L.-M.); (L.B.); (F.L.-G.); (B.C.); (M.F.-F.); (I.P.); (E.A.-L.); (E.D.-T.); (M.G.-F.)
| |
Collapse
|
34
|
Saha P, Talwar P. Identification of PPREs and PPRE associated genes in the human genome: insights into related kinases and disease implications. Front Immunol 2024; 15:1457648. [PMID: 39434882 PMCID: PMC11491715 DOI: 10.3389/fimmu.2024.1457648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction "Peroxisome Proliferator-Activated Receptors" (PPARs) belong to the class of transcription factors (TF) identified as Nuclear Receptors (NR). Upon activation by peroxisome proliferators (PPs), PPARs modulate a diverse range of genes, consequently regulating intra-cellular lipid metabolism, glucose uptake, apoptosis, and cell proliferation. Subsequent to the heterodimerization of Retinoid X Receptors (RXR) with PPARs induced by the binding of activators to PPARs, facilitates the binding of the resulting complex to Peroxisome Proliferator-Activated Receptors Response Elements (PPRE), with a consensus sequence 5'AGGTCANAGGTCA-3', and regulate the transcription of the targeted genes. Methods A comprehensive screening of PPRE within the whole human genome was performed using the Genome Workbench and UCSC Genome Browser to find the associated genes. Subsequently, the kinase subset was isolated from the extracted list of PPRE-related genes. Functional enrichment of the kinases was performed using FunRich, ToppGene, and ShinyGO. Network analysis and enrichment studies were then further performed using NDEx to elucidate these identified kinases' connections and significance. Additionally, the disease association of the PPRE kinases was analyzed using DisGeNET data in R studio and the COSMIC dataset. Results A comprehensive analysis of 1002 PPRE sequences within the human genome (T2T), yielded the identification of 660 associated genes, including 29 kinases. The engagement of these kinases in various biological pathways, such as apoptosis, platelet activation, and cytokine pathways, revealed from the functional enrichment analysis, illuminates the multifaceted role of PPAR in the regulation of cellular homeostasis and biological processes. Network analysis reveals the kinases interact with approximately 5.56% of the Human Integrated Protein-Protein Interaction rEference (HIPPIE) network. Disease association analysis using DisGeNET and COSMIC datasets revealed the significant roles of these kinases in cellular processes and disease modulation. Discussion This study elucidates the regulatory role of PPAR-associated genes and their association with numerous biological pathways. The involvement of the kinases with disease-related pathways highlights new potential for the development of therapeutic strategies designed for disease management and intervention.
Collapse
|
35
|
Kumar M, Patel K, Chinnapparaj S, Sharma T, Aggarwal A, Singla N, Karthigeyan M, Singh A, Sahoo SK, Tripathi M, Takkar A, Gupta T, Pal A, Attri SV, Bansal YS, Ratho RK, Gupta SK, Khullar M, Vashishta RK, Mukherjee KK, Grover VK, Prasad R, Chatterjee A, Gowda H, Bhagat H. Dysregulated Genes and Signaling Pathways in the Formation and Rupture of Intracranial Aneurysm. Transl Stroke Res 2024; 15:865-879. [PMID: 37644376 DOI: 10.1007/s12975-023-01178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 03/21/2023] [Accepted: 07/10/2023] [Indexed: 08/31/2023]
Abstract
Intracranial aneurysm (IA) has the potential to rupture. Despite scientific advances, we are still not in a position to screen patients for IA and identify those at risk of rupture. It is critical to comprehend the molecular basis of disease to facilitate the development of novel diagnostic strategies. We used transcriptomics to identify the dysregulated genes and understand their role in the disease biology. In particular, RNA-Seq was performed in tissue samples of controls, unruptured IA, and ruptured IA. Dysregulated genes (DGs) were identified and analyzed to understand the functional aspects of molecules. Subsequently, candidate genes were validated at both transcript and protein level. There were 314 DGs in patients with unruptured IA when compared to control samples. Out of these, SPARC and OSM were validated as candidate molecules in unruptured IA. PI3K-AKT signaling pathway was found to be an important pathway for the formation of IA. Similarly, 301 DGs were identified in the samples of ruptured IA when compared with unruptured IAs. CTSL was found to be a key candidate molecule which along with Hippo signaling pathway may be involved in the rupture of IA. We conclude that activation of PI3K-AKT signaling pathway by OSM along with up-regulation of SPARC is important for the formation of IA. Further, regulation of Hippo pathway through PI3K-AKT signaling results in the down-regulation of YAP1 gene. This along with up-regulation of CTSL leads to further weakening of aneurysm wall and its subsequent rupture.
Collapse
Affiliation(s)
- Munish Kumar
- Division of Neuro-anesthesia, Department of Anesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Krishna Patel
- Institute of Bioinformatics, International Tech Park, Bangalore, India
| | - Shobia Chinnapparaj
- Division of Neuro-anesthesia, Department of Anesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Tanavi Sharma
- Division of Neuro-anesthesia, Department of Anesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashish Aggarwal
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Navneet Singla
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhivanan Karthigeyan
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Apinderpreet Singh
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sushanta Kumar Sahoo
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manjul Tripathi
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aastha Takkar
- Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Tulika Gupta
- Department of Anatomy, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Arnab Pal
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Savita Verma Attri
- Pediatric Biochemistry, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Yogender Singh Bansal
- Department of Forensic Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Radha Kanta Ratho
- Department of Virology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sunil K Gupta
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhu Khullar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rakesh Kumar Vashishta
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Kanchan Kumar Mukherjee
- Department of Neurosurgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vinod Kumar Grover
- Division of Neuro-anesthesia, Department of Anesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajendra Prasad
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aditi Chatterjee
- Institute of Bioinformatics, International Tech Park, Bangalore, India
| | - Harsha Gowda
- Institute of Bioinformatics, International Tech Park, Bangalore, India
| | - Hemant Bhagat
- Division of Neuro-anesthesia, Department of Anesthesia and Intensive Care, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
36
|
Supradit K, Wongprasert K, Tangphatsornruang S, Yoocha T, Sonthirod C, Pootakham W, Thitapakorn V, Butthongkomvong K, Phanaksri T, Kunjantarachot A, Klongprateeppon H, Sattavacharavech P, Prasopdee S. microRNA profiling of exosomes derived from plasma and their potential as biomarkers for Opisthorchis viverrini-associated cholangiocarcinoma. Acta Trop 2024; 258:107362. [PMID: 39151716 DOI: 10.1016/j.actatropica.2024.107362] [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: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Cholangiocarcinoma (CCA) is a life-threatening disease that impacts patients worldwide. In Southeast Asian countries, the liver fluke Opisthorchis viverrini plays a major role in inducing carcinogenesis of the bile ducts. Due to its asymptomatic nature, O. viverrini infections are rarely treated, consequently leading to the development of advanced stages of CCA before diagnosis. Despite the current use of exosomal microRNAs (miRNA) as diagnostic biomarkers for the early detection of many types of cancer, the applications for miRNA remain limited with CCA. Circulating exosomes, membranous vesicles essential for intercellular communication, were found to contain unique miRNA. In this study, we conducted next-generation sequencing (Ion Torrent PGM) and bioinformatics to characterize and compare the contents of exosomal miRNA derived from the plasma of CCA patients, O. viverrini-infected patients, and healthy individuals, as well as to identify and validate key molecules as markers for screening the diagnosis of CCA and O. viverrini infection. The obtained results showed the success of using NGS technology in discovering exosomal miRNAs, specifically miR-194-5p and miR-192-5p, both of which were upregulated in the O. viverrini-infected group. Interestingly, miR-192-5p was upregulated while miR-194-5p was downregulated in CCA, suggesting their potential use as biomarkers for screening CCA and O. viverrini infection, especially in O. viverrini-endemic areas.
Collapse
Affiliation(s)
- Kittiya Supradit
- Department of Radiological technology, Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand; Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Kanokpan Wongprasert
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sithichoke Tangphatsornruang
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Thippawan Yoocha
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Chutima Sonthirod
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Wirulda Pootakham
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Veerachai Thitapakorn
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand; Research Unit in Opisthorchiasis, Cholangiocarcinoma, and Neglected Parasitic Diseases, Thammasat University, Pathum Thani, 12120, Thailand
| | | | - Teva Phanaksri
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | - Anthicha Kunjantarachot
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | | | | | - Sattrachai Prasopdee
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand; Research Unit in Opisthorchiasis, Cholangiocarcinoma, and Neglected Parasitic Diseases, Thammasat University, Pathum Thani, 12120, Thailand.
| |
Collapse
|
37
|
Gatasheh MK. Identifying key genes against rutin on human colorectal cancer cells via ROS pathway by integrated bioinformatic analysis and experimental validation. Comput Biol Chem 2024; 112:108178. [PMID: 39191167 DOI: 10.1016/j.compbiolchem.2024.108178] [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: 03/13/2024] [Revised: 08/04/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
Colorectal cancer (CRC) poses a significant global health challenge, characterized by substantial prevalence variations across regions. This study delves into the therapeutic potential of rutin, a polyphenol abundant in fruits, for treating CRC. The primary objectives encompass identifying molecular targets and pathways influenced by rutin through an integrated approach combining bioinformatic analysis and experimental validation. Employing Gene Set Enrichment Analysis (GSEA), the study focused on identifying potential differentially expressed genes (DEGs) associated with CRC, specifically those involved in regulating reactive oxygen species, metabolic reprogramming, cell cycle regulation, and apoptosis. Utilizing diverse databases such as GEO2R, CTD, and Gene Cards, the investigation revealed a set of 16 targets. A pharmacological network analysis was subsequently conducted using STITCH and Cytoscape, pinpointing six highly upregulated genes within the rutin network, including TP53, PCNA, CDK4, CCNEB1, CDKN1A, and LDHA. Gene Ontology (GO) analysis predicted functional categories, shedding light on rutin's potential impact on antioxidant properties. KEGG pathway analysis enriched crucial pathways like metabolic and ROS signaling pathways, HIF1a, and mTOR signaling. Diagnostic assessments were performed using UALCAN and GEPIA databases, evaluating mRNA expression levels and overall survival for the identified targets. Molecular docking studies confirmed robust binding associations between rutin and biomolecules such as TP53, PCNA, CDK4, CCNEB1, CDKN1A, and LDHA. Experimental validation included inhibiting colorectal cell HT-29 growth and promoting cell growth with NAC through MTT assay. Flow cytometric analysis also observed rutin-induced G1 phase arrest and cell death in HT-29 cells. RT-PCR demonstrated reduced expression levels of target biomolecules in HT-29 cells treated with rutin. This comprehensive study underscores rutin's potential as a promising therapeutic avenue for CRC, combining computational insights with robust experimental evidence to provide a holistic understanding of its efficacy.
Collapse
Affiliation(s)
- Mansour K Gatasheh
- Department of Biochemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
38
|
Durrani IA, John P, Bhatti A, Khan JS. Network medicine based approach for identifying the type 2 diabetes, osteoarthritis and triple negative breast cancer interactome: Finding the hub of hub genes. Heliyon 2024; 10:e36650. [PMID: 39281650 PMCID: PMC11401126 DOI: 10.1016/j.heliyon.2024.e36650] [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: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024] Open
Abstract
The increasing prevalence of multi-morbidities, particularly the incidence of breast cancer in diabetic/osteoarthritic patients emphasize on the need for exploring the underlying molecular mechanisms resulting in carcinogenesis. To address this, present study employed a systems biology approach to identify switch genes pivotal to the crosstalk between diseased states resulting in multi-morbid conditions. Hub genes previously reported for type 2 diabetes mellitus (T2DM), osteoarthritis (OA), and triple negative breast cancer (TNBC), were extracted from published literature and fed into an integrated bioinformatics analyses pipeline. Thirty-one hub genes common to all three diseases were identified. Functional enrichment analyses showed these were mainly enriched for immune and metabolism associated terms including advanced glycation end products (AGE) pathways, cancer pathways, particularly breast neoplasm, immune system signalling and adipose tissue. The T2DM-OA-TNBC interactome was subjected to protein-protein interaction network analyses to identify meta hub/clustered genes. These were prioritized and wired into a three disease signalling map presenting the enriched molecular crosstalk on T2DM-OA-TNBC axes to gain insight into the molecular mechanisms underlying disease-disease interactions. Deciphering the molecular bases for the intertwined metabolic and immune states may potentiate the discovery of biomarkers critical for identifying and targeting the immuno-metabolic origin of disease.
Collapse
Affiliation(s)
- Ilhaam Ayaz Durrani
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | - Peter John
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | - Attya Bhatti
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, 44000, Pakistan
| | | |
Collapse
|
39
|
Alghamdi KS, Kassar RH, Farrash WF, Obaid AA, Idris S, Siddig A, Shakoori AM, Alshehre SM, Minshawi F, Mujalli A. Key Disease-Related Genes and Immune Cell Infiltration Landscape in Inflammatory Bowel Disease: A Bioinformatics Investigation. Int J Mol Sci 2024; 25:9751. [PMID: 39273699 PMCID: PMC11396460 DOI: 10.3390/ijms25179751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/29/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024] Open
Abstract
Inflammatory Bowel Diseases (IBD), which encompass ulcerative colitis (UC) and Crohn's disease (CD), are characterized by chronic inflammation and tissue damage of the gastrointestinal tract. This study aimed to uncover novel disease-gene signatures, dysregulated pathways, and the immune cell infiltration landscape of inflamed tissues. Eight publicly available transcriptomic datasets, including inflamed and non-inflamed tissues from CD and UC patients were analyzed. Common differentially expressed genes (DEGs) were identified through meta-analysis, revealing 180 DEGs. DEGs were implicated in leukocyte transendothelial migration, PI3K-Akt, chemokine, NOD-like receptors, TNF signaling pathways, and pathways in cancer. Protein-protein interaction network and cluster analysis identified 14 central IBD players, which were validated using eight external datasets. Disease module construction using the NeDRex platform identified nine out of 14 disease-associated genes (CYBB, RAC2, GNAI2, ITGA4, CYBA, NCF4, CPT1A, NCF2, and PCK1). Immune infiltration profile assessment revealed a significantly higher degree of infiltration of neutrophils, activated dendritic cells, plasma cells, mast cells (resting/activated), B cells (memory/naïve), regulatory T cells, and M0 and M1 macrophages in inflamed IBD tissue. Collectively, this study identified the immune infiltration profile and nine disease-associated genes as potential modulators of IBD pathogenesis, offering insights into disease molecular mechanisms, and highlighting potential disease modulators and immune cell dynamics.
Collapse
Affiliation(s)
- Kawthar S Alghamdi
- Department of Biology, College of Science, University of Hafr Al Batin, Hafar Al-Batin 39511, Saudi Arabia
| | - Rahaf H Kassar
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Wesam F Farrash
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Ahmad A Obaid
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Shakir Idris
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Alaa Siddig
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Afnan M Shakoori
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Sallwa M Alshehre
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Faisal Minshawi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Abdulrahman Mujalli
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| |
Collapse
|
40
|
Sicairos B, Zhou J, Hu Z, Zhang Q, Shi WQ, Du Y. Proteomic analysis reveals the dominant effect of ipomoeassin F on the synthesis of membrane and secretory proteins in triple-negative breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605505. [PMID: 39131350 PMCID: PMC11312459 DOI: 10.1101/2024.07.28.605505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Ipomoeassin F (Ipom-F) is a natural compound with embedded carbohydrates that exhibits a potent cytotoxic effect on triple-negative breast cancer (TNBC) cells. The mechanism behind this selective potency remains unclear. To elucidate this mechanism, we analyzed the proteome profiles of the TNBC MDA-MB-231 cells after exposure to Ipom-F at different time points and increasing doses using a quantitative proteomic method. Our proteomic data demonstrate that the major effect of Ipom-F on MDA-MB-231 cells is the inhibition of membrane and secreted protein expression. Our proteomic data are consistent with the recently uncovered molecular mechanism of action of Ipom-F, which binds to Sec61-α and inhibits the co-translational import of proteins into the endoplasmic reticulum. We have defined a subset of membrane and secreted proteins particularly sensitive to Ipom-F. Analysis of the expression of these Ipom-F-sensitive proteins in cancer cell lines and breast cancer tissues demonstrates that some of these proteins are upregulated in TNBC cells. Thus, it is likely that TNBC cells may have adapted to the elevated levels of some proteins identified as sensitive to Ipom-F in this study; inhibition of the expression of these proteins leads to a crisis in proliferation and/or survival for the cells.
Collapse
Affiliation(s)
- Brihget Sicairos
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Jianhong Zhou
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Zhijian Hu
- Feinstein Institute for Medical Research, Northwell Health, 350 Community Dr., Manhasset, New York, 11030, USA
| | - Qingyang Zhang
- Department of Mathematical Sciences, University of Arkansas, Fayetteville, Arkansas 72701, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, Indiana 47306, USA
| | - Yuchun Du
- Department of Biological Sciences, University of Arkansas, Fayetteville, Arkansas 72701, USA
| |
Collapse
|
41
|
Bhardwaj M, Begum F, Singh D, Krupanidhi S, Yadav VK, Sahoo DK, Patel A, Singh S. Identification of Biomarkers Associated With Paget's Disease of Bone and Bone Metastasis From Breast Cancer Patients. Cancer Rep (Hoboken) 2024; 7:e70003. [PMID: 39233667 PMCID: PMC11375332 DOI: 10.1002/cnr2.70003] [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: 03/09/2024] [Revised: 05/22/2024] [Accepted: 08/11/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND The bone is among the most frequently chosen sites for the metastatic spread of breast cancer. The prediction of biomarkers for BM (Bone Metastasis) and PDB (Paget's disease of bone) initiated from breast cancer could be critically important in categorizing individuals with a higher risk and providing targeted treatment for PDB and BM. AIMS This research aims to investigate the common key candidate biomarkers that contribute to BM-BCa (Bone metastasis of breast cancer) and PDB by employing network decomposition and functional enrichment studies. METHODS AND RESULTS This research analyzed high-throughput transcriptome sequencing (RNA-Seq). For this work, the dataset (GSE121677) was downloaded from GEO (Gene Expression Omnibus), and DEGs were identified using Galaxy and R script 4.3. Using STRING (Search Tool for the Retrieval of Interacting Genes), high-throughput research created a protein-protein interaction network (PPIN). The BM-PDB-interactome was created using Cytoscape 3.9.1 and PDB biomarkers, with the top 3% DEGs from BM-BCa. Functional Enrichment Analysis (Funrich 3.1.3) and DAVID 6.8 performed functional and gene set enrichment analysis (GSEA) of putatively essential biomarkers. TCGA (The Cancer Genome Atlas) validated the discovered genes. Based on our research, we identified 1262 DEGs; among these DEGs, 431 genes were upregulated, and 831 genes were downregulated. During the third growth of the interactome, 20 more genes were pinned to the BM-PDB interactome. RAC2, PIAS1, EP300, EIF2S1, and LRP6 are among the additional 25% of genes identified to interact with the BM-PDB interactome. To corroborate the findings of the research presented, additional functional and gene set enrichment analyses have been performed. CONCLUSION Of the five reported genes (RAC2, PIAS1, EP300, EIF2S1, and LRP6), RAC2 was identified to function as the common key potential biomarker in the BM-PDB interactome analysis and validated by TCGA in the study presented.
Collapse
Affiliation(s)
- Mahima Bhardwaj
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research (Deemed to Be University), Guntur, Andhra Pradesh, India
| | - Farhana Begum
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research (Deemed to Be University), Guntur, Andhra Pradesh, India
| | - Duleswar Singh
- Department of Biotechnology, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Srirama Krupanidhi
- Department of Biotechnology, Vignan's Foundation for Science, Technology and Research (Deemed to Be University), Guntur, Andhra Pradesh, India
| | - Virendra Kumar Yadav
- Department of Biotechnology, Faculty of Energy Technology, Pandit Deendayal Energy University, Gandhinagar, Gujarat, India
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Ashish Patel
- Department of Lifesciences, Hemchandracharya North Gujarat University, Patan, Gujarat, India
| | - Sachidanand Singh
- Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, India
| |
Collapse
|
42
|
Caglar HO, Aytatli A, Barlak N, Aydin Karatas E, Tatar A, Sahin A, Karatas OF. Bioinformatics approach combined with experimental verification reveals OAS3 gene implicated in paclitaxel resistance in head and neck cancer. Head Neck 2024; 46:2178-2196. [PMID: 38752376 DOI: 10.1002/hed.27803] [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: 10/26/2023] [Revised: 04/18/2024] [Accepted: 05/05/2024] [Indexed: 08/09/2024] Open
Abstract
BACKGROUND This study aimed to identify a candidate gene associated with paclitaxel (PTX) resistance and to evaluate functionally its biological role in the PTX-resistant head and neck squamous cell carcinoma (HNSCC) cell lines and clinical specimens. METHODS Microarray data series containing samples of different types of cancers resistant to PTX were analyzed and then a candidate gene associated with PTX resistance was identified using various bioinformatics tools. After the suppression of the target gene expression, changes in cell viability and colony-forming ability were evaluated in PTX-resistant FaDu and SCC-9 cell lines. RESULTS Bioinformatics analyses of upregulated genes in PTX-resistant cancer cells indicated that OAS3 was associated with PTX resistance. The downregulation of OAS3 expression significantly reduced the viability and colony-forming capacity of PTX-resistant SCC-9 cells by inducing apoptosis and cell cycle arrest at G0/G1 phase. CONCLUSIONS The therapeutic targeting of OAS3 may resensitize PTX-resistant HNSCC cells with high OAS3 expression to PTX treatment.
Collapse
Affiliation(s)
- Hasan Onur Caglar
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
| | - Abdulmelik Aytatli
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
- Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| | - Neslisah Barlak
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
- Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| | - Elanur Aydin Karatas
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
- Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| | - Arzu Tatar
- Department of Otorhinolaryngology Diseases, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Abdulkadir Sahin
- Department of Otorhinolaryngology Diseases, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Omer Faruk Karatas
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, Erzurum, Turkey
- Molecular Cancer Biology Laboratory, High Technology Application and Research Center, Erzurum Technical University, Erzurum, Turkey
| |
Collapse
|
43
|
Bagheri-Hosseinabadi Z, Eshkevari SMS, Khalighfard S, Alizadeh AM, Khori V, Amiriani T, Poorkhani A, Sadani S, Esmati E, Lashgari M, Mahmoodi M, Hajizadeh MR. A systematic approach introduced some immune system targets in rectal cancer by considering cell-free DNA methylation in response to radiochemotherapy. Cytokine 2024; 181:156666. [PMID: 38906038 DOI: 10.1016/j.cyto.2024.156666] [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: 03/03/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND This study aims to investigate cell-free DNA (cfDNA) methylation of genes involved in some immune system targets as biomarkers of radioresistance in patients with non-metastatic rectal cancer. METHODS Gene expression (GSE68204, GPL6480, and GSE15781) and DNA methylation profiles (GSE75548 and GSE139404) of rectal cancer patients were obtained from the Gene Expression Omnibus (GEO) database. GEO2R and FunRich software were first used to identify genes with significant expression differences. Enricher softwer was then used to analyze Gene Ontology and detect pathway enrichment of hub genes. Blood samples were then taken from 43 rectal cancer patients. After cfDNA extraction from samples, it was treated with bisulfite and analyzed by methylation-specific PCR. RESULTS 1088 genes with high and 629 with low expression were identified by GEO2R and FunRich software. A total of five high-expression hub genes, including CDH24, FGF18, CCND1, IFITM1, UBE2V1, and three low-expression hub genes, including CBLN2, VIPR2, and IRF4, were identified from UALCAN and DNMIVD databases. Methylation-specific PCR indicated a significant difference in hub gene methylation between cancerous and non-cancerous individuals. Radiochemotherapy significantly affected hub gene methylation. There was a considerable difference in the methylation rate of hub genes between patients who responded to radiochemotherapy and those who did not. CONCLUSIONS Evaluating gene methylation patterns might be an appropriate diagnostic tool to predict radiochemotherapy response and develop targeted therapeutic agents.
Collapse
Affiliation(s)
- Zahra Bagheri-Hosseinabadi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | | | - Ali Mohammad Alizadeh
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran; Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Amirhoushang Poorkhani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Somayeh Sadani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ebrahim Esmati
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzih Lashgari
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mahmoodi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Hajizadeh
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
44
|
Sailer AL, Jevtic Z, Stoll B, Wörtz J, Sharma K, Urlaub H, Dyall-Smith M, Pfeiffer F, Marchfelder A, Lenz C. Iron starvation results in up-regulation of a probable Haloferax volcanii siderophore transporter. Front Microbiol 2024; 15:1422844. [PMID: 39206359 PMCID: PMC11349517 DOI: 10.3389/fmicb.2024.1422844] [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: 04/24/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The response of the haloarchaeal model organism Haloferax volcanii to iron starvation was analyzed at the proteome level by data-independent acquisition mass spectrometry. Cells grown in minimal medium with normal iron levels were compared to those grown under low iron conditions, with samples being separated into membrane and cytoplasmic fractions in order to focus on import/export processes which are frequently associated with metal homeostasis. Iron starvation not only caused a severe retardation of growth but also altered the levels of many proteins. Using a comprehensive annotated spectral library and data-independent acquisition mass spectrometry (DIA-MS), we found that iron starvation resulted in significant changes to both the membrane and the soluble proteomes of Hfx. volcanii. The most affected protein is the RND family permease HVO_A0467, which is 44-fold enriched in cells grown under iron starvation. The gene HVO_A0467 can be deleted suggesting that it is not essential under standard conditions. Compared to wild type cells the deletion strain shows only slight changes in growth and cell morphologies show no differences. Molecular docking predictions indicated that HVO_A0467 may be an exporter of the siderophore schizokinen for which a potential biosynthesis cluster is encoded in the Hfx. volcanii genome. Together, these findings confirm the importance of iron for archaeal cells and suggest HVO_0467 as a siderophore exporter.
Collapse
Affiliation(s)
| | - Zivojin Jevtic
- Department of Biomedicine, University Children’s Hospital, University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Kundan Sharma
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics Group, Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Mike Dyall-Smith
- Computational Systems Biochemistry, Max Planck Institute for Biochemistry, Martinsried, Germany
- Veterinary Biosciences, Faculty of Science, Melbourne Veterinary School, University of Melbourne, Parkville, VIC, Australia
| | - Friedhelm Pfeiffer
- Biology II, Ulm University, Ulm, Germany
- Computational Systems Biochemistry, Max Planck Institute for Biochemistry, Martinsried, Germany
| | | | - Christof Lenz
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics Group, Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
45
|
Zertuche-Martínez C, Velázquez-Enríquez JM, González-García K, Santos-Álvarez JC, Romero-Tlalolini MDLÁ, Pina-Canseco S, Pérez-Campos Mayoral L, Muriel P, Villa-Treviño S, Baltiérrez-Hoyos R, Arellanes-Robledo J, Vásquez-Garzón VR. Discovery of candidate biomarkers from plasma-derived extracellular vesicles of patients with cirrhosis and hepatocellular carcinoma: an exploratory proteomic study. Mol Omics 2024; 20:483-495. [PMID: 39011654 DOI: 10.1039/d4mo00043a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Extracellular vesicles (EVs) represent an attractive source of biomarkers due to their biomolecular cargo. The aim of this study was to identify candidate protein biomarkers from plasma-derived EVs of patients with liver cirrhosis (LC) and hepatocellular carcinoma (HCC). Plasma-derived EVs from healthy participants (HP), LC, and HCC patients (eight samples each) were subjected to label-free quantitative proteomic analysis using LC-MS/MS. A total of 248 proteins were identified, and differentially expressed proteins (DEPs) were obtained after pairwise comparison. We found that DEPs mainly involve complement cascade activation, coagulation pathways, cholesterol metabolism, and extracellular matrix components. By choosing a panel of up- and down-regulated proteins involved in cirrhotic and carcinogenesis processes, TGFBI, LGALS3BP, C7, SERPIND1, and APOC3 were found to be relevant for LC patients, while LRG1, TUBA1C, TUBB2B, ACTG1, C9, HP, FGA, FGG, FN1, PLG, APOB and ITIH2 were associated with HCC patients, which could discriminate both diseases. In addition, we identified the top shared proteins in both diseases, which included LCAT, SERPINF2, A2M, CRP, and VWF. Thus, our exploratory proteomic study revealed that these proteins might play an important role in the disease progression and represent a panel of candidate biomarkers for the prognosis and diagnosis of LC and HCC.
Collapse
Affiliation(s)
- Cecilia Zertuche-Martínez
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Karina González-García
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | | | - Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina UNAM UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Laura Pérez-Campos Mayoral
- Centro de Investigación Facultad de Medicina UNAM UABJO, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico
| | - Pablo Muriel
- Laboratorio de Hepatología Experimental, Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07000, Mexico
| | - Saúl Villa-Treviño
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07360, Mexico
| | - Rafael Baltiérrez-Hoyos
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico.
| | | | - Verónica Rocío Vásquez-Garzón
- CONAHCYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Oaxaca, Mexico.
| |
Collapse
|
46
|
Vassileff N, Spiers JG, Lee JD, Woodruff TM, Ebrahimie E, Mohammadi Dehcheshmeh M, Hill AF, Cheng L. A Panel of miRNA Biomarkers Common to Serum and Brain-Derived Extracellular Vesicles Identified in Mouse Model of Amyotrophic Lateral Sclerosis. Mol Neurobiol 2024; 61:5901-5915. [PMID: 38252383 PMCID: PMC11249427 DOI: 10.1007/s12035-023-03857-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron disease characterised by the deposition of aggregated proteins including TAR DNA-binding protein 43 (TDP-43) in vulnerable motor neurons and the brain. Extracellular vesicles (EVs) facilitate the spread of neurodegenerative diseases and can be easily accessed in the bloodstream. This study aimed to identify a panel of EV miRNAs that can capture the pathology occurring in the brain and peripheral circulation. EVs were isolated from the cortex (BDEVs) and serum (serum EVs) of 3 month-old and 6-month-old TDP-43*Q331K and TDP-43*WT mice. Following characterisation and miRNA isolation, the EVs underwent next-generation sequencing where 24 differentially packaged miRNAs were identified in the TDP-43*Q331K BDEVs and 7 in the TDP-43*Q331K serum EVs. Several miRNAs, including miR-183-5p, were linked to ALS. Additionally, miR-122-5p and miR-486b-5p were identified in both panels, demonstrating the ability of the serum EVs to capture the dysregulation occurring in the brain. This is the first study to identify miRNAs common to both the serum EVs and BDEVs in a mouse model of ALS.
Collapse
Affiliation(s)
- Natasha Vassileff
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Jereme G Spiers
- Clear Vision Research, Eccles Institute of Neuroscience, John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, ACT, Australia
- School of Medicine and Psychology, College of Health and Medicine, The Australian National University, Acton, ACT, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Esmaeil Ebrahimie
- Genomics Research Platform, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, 3000, Australia
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, SA, 5371, Australia
- School of BioSciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | | | - Andrew F Hill
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
- Institute for Health and Sport, Victoria University, Footscray, Victoria, Australia
| | - Lesley Cheng
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
47
|
Rice SJ, Belani CP. Characterization of effective, simple, and low-cost precipitation methods for depleting abundant plasma proteins to enhance the depth and breadth of plasma proteomics. Proteomics 2024; 24:e2400071. [PMID: 38700387 DOI: 10.1002/pmic.202400071] [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: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Plasma is an abundant source of proteins and potential biomarkers to aid in the detection, diagnosis, and prognosis of human diseases. These proteins are often present at low levels in the blood and difficult to identify and measure due to the large dynamic range of proteins. The goal of this work was to characterize and compare various protein precipitation methods related to how they affect the depth and breadth of plasma proteomic studies. Abundant protein precipitation with perchloric acid (PerCA) can increase protein identifications and depth of plasma proteomic studies. Three acid- and four solvent-based precipitation methods were evaluated. All methods tested provided excellent plasma proteomic coverage (>600 identified protein groups) and detected protein in the low pg/mL range. Functional enrichment analysis revealed subtle differences within and larger changes between the precipitant groups. Methanol-based precipitation outperformed the other methods based on identifications and reproducibility. The methods' performance was verified using eight lung cancer patient samples, where >700 protein groups were measured and proteins with an estimated plasma concentration of ∼10 pg/mL were detected. Various protein precipitation agents are amenable to extending the depth and breadth of plasma proteomes. These data can guide investigators to implement inexpensive, high-throughput methods for their plasma proteomic workflows.
Collapse
Affiliation(s)
- Shawn J Rice
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Chandra P Belani
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
48
|
Naveed M, Ali I, Aziz T, Javed K, Saleem A, Hanif N, Alharbi M. Investigating the anti-cancer compounds from Calliandra harrisii for precision medicine in pancreatic cancer via in-silico drug design and GC-MS analysis. Z NATURFORSCH C 2024; 79:209-220. [PMID: 38635803 DOI: 10.1515/znc-2024-0057] [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: 03/12/2024] [Accepted: 04/06/2024] [Indexed: 04/20/2024]
Abstract
Pancreatic cancer is a fatal illness caused by mutations in multiple genes. Pancreatic cancer damages the organ that helps in digestion, resulting in symptoms including fatigue, bloating, and nausea. The use of medicinal plants has been crucial in the treatment of numerous disorders. The medicinal plant Calliandra Harrisi has been widely exploited for its possibilities in biology and medicine. The current study aimed to assess the biopotential of biologically active substances against pancreatic cancer. The GC-MS data of these phytochemicals from Calliandra Harrisi were further subjected to computational approaches with pancreatic cancer genes to evaluate their potential as therapeutic candidates. Molecular docking analysis revealed that N-[Carboxymethyl] maleamic acid is the leading molecule responsible for protein denaturation inhibition, having the highest binding affinity of 6.8 kJ/mol among all other compounds with KRAS inflammatory proteins. Furthermore, ADMET analysis and Lipinski's rule validation were also performed revealing its higher absorption in the gastrointestinal tract. The results of the hepatotoxicity test demonstrated that phytochemicals are non-toxic, safe to use, and do not cause necrosis, fibrosis, or vacuolar degeneration even at excessive levels. Calliandra Harrisi has phytoconstituents that have a variety of pharmacological uses in consideration.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, 66901 University of Central Punjab , Lahore 54590, Pakistan
| | - Imran Ali
- Department of Biotechnology, Faculty of Science and Technology, 66901 University of Central Punjab , Lahore 54590, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, 37796 University of Ioannina , Arta 47132, Greece
| | - Khushbakht Javed
- Department of Biotechnology, Faculty of Science and Technology, 66901 University of Central Punjab , Lahore 54590, Pakistan
| | - Ayesha Saleem
- Department of Biotechnology, Faculty of Science and Technology, 66901 University of Central Punjab , Lahore 54590, Pakistan
| | - Nimra Hanif
- Department of Biotechnology, Faculty of Science and Technology, 66901 University of Central Punjab , Lahore 54590, Pakistan
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, 37850 King Saud University , P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
49
|
Gudivada IP, Amajala KC. Integrative Bioinformatics Analysis for Targeting Hub Genes in Hepatocellular Carcinoma Treatment. Curr Genomics 2024; 26:48-80. [PMID: 39911278 PMCID: PMC11793067 DOI: 10.2174/0113892029308243240709073945] [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: 03/22/2024] [Revised: 04/24/2024] [Accepted: 06/11/2024] [Indexed: 02/07/2025] Open
Abstract
Background The damage in the liver and hepatocytes is where the primary liver cancer begins, and this is referred to as Hepatocellular Carcinoma (HCC). One of the best methods for detecting changes in gene expression of hepatocellular carcinoma is through bioinformatics approaches. Objective This study aimed to identify potential drug target(s) hubs mediating HCC progression using computational approaches through gene expression and protein-protein interaction datasets. Methodology Four datasets related to HCC were acquired from the GEO database, and Differentially Expressed Genes (DEGs) were identified. Using Evenn, the common genes were chosen. Using the Fun Rich tool, functional associations among the genes were identified. Further, protein-protein interaction networks were predicted using STRING, and hub genes were identified using Cytoscape. The selected hub genes were subjected to GEPIA and Shiny GO analysis for survival analysis and functional enrichment studies for the identified hub genes. The up-regulating genes were further studied for immunohistopathological studies using HPA to identify gene/protein expression in normal vs HCC conditions. Drug Bank and Drug Gene Interaction Database were employed to find the reported drug status and targets. Finally, STITCH was performed to identify the functional association between the drugs and the identified hub genes. Results The GEO2R analysis for the considered datasets identified 735 upregulating and 284 downregulating DEGs. Functional gene associations were identified through the Fun Rich tool. Further, PPIN network analysis was performed using STRING. A comparative study was carried out between the experimental evidence and the other seven data evidence in STRING, revealing that most proteins in the network were involved in protein-protein interactions. Further, through Cytoscape plugins, the ranking of the genes was analyzed, and densely connected regions were identified, resulting in the selection of the top 20 hub genes involved in HCC pathogenesis. The identified hub genes were: KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Further, GEPIA and Shiny GO analyses provided insights into survival ratios and functional enrichment studied for the hub genes. The HPA database studies further found that upregulating genes were involved in changes in protein expression in Normal vs HCC tissues. These findings indicated that hub genes were certainly involved in the progression of HCC. STITCH database studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, could be used as leads to identify novel drugs, and identified hub genes could also be considered as potential and promising drug targets as they are involved in the gene-chemical interaction networks. Conclusion The present study involved various integrated bioinformatics approaches, analyzing gene expression and protein-protein interaction datasets, resulting in the identification of 20 top-ranked hubs involved in the progression of HCC. They are KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Gene-chemical interaction network studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, can be used as leads to identify novel drugs, and the identified hub genes can be promising drug targets. The current study underscores the significance of targeting these hub genes and utilizing existing molecules to generate new molecules to combat liver cancer effectively and can be further explored in terms of drug discovery research to develop treatments for HCC.
Collapse
Affiliation(s)
- Indu Priya Gudivada
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Krishna Chaitanya Amajala
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| |
Collapse
|
50
|
Zhu Y, Chen X, Tang R, Li G, Yang J, Hong S. Comprehensive analysis of hub genes associated with cisplatin-resistance in ovarian cancer and screening of therapeutic drugs through bioinformatics and experimental validation. J Ovarian Res 2024; 17:142. [PMID: 38987777 PMCID: PMC11234624 DOI: 10.1186/s13048-024-01461-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: 12/12/2023] [Accepted: 06/18/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND To identify key genes associated with cisplatin resistance in ovarian cancer, a comprehensive analysis was conducted on three datasets from the GEO database and through experimental validation. METHODS Gene expression profiles were retrieved from the GEO database. DEGs were identified by comparing gene expression profiles between cisplatin-sensitive and resistant ovarian cancer cell lines. The identified genes were further subjected to GO, KEGG, and PPI network analysis. Potential inhibitors of key genes were identified through methods such as LibDock nuclear molecular docking. In vitro assays and RT-qPCR were performed to assess the expression levels of key genes in ovarian cancer cell lines. The sensitivity of cells to chemotherapy and proliferation of key gene knockout cells were evaluated through CCK8 and Clonogenic assays. RESULTS Results showed that 12 genes influenced the chemosensitivity of the ovarian cancer cell line SKOV3, and 9 genes were associated with the prognosis and survival outcomes of ovarian cancer patients. RT-qPCR results revealed NDRG1, CYBRD1, MT2A, CNIH3, DPYSL3, and CARMIL1 were upregulated, whereas ERBB4, ANK3, B2M, LRRTM4, EYA4, and SLIT2 were downregulated in cisplatin-resistant cell lines. NDRG1, CYBRD1, and DPYSL3 knock-down significantly inhibited the proliferation of cisplatin-resistant cell line SKOV3. Finally, photofrin, a small-molecule compound targeting CYBRD1, was identified. CONCLUSION This study reveals changes in the expression level of some genes associated with cisplatin-resistant ovarian cancer. In addition, a new small molecule compound was identified for the treatment of cisplatin-resistant ovarian cancer.
Collapse
Affiliation(s)
- Yunshan Zhu
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Xuehong Chen
- Hospital Department of Obstetrics and Gynecology, Linhai Second People's Hospital, TaiZhou, 317016, China
| | - Rongrong Tang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Guangxiao Li
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China
| | - Jianhua Yang
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China.
| | - Shihao Hong
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, 310016, China.
| |
Collapse
|