{"title":"Pathogenic Mechanism of the Lactylation-Related Gene DCBLD1 in Ulcerative Colitis: A Multi-Omics and Machine Learning Analysis.","authors":"Changan Chen, Yuping Yang, Tingmei Yang, Caiyuan Yu, Guixia Zhang, Lijiao Cui, Yu Zhou, Zhenkai Li, Zihang Hu, Yijie Weng","doi":"10.2174/0113862073403664250911055311","DOIUrl":"10.2174/0113862073403664250911055311","url":null,"abstract":"<p><strong>Background: </strong>The incidence of ulcerative colitis (UC) has been steadily increasing in recent years. Current treatments are only effective for some patients, highlighting the need to find novel therapeutic targets. Lactylation, a post-translational modification, remains poorly understood in UC. This study examines the role of the lactylation-related gene DCBLD1 in the pathogenesis of UC through multi-omics analysis.</p><p><strong>Methods: </strong>Summary-data-based Mendelian Randomization (SMR) analysis identified DCBLD1 as a lactylation-related gene associated with UC risk. Single-cell RNA sequencing (scRNA-seq) examined DCBLD1 expression in UC and healthy intestinal tissues, coupled with cellular communication, metabolic pathway, KEGG enrichment, and GO annotations. Diagnostic models were built based on differential expression between DCBLD1+ and DCBLD1- epithelial cells. In addition, RNA sequencing (RNA-seq) was used for analysis. Ultimately, qPCR was performed to validate DCBLD1 expression.</p><p><strong>Results: </strong>SMR demonstrated that DCBLD1 positively correlated with UC risk. scRNA-seq revealed that DCBLD1+ epithelial cells exhibited enhanced cellular communication and metabolic activity. Seventeen hub genes were screened for machine learning, yielding AUC values of 0.69 (CATboost), 0.63 (XGBoost), and 0.55 (NGboost) in the test set. RNA-seq confirmed the association of DCBLD1 with immune responses. qPCR confirmed elevated DCBLD1 expression in UC tissues versus controls.</p><p><strong>Discussion: </strong>Intestinal epithelial cells expressing DCBLD1 may promote inflammation in UC by lactylation, regulating immunometabolism, and participating in immunological responses, all of which require further investigation in the future.</p><p><strong>Conclusion: </strong>DCBLD1 may promote UC progression through lactylation, immune-metabolic regulation, and involvement in immune responses, serving as a potential therapeutic target.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145063440","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Exploring the Protective Effect and Potential Mechanism of Acutumidine on Myocardial Ischemia based on \"Compound-Target-Pathway\" Network.","authors":"Jia Shao, Rongrong Zheng, Meng Liu, Yingying Yu, Ling Luo, Jinxia Wei","doi":"10.2174/0113862073399707250827110100","DOIUrl":"https://doi.org/10.2174/0113862073399707250827110100","url":null,"abstract":"<p><strong>Introduction: </strong>Menispermi Rhizoma is a traditional Chinese medicine with significant Anti-Myocardial Ischemia (MI) effects. Acutumidine is a major alkaloid component of Menispermi Rhizoma. However, the effectiveness and potential mechanism of acutumidine in treating MI have been rarely studied. This research aims to explore the effect and mechanism of acutumidine on MI.</p><p><strong>Methods: </strong>The function and mechanism of acutumidine in ameliorating MI were investigated via a comprehensive strategy of experimental evaluation, network pharmacology, and molecular docking. Firstly, the oxygen glucose deprivation (OGD) model of H9c2 cardiomyocytes was established to confirm the effects of acutumidine on MI. Then, network pharmacology was used to predict the potential targets and mechanisms of acutumidine in MI. The intersection targets between acutumidine and MI were acquired and used to construct a protein-protein interaction network. GO and KEGG enrichment analyses were performed using the Metascape database to reveal the probable mechanism of acutumidine on MI. Finally, the key potential targets of acutumidine were validated by molecular docking.</p><p><strong>Results: </strong>Cell experiments showed that acutumidine protected H9c2 cells against OGD injury by increasing SOD and GSH levels, decreasing LDH, CK, and MDA levels, and reducing apoptosis rates. Network pharmacology showed that the protective effect of acutumidine on MI was related to PI3K/AKT, HIF-1, and Ras signaling pathways. Molecular docking studies further showed that MAPK1, ESR1, EGFR, IGF1, and CASP3 are the core targets of acutumidine in treating MI.</p><p><strong>Discussions: </strong>All research results suggested that acutumidine could inhibit oxidative stress and cell apoptosis.</p><p><strong>Conclusions: </strong>Acutumidine exhibits significant effects on MI, exerting pharmacological effects through multiple targets and pathways.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145063393","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Mechanisms of Propolis Ethanol Extracts to Alleviate Sarcopenia based on Network Pharmacology and Experimental Validation.","authors":"Songhao Tian, Tao Chen, Congying Song, Hongru Guo, Conglin Jiang, Wei Feng","doi":"10.2174/0113862073397434250731053716","DOIUrl":"https://doi.org/10.2174/0113862073397434250731053716","url":null,"abstract":"<p><strong>Introduction: </strong>Sarcopenia (Sar) is an age-related loss of muscle mass and function. Propolis, a natural product with anti-inflammatory properties, may help prevent Sar, but its active components and mechanisms remain unclear.</p><p><strong>Methods: </strong>Network pharmacology identified intersecting targets of propolis ethanol extract (PEE) and Sar. PPI and CTP networks highlighted key compounds and targets, verified by molecular docking. In vitro, apigenin (Ap), the predicted main compound, was tested on D-galactoseinduced senescent C2C12 myoblasts via cell viability and Western blotting.</p><p><strong>Results: </strong>Twelve overlapping targets were identified between PEE and Sar, with TNFα and IL6 highlighted as hub targets. Network analysis determined Ap as the main active compound. Molecular docking revealed strong binding affinities of Ap with TNFα and IL6. In vitro experiments demonstrated that Ap significantly enhanced the viability and differentiation of senescent C2C12 cells, downregulated TNFα and IL6 expression, and inhibited JAK2 and STAT3 phosphorylation, indicating suppression of the JAK-STAT signaling pathway.</p><p><strong>Discussion: </strong>The findings suggest that PEE, primarily through Ap, alleviates Sar by targeting inflammatory pathways and suppressing JAK-STAT signaling, thereby promoting muscle regeneration. The integration of network pharmacology, molecular docking, and in vitro validation provides mechanistic insights supporting the therapeutic potential of PEE in Sar. Limitations include the absence of in vivo confirmation, which warrants further animal and clinical studies to validate these effects and explore translational applications.</p><p><strong>Conclusion: </strong>This study identifies Ap as the key active compound in PEE that alleviates Sar by downregulating TNFα and IL6 and inhibiting the JAK-STAT pathway. The results provide a molecular basis for the use of propolis as a natural intervention for Sar and support its development as a functional food or therapeutic agent targeting age-related muscle degeneration.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144999824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Acupoint Catgut Embedding Alleviates Neuropathic Pain by Regulating Sigma-1 Receptor Expression.","authors":"Heng-Tao Xie, Xiao-Bo Feng, Kai-Rong Du","doi":"10.2174/0113862073377826250728055125","DOIUrl":"https://doi.org/10.2174/0113862073377826250728055125","url":null,"abstract":"<p><strong>Introduction: </strong>Acupoint Catgut Embedding (ACE), also known as acupuncture catgut implantation, exerts analgesic effects by inhibiting Sig-1R. This study aimed to evaluate the modulatory effect of ACE on Sig-1R and its mechanism of action in alleviating nerve pain.</p><p><strong>Methods: </strong>We assessed behavioral changes in mechanosensitive and thermosensitive pain in rats. Spinal cord tissue damage was examined using HE staining, while apoptosis was evaluated through TUNEL staining. Sig-1R expression in spinal cord tissue was analyzed via immunohistochemistry.</p><p><strong>Results: </strong>ACE and Sig-1R antagonists significantly reduced paw withdrawal frequency (PWF), decreased the expression of Bax and cleaved caspase-3 proteins, and alleviated morphological damage in spinal cord cells. They also increased the expression of Bcl-2 and prolonged paw withdrawal latency (PWL) in rats. Additionally, ACE and Sig-1R antagonists reduced levels of TNF-α, IL-1β, and IL-6, as well as malondialdehyde (MDA), while elevating levels of Superoxide Dismutase (SOD) and Glutathione Peroxidase (GPx) in both serum and spinal cord tissues. Furthermore, they downregulated the protein expression of p-ERK1/2, p38 MAPK, and Nox2, reduced the number of Th1 and Th17 cells, and increased the number of Th2 and Treg cells.</p><p><strong>Discussion: </strong>Currently, the mechanism of action of ACE on neuropathic pain caused by peripheral nerve injury based on Sig-1R is still unclear. This study evaluated the mechanism by which ACE alleviates neuralgia by regulating the expression of Sig-1R in the spinal cord. In future work, we aim to conduct additional experiments to determine the precise localization of T cells within the spinal cord and to further investigate their direct interactions with glial cells.</p><p><strong>Conclusion: </strong>ACE effectively alleviates nerve pain by modulating Sig-1R expression in the spinal cord, thereby regulating inflammatory responses, oxidative stress, and associated signaling pathways.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144945601","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ming Chen, Wenjian Huang, Yongyan Huang, Mushi Chen, Guobiao Yan
{"title":"RuAn Tablets Relieve Mammary Gland Hyperplasia by Regulating PGE2 Synthesis.","authors":"Ming Chen, Wenjian Huang, Yongyan Huang, Mushi Chen, Guobiao Yan","doi":"10.2174/0113862073391606250728082957","DOIUrl":"https://doi.org/10.2174/0113862073391606250728082957","url":null,"abstract":"<p><strong>Introduction: </strong>RuAn Tablets (RATs) are a traditional Chinese formulation used for the treatment of mammary gland hyperplasia (MGH), one of the most prevalent gynecological diseases. However, the complexity of RATs composition has hindered the elucidation of the mechanism.</p><p><strong>Methods: </strong>In this study, we employed an integrated approach combining ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS), network pharmacology, and experimental validation to identify the active components of RATs and explore their potential mechanisms in alleviating MGH.</p><p><strong>Results: </strong>Through UHPLC-MS analysis and database screening (SymMap and TCMSP), four active compounds were screened, including Betulinic acid (BA), Dioscin, Icariside I, and Neohesperidin. These compounds were predicted to interact with 321 potential targets, of which 22 were identified as key targets for RATs in MGH intervention. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed that these targets were primarily associated with prostaglandin synthesis and metabolism, particularly the synthesis of prostaglandin E2 (PGE2). Molecular docking studies demonstrated strong binding affinities between the active compounds and core target proteins, including PTGS2, EGFR, and TP53. Experimental validation further confirmed that RATs significantly reduced PGE2 levels in mammary proliferating cells by over 60%.</p><p><strong>Discussion: </strong>Integrating findings from network pharmacology and experimental validation suggests that RATs exert their therapeutic effects on MGH primarily by regulating PGE2 synthesis.</p><p><strong>Conclusion: </strong>In summary, this study provides a comprehensive understanding of the molecular mechanisms underlying RATs' efficacy and highlights their potential as a therapeutic agent for MGH.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144945671","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Hua-Zhuo-Ning-Fu Decoction Ameliorates Psoriasis by Inhibiting TNF-Α/IL-6 and PI3K/AKT Signaling Pathway: A Network Pharmacology Approach and Experimental Validation.","authors":"Zhuzhu Wu, Wenke Ma, Zitong Guan, Mengxue Han, Shuai Man, Zhenguo Wang","doi":"10.2174/0113862073390038250723201614","DOIUrl":"https://doi.org/10.2174/0113862073390038250723201614","url":null,"abstract":"<p><strong>Introduction: </strong>Hua-Zhuo-Ning-Fu decoction (HZD) is a traditional Chinese medicine prescription that has been clinically used by Chinese medical master Wang Xinlu for treating psoriasis. However, the specific molecular mechanisms remain unclear.</p><p><strong>Methods: </strong>To identify the effective compounds of HZD and psoriasis-related genes, we conducted comprehensive searches in public databases, including TCMSP, SwissTargetPrediction, Gene Cards, and OMIM. Based on the degree values, core genes of HZD against psoriasis were determined. Furthermore, the affinity energy between the active compounds of HZD and their core targets was validated via molecular docking. Finally, the anti-psoriasis effects and potential mechanisms of HZD were examined in M5-stimulated HaCaT cells in vitro and IMQ-induced psoriasis mice in vivo.</p><p><strong>Results: </strong>Network pharmacological analysis of HZD for psoriasis treatment identified 43 active components and 243 targets. Topological and molecular docking analyses identified interleukin (IL)-6 and tumor necrosis factor-α (TNF-α) as core targets for its anti-psoriasis effects. Specifically, the docking energy of isovitexin with IL-6 was lower (-7.30 kcal/mol), and that of baicalin with TNF-α was lower (-6.70 kcal/mol). KEGG analysis revealed that the main pathway was the PI3K/AKT pathway. HZD inhibited cell viability, inflammation, and oxidative stress in M5- induced HaCaT cells. Animal experiments demonstrated that HZD alleviated psoriatic dermatitis, histopathological features, and inflammation in IMQ-induced mice with psoriatic plaques. Notably, HZD inhibited the expression of TNF-α and IL-6 and the activation of the PI3K/AKT pathway both in vivo and in vitro.</p><p><strong>Discussion: </strong>Specific upstream/downstream regulators of the PI3K/AKT axis regulated by HZD still need to be explored. Further investigation is essential to clarify the functional relationship between the predicted targets and active components.</p><p><strong>Conclusion: </strong>In summary, HZD potentially mitigated inflammatory responses by targeting the TNF-α and IL-6 proteins, interfered with the PI3K/AKT pathway, and consequently drove the anti-psoriatic effect in IMQ-induced mice. Our findings provide a theoretical basis for HZD's clinical use in psoriasis treatment.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144945591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Novel Benzimidazole-Thiazolidine-2,4-dione Hybrid Compounds as Promising PPAR-γ Modulators with Antidiabetic Activity: Design, Synthesis, Molecular Docking, and In Vitro Evaluation.","authors":"Jisha Prems, Anton Smith A, Lal Prasanth M L","doi":"10.2174/0113862073403893250805011749","DOIUrl":"https://doi.org/10.2174/0113862073403893250805011749","url":null,"abstract":"<p><strong>Introduction: </strong>Design, synthesis, and molecular docking studies of a novel series of Benzimidazole-Thiazolidine-2,4-dione hybrid compounds as PPAR-γ modulators for antidiabetic activity. Thiazolidinediones function as agonists of PPAR-γ, influencing the expression of numerous genes involved in the regulation of glucose, lipid, and protein metabolism. They improve insulin sensitivity by promoting glucose uptake in adipose tissue and skeletal muscle, and by reducing hepatic gluconeogenesis.</p><p><strong>Objectives: </strong>To perform the synthesis, molecular docking studies, in vitro cytotoxicity assessment, glucose uptake assay, and in vitro PPAR-γ transcription factor assay of Benzimidazole- Thiazolidine-2,4-dione hybrid compounds as antidiabetic agents.</p><p><strong>Methods: </strong>A molecular hybridization strategy was employed for the synthesis of Benzimidazole- Thiazolidine-2,4-dione hybrid compounds, and their chemical identities were confirmed using 1H-NMR, 13C-NMR, and LC-MS techniques. Molecular docking studies were conducted to analyze the binding affinities of the synthesized analogs with the target protein. The MTT assay was used to evaluate the cytotoxic effects of the new compounds, which were subsequently tested for glucose uptake using rat L6 myotubes as a model. Furthermore, the compounds underwent an in vitro PPAR-γ transcription factor assay.</p><p><strong>Results: </strong>Synthesized compounds were screened for their activation potential of PPAR-γ expression in HepG2 cell lines, and several compounds, notably PT14 and PT18, displayed an activation potential higher than the reference standard pioglitazone.</p><p><strong>Discussion: </strong>Compounds PT18 and PT14 exhibited significant PPAR-γ expression in comparison to the commercially available PPAR-γ agonist pioglitazone, at a concentration of 1μM which was used as the benchmark of agonist action.</p><p><strong>Conclusion: </strong>Compounds PT14 and PT18 are promising candidates to investigate further, as they exhibit significant cell viability, glucose uptake, and PPAR-γ transcription factor activation in nuclear extracts. These encouraging findings warrant further exploration through in vivo pharmacokinetic and antidiabetic efficacy studies, which will help establish their therapeutic potential and safety profile in biological systems.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144871796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Dysregulation of the cAMP Signaling Pathway Mediated by Shared Hub Genes: Diagnostic, Prognostic, and Therapeutic Insights into Thyroid Cancer and Graves' Disease.","authors":"Tingting Hu, Ruqiong Sun, Juanfei Zhu","doi":"10.2174/0113862073389123250723210633","DOIUrl":"https://doi.org/10.2174/0113862073389123250723210633","url":null,"abstract":"<p><strong>Background: </strong>Graves' disease and Thyroid cancer share overlapping molecular mechanisms that may reveal potential biomarkers and therapeutic targets. Identifying shared hub genes can provide insights into disease progression and improve diagnosis and treatment strategies.</p><p><strong>Methodology: </strong>Gene expression profiles from Graves' disease (GSE71956) and thyroid cancer (GSE153659) datasets were analyzed to identify differentially expressed genes (DEGs) using the limma package. Common DEGs were identified through cross-dataset comparison, and hub genes were determined based on the degree method. CD44, RHOC, HCN4, and MYH10 hub genes were validated using RT-qPCR in thyroid cancer and normal cell lines and analyzed for functional roles via siRNA-mediated knockdown experiments. miRNA regulators of hub genes were predicted using TargetScan and validated with RT-qPCR. Promoter methylation, mutation, and copy number variation (CNV) analyses were conducted using OncoDB and cBioPortal. Prognostic significance was assessed using the cSurvival and GENT2 databases. Functional enrichment analysis was performed using DAVID, GeneMANIA, and GSCA. miRNA target prediction was carried out with TargetScan, and expression correlation analysis was performed using GEPIA2.</p><p><strong>Results: </strong>Twenty-three common DEGs were identified, with CD44, RHOC, HCN4, and MYH10 emerging as key hub genes. RT-qPCR showed significant (p < 0.001) upregulation of these genes in thyroid cancer cell lines. Functional analysis linked these genes to cAMP signaling and EMT pathways, with promoter hypomethylation, CNV amplifications, and mutations contributing to their dysregulation. CD44 and RHOC knockdown significantly (p < 0.01) reduced proliferation, colony formation, and migration in SW579 cells. Predicted tumor-suppressive miRNAs, including hsa-miR-199a-3p and hsa-miR-455-3p.2, were significantly (p < 0.0001) downregulated in thyroid cancer.</p><p><strong>Conclusion: </strong>CD44, RHOC, HCN4, and MYH10 were identified as shared hub genes between Graves' disease and thyroid cancer, with functional roles in oncogenic pathways. Their diagnostic and therapeutic potential offers novel insights into thyroid disease pathogenesis.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144798368","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Knockdown of NFS1 and Oxaliplatin Combination Induces Pyroptosis via the Caspase-3/GSDME Pathway in Gastric Cancer Cells.","authors":"Weiwei Jiang, Miao Zhang, Nan Hu, Chenxi Mao, Yiqian Han, Luming Zhao, Mingtong Liang, Fenglei Wu","doi":"10.2174/0113862073386878250801103600","DOIUrl":"https://doi.org/10.2174/0113862073386878250801103600","url":null,"abstract":"<p><strong>Introduction: </strong>This study aimed to elucidate the role of NFS1 in gastric cancer (GC) prognosis, pyroptosis, and oxaliplatin chemosensitivity, and to explore its interaction with the MAPK signaling pathway.</p><p><strong>Methods: </strong>GC mRNA expression and clinical survival data were obtained from The Cancer Genome Atlas Gastric Adenocarcinoma (TCGA-STAD). Kaplan-Meier analysis assessed the prognostic significance of NFS1. R software facilitated NFS1 expression analysis and KEGG pathway enrichment. Pyroptosis was evaluated using Cell Counting Kit-8, flow cytometry, and morphological analysis. Western blotting quantified pyroptosis-related protein expression. RNA sequencing libraries were prepared and sequenced on the Illumina platform.</p><p><strong>Results: </strong>Oxaliplatin treatment significantly reduced cell viability and induced pyroptosis, which was markedly attenuated by GSDME deficiency. Oxaliplatin activated caspase-3 and cleaved GSDME, effects that were reversed by the caspase-3 inhibitor Z-DEVD. NFS1 knockdown enhanced GSDME and caspase-3 cleavage, increasing pyroptosis (PI and Annexin-V doublepositive cells) compared to controls. KEGG analysis of RNA sequencing and TCGA data highlighted the MAPK signaling pathway. Western blotting confirmed that oxaliplatin combined with NFS1 knockdown suppressed MAPK pathway proteins.</p><p><strong>Discussion: </strong>The caspase-3/GSDME axis mediates oxaliplatin-induced GC pyroptosis. High NFS1 expression inhibits GSDME activation, promotes MAPK protein activation, and reduces oxaliplatin sensitivity. These findings suggest that the caspase-3/GSDME pathway offers a novel mechanism for oxaliplatin's antitumor effects. NFS1 may serve as an independent prognostic biomarker in GC, influencing disease progression through MAPK regulation.</p><p><strong>Conclusion: </strong>NFS1 is a promising therapeutic target for gastric cancer, especially in the study of oxaliplatin-based chemotherapy in combination with a treatment regimen that triggers pyroptosis.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144798369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Baihe Dihuang Danshen Decoction Alleviates Myocardial Ischemia-Reperfusion Injury in Depression-Induced Rats by Inhibiting Ferroptosis.","authors":"Ling Huang, Cuihua Liu, Yudan Liang, Wenquan Huang, Shizhong Zhang, Qiang Xu, Kuncheng Qiu","doi":"10.2174/0113862073387437250803043736","DOIUrl":"https://doi.org/10.2174/0113862073387437250803043736","url":null,"abstract":"<p><strong>Introduction: </strong>The comorbidity of myocardial ischemia reperfusion injury (MIRI) and depression (DEP) may worsen the prognosis of coronary heart disease surgery. Currently, research on medications and therapeutic mechanisms for MIRI combined with DEP is still insufficient. This study aims to explore the relationship between DEP and MIRI, and the therapeutic effects and mechanisms of Baihe Dihuang Danshen decoction (BDDSD) on DEP combined with MIRI.</p><p><strong>Methods: </strong>SD rats were assigned to a final experimental framework of six groups (Sham, MIRI, DEP+MIRI, BDDSD, DEP drug control, MIRI drug control). DEP was induced via 6-week chronic unpredictable mild stress (CUMS), with BDDSD administered during the final 2 weeks. MIRI was then induced by 30-minute coronary artery ligation and 2-hour reperfusion. DEP severity was assessed using behavioral tests (open field, elevated plus maze, sucrose preference, forced swimming). MIRI outcomes were evaluated via infarct size, histopathology, serum markers (LDH, IL-6, IL-1β), myocardial oxidative stress (MDA, GSH, SOD, Fe²⁺), and NADPH/FSP1/CoQ10 pathway proteins (FSP1, CoQ10, FTL, NOX2, NOX4, COX2).</p><p><strong>Results: </strong>Compared with the MIRI group, DEP significantly exacerbated MIRI, manifested by increased serum IL-6 and IL-1β levels, enlarged infarction area, and aggravated oxidative damage (elevated MDA/Fe²⁺, decreased SOD/GSH). Compared with the DEP+MIRI group, BDDSD intervention relieved DEP of rats, and subsequently reduced infarction area; decreased serum LDH, IL-6, and IL-1β; lowered myocardial MDA and Fe²⁺ while increasing SOD and GSH; upregulated FSP1/CoQ10/FTL; and downregulated NOX2/NOX4/COX2 expression.</p><p><strong>Discussion: </strong>DEP can aggravate inflammation and oxidative stress, promoting cardiac ferroptosis, thereby exacerbating MIRI. Our results demonstrate that BDDSD alleviates MIRI-DEP comorbidity through a dual mechanism, mitigating depressive symptoms and inhibiting myocardial ferroptosis via modulation of the NADPH/FSP1/CoQ10 pathway. Although the efficacy of BDDSD is encouraging, its dose-effect relationship and long-term safety require further study.</p><p><strong>Conclusion: </strong>BDDSD effectively treats DEP-MIRI comorbidity through its dual mechanism, mitigating DEP and protecting against myocardial ferroptosis. Our study not only offers a novel therapeutic strategy for patients with DEP requiring coronary heart disease surgery but also provides new targets for developing drugs to treat MIRI combined with DEP.</p>","PeriodicalId":10491,"journal":{"name":"Combinatorial chemistry & high throughput screening","volume":" ","pages":""},"PeriodicalIF":1.7,"publicationDate":"2025-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144798367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}