PharmacologyPub Date : 2025-02-05DOI: 10.1159/000543902
Gunhyuk Park, Yumi Jang, Byeong Cheol Moon, Hye-Sun Lim
{"title":"Batryticatus bombyx improved atopic dermatitis in NC/Nga mice and impact on inflammation and recovery of skin barrier via STAT1/P38/NF-κB downregulation and HO-1/Nrf2 activation in HaCaT keratinocytes.","authors":"Gunhyuk Park, Yumi Jang, Byeong Cheol Moon, Hye-Sun Lim","doi":"10.1159/000543902","DOIUrl":"https://doi.org/10.1159/000543902","url":null,"abstract":"<p><strong>Introduction: </strong>Batryticatus bombyx (BB) (Beauveria bassiana Vuill.) is used as an herbal medicine and food additive. Although BB has neuroprotective and anti-apoptotic effects, its impacts on atopic dermatitis (AD) are unknown. We evaluated the effects of BB in an NC/Nga mouse model of house dust mite (HDM)-induced AD, in TNF-α and IFN-γ (TI)-stimulated HaCaT keratinocytes, and in a 3D human skin model.</p><p><strong>Methods: </strong>NC/Nga mice were induced with HDM and treated with BB for 6 weeks. We assessed skin symptoms, serum levels of IgE, histamine, and IL-6, immune cell counts, and performed histological analysis of dorsal skin tissue. Additionally, we induced inflammation in HaCaT cells and a 3D human skin model using TNF-α/IFN-γ. We measured inflammatory cytokine levels, skin hydration factors, and delved into underlying mechanisms via ELISA, real-time PCR, and Western blot.</p><p><strong>Results: </strong>BB improved skin lesions, reduced thickness, and inflammatory cell infiltration in HDM-induced mice. It alleviated scratching, improved moisture retention, and reduced IgE, histamine, and IL-6 levels. In HaCaT cells, BB inhibited thymic stromal lymphopoietin and increased hyaluronan content. It also upregulated aquaporin-3, hyaluronan synthase-1/3, filaggrin, involucrin, and occludin, enhancing skin hydration and tight junction stability. BB decreased STAT1, p38 MAPK, and NF-κB p65 levels in HaCaT cells and exhibited antioxidant effects by increasing HO-1/Nrf2 expression.</p><p><strong>Conclusion: </strong>BB may serve as a therapeutic alternative for AD treatment by inhibiting skin inflammation.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"1-21"},"PeriodicalIF":2.9,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143190236","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}
PharmacologyPub Date : 2025-02-03DOI: 10.1159/000543234
Kimberly D Herman, Ian Holyer, Duncan C Humphries, James A Roper, Kristoffer Peterson, Fredrik R Zetterberg, Anders Pedersen, Alison C Mackinnon, Robert J Slack
{"title":"Pharmacological characterization of GB1908, a selective galectin-1 carbohydrate binding domain inhibitor for the treatment of cancer.","authors":"Kimberly D Herman, Ian Holyer, Duncan C Humphries, James A Roper, Kristoffer Peterson, Fredrik R Zetterberg, Anders Pedersen, Alison C Mackinnon, Robert J Slack","doi":"10.1159/000543234","DOIUrl":"https://doi.org/10.1159/000543234","url":null,"abstract":"<p><strong>Introduction: </strong>Galectin-1 (Gal-1) is a lectin that has been shown to be involved in a number of pro-tumorigenic mechanisms and has also been shown to be immune-suppressive. Therefore, pharmacological blockade of Gal-1 has the potential to be therapeutically beneficial in cancers that overexpress this lectin where it is hypothesized to be driving cancer progression.</p><p><strong>Methods: </strong>GB1908 is a novel, selective and high affinity inhibitor of the Gal-1 carbohydrate recognition domain and in this study we have pharmacologically characterized this small molecule in a range of in vitro and in vivo systems in the context of cancer therapy. In addition, we used a data-driven approach to identify the cancer types which may benefit from Gal-1 inhibitor therapy.</p><p><strong>Results: </strong>The selectivity of GB1908 for Gal-1 compared with galectin-3 (Gal-3) was confirmed in biophysical and cellular assays. GB1908 attenuated Gal-1-induced T cell (Jurkat) apoptosis and reduced the production of immunosuppressive cytokines in a stromal non-small cell lung cancer (NSCLC) tumor microenvironment model. Breast carcinoma and metastatic skin cutaneous melanoma were identified as cancers in which high Gal-1 expression correlated with poorer survival outcomes in patients. Treatment with GB1908 slowed tumor growth in syngeneic mouse models of these cancers.</p><p><strong>Conclusion: </strong>The inhibition of both tumor growth and immune-suppressive cytokines, in cancers in which high Gal-1 is associated with poorer survival outcomes, suggests a potential therapeutic benefit for Gal-1 inhibitors such as GB1908.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"1-19"},"PeriodicalIF":2.9,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143123420","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}
PharmacologyPub Date : 2025-01-27DOI: 10.1159/000543797
Akira Sato, Yuta Ogawa, Ayano Yabuki, Genta Sato, Hina Nemoto, Makoto Ohira
{"title":"Estrogen receptors and platelet-activating acetylhydrolase activity in the pathogenesis of systemic lupus erythematosus.","authors":"Akira Sato, Yuta Ogawa, Ayano Yabuki, Genta Sato, Hina Nemoto, Makoto Ohira","doi":"10.1159/000543797","DOIUrl":"https://doi.org/10.1159/000543797","url":null,"abstract":"<p><strong>Introduction: </strong>Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease that affects various body systems, including the skin and facial features. Estrogen promotes lupus in human and mouse models of SLE. In this study, we conducted an in vivo study to investigate the relationship between two estrogen receptors (ERα and ERβ) and platelet-activating factor acetylhydrolase (PAF-AH) on the symptoms of SLE.</p><p><strong>Methods: </strong>Skin and facial features, serum PAF-AH activity, tumor-necrosis factor-alpha (TNF-α), and anti-double-stranded DNA (anti-dsDNA) antibody titer, all of which are central to the classification and pathogenesis of SLE, were measured in SLE model mice injected with various ER antagonists.</p><p><strong>Results: </strong>In SLE mice, the injection of an ERβ antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-α]pyrimidine-3-yl]-phenol induced the expression of facial erythema (especially around a cheek). The injection of an ERα/β antagonist tamoxifen induced hair-loss on the head and the neck. The injection of an ERα antagonist methylpiperidino pyrazole or tamoxifen increased PAF-AH activity and decreased TNF-α levels and anti-dsDNA antibody in mouse serum.</p><p><strong>Conclusions: </strong>The activation of ERα and inactivation of ERβ can cause inflammatory and cutaneous symptoms of SLE, respectively, implying that ERα antagonists and/or ERβ agonists can be useful for the prevention and treatment of SLE.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"1-12"},"PeriodicalIF":2.9,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143053310","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":"Safety, tolerability, pharmacokinetics and effects of gene polymorphisms on Furaprevir (TG-2349), a novel hepatitis C inhibitor: a randomized phase Ι study.","authors":"Su-Mei Xu, Ying-Jun Zhang, Li-Wen Chang, Cheng-Yuan Tsai, Dai Li, Ping-Sheng Xu","doi":"10.1159/000543416","DOIUrl":"https://doi.org/10.1159/000543416","url":null,"abstract":"<p><strong>Introduction: </strong>The purpose of our study was to evaluate the safety, tolerability, and pharmacokinetics of furaprevir, a new highly selective hepatitis C virus NS3/4A protease inhibitor.</p><p><strong>Methods: </strong>The study was divided into 2 parts: Part A (single ascending-dose study, SAD) and Part B (multiple ascending-dose study, MAD). A total of 62 healthy subjects were enrolled in the studies. DNA samples were extracted from all subjects and genotypes of CYP3A5*3, CYP3A4*1G and POR*28 were analyzed by ligase detection reaction (LDR).</p><p><strong>Results: </strong>We used nonlinear mixed effects model (NONMEM) to construct furaprevir population pharmacokinetics model. (1) In SAD, Cmax and AUC were greater than dose increased ratio in the dose rang of 100-400 mg; (2) In MAD, Cmax and AUC increased in an approximately dose-proportional manner in the dose range of 200-600 mg; (3) A one-compartment model with transit absorption described the plasma concentrations of furaprevir. The apparent clearance (CL) was estimated at 33.4 L/h. The distribution volume of compartment (V2) was 219.0 L. No serious adverse event occurred in the studies. But other screening gene mutations had no statistically significant effects on the pharmacokinetics of furaprevir.</p><p><strong>Conclusion: </strong>Food significantly impacts the bioavailability of furaprevir. Furaprevir does not accumulate in vivo after multiple rising doses and has demonstrated safety and tolerability in healthy subjects, supporting its further investigation in patients with hepatitis C.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"1-24"},"PeriodicalIF":2.9,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010234","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}
PharmacologyPub Date : 2025-01-08DOI: 10.1159/000543293
Sufia Husain, Nervana Mustafa Kamal Bayoumy, Fatima Noor, Hanan Hagar
{"title":"Nephroprotective Effects of Curcumin in Murine Models of Focal and Segmental Glomerulosclerosis.","authors":"Sufia Husain, Nervana Mustafa Kamal Bayoumy, Fatima Noor, Hanan Hagar","doi":"10.1159/000543293","DOIUrl":"https://doi.org/10.1159/000543293","url":null,"abstract":"<p><strong>Introduction: </strong>This study aims to explore the reno-protective effect of Curcumin in focal and segmental glomerulosclerosis (FSGS) in Murine models, a common chronic glomerulopathy that leads to end stage renal disease.</p><p><strong>Methods: </strong>Adult Wistar rats were used in this experiment. One group was treated with intravenous Adriamycin (ADR) injection to induce FSGS similar to that seen in humans and a second group was co-administered ADR and Curcumin (ADR-CUR). Saline treated rats served as controls. Renal injury was assessed by measuring the levels of serum creatinine, blood urea nitrogen (BUN), triglyceride and urinary protein. The homogenates of renal cortex were used to estimate the renal content of the inflammatory marker, tumor necrosis factor-a (TNF-a); oxidative stress marker, Malonaldehyde (MDA); and two anti-oxidants superoxide dismutase (SOD) and reduced glutathione (GSH). In addition, the rat kidneys were harvested by ends of week-8 and week-12 and examined for histological abnormalities.</p><p><strong>Results: </strong>The ADR-treated rats showed biochemical and histological evidence of FSGS, in the form of proteinuria, elevated serum creatinine, BUN and triglycerides, elevated renal TNF-a and MDA, and segmental glomerulosclerosis. The ADR-CUR treated rats showed significant correction of all these variables. Co-administration with Curcumin resulted in improvement of the proteinuria, serum creatinine, BUN and triglyceride. The renal tissue levels of anti-oxidants SOD and GSH increased and that of TNF-a and MDA decreased and the histology revealed reduction in the extent of segmental glomerulosclerosis. The FSGS associated renal damage was notably antagonized by curcumin treatment.</p><p><strong>Conclusion: </strong>Our findings confirm the reno-protective effects of Curcumin as a potential therapeutic agent in protection against the progression of FSGS and indicate that it is mitigated by inhibition of oxidant injury and inflammation and also by promotion of antioxidants. Curcumin ameliorated the ADR-induced FSGS in murine models. It may be a promising compound in the treatment of FSGS in human subjects. More human studies are needed to further elucidate its effects in FSGS.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"1-17"},"PeriodicalIF":2.9,"publicationDate":"2025-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142953662","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}
PharmacologyPub Date : 2025-01-01Epub Date: 2024-07-30DOI: 10.1159/000540427
Ivan R Milovanovic, Ana V Pejcic
{"title":"Drug-Drug Interactions in Hospitalized Urological Patients: A Retrospective Cohort Study.","authors":"Ivan R Milovanovic, Ana V Pejcic","doi":"10.1159/000540427","DOIUrl":"10.1159/000540427","url":null,"abstract":"<p><strong>Introduction: </strong>Exposure to potential drug-drug interactions (pDDIs) can be a notable source of avoidable drug-related harm that requires adequate management to prevent medical errors. We aimed to evaluate pDDIs and associated factors in hospitalized urological patients on admission, during hospitalization, and on discharge.</p><p><strong>Methods: </strong>A retrospective cohort study was conducted at the Clinic of Urology of the University Clinical Centre Kragujevac, Serbia. To detect pDDIs, we used Lexicomp, which categorizes pDDIs as follows: X (avoid combination), D (consider therapy modification), C (monitor therapy), B (no action needed), and A (no known interaction). Multiple linear regression analysis was used to identify factors associated with the number of pDDIs.</p><p><strong>Results: </strong>More than half of the 220 included patients had at least one pDDI on admission and discharge (57.3% and 63.6%, respectively), whereas 95.0% had at least one pDDI during hospitalization. The total number and number of X, D, C, and B categories of pDDIs were the highest during hospitalization and the lowest on admission. Duration of hospitalization, arrhythmias, dementia, renal failure, cancer, surgery during hospitalization, number of prescribed drugs, and various pharmacological drug classes were risk factors for a higher number of pDDIs, while age, ischemic heart disease, hypertension, and development of infection during hospitalization were protective factors in at least one of the stages. The impact of renal colic depended on the stage and category of pDDI.</p><p><strong>Conclusion: </strong>More than half of the urological patients were exposed to at least one pDDIs at all stages. Medical professionals should regularly screen for pDDIs, particularly in patients with risk factors.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"15-25"},"PeriodicalIF":2.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141856287","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}
PharmacologyPub Date : 2025-01-01Epub Date: 2024-08-12DOI: 10.1159/000540882
Gregory C Havton, Alex T C Tai, Surabhi Vasisht, Daryl L Davies, Liana Asatryan
{"title":"Preclinical Evaluation of Sodium Butyrate's Potential to Reduce Alcohol Consumption: A Dose-Escalation Study in C57BL/6J Mice in Antibiotic-Enhanced Binge-Like Drinking Model.","authors":"Gregory C Havton, Alex T C Tai, Surabhi Vasisht, Daryl L Davies, Liana Asatryan","doi":"10.1159/000540882","DOIUrl":"10.1159/000540882","url":null,"abstract":"<p><strong>Introduction: </strong>In our earlier efforts to establish gut-brain axis during alcohol use disorder (AUD), we have demonstrated that supplementation of C57BL/6J male mice with 8 mg/mL sodium butyrate, a major short-chain fatty acid, in drinking water reduced ethanol intake and neuroinflammatory response in antibiotic (ABX)-enhanced voluntary binge-like alcohol consumption model, drinking in the dark (DID).</p><p><strong>Methods: </strong>To further evaluate the preclinical potential of SB, we have set a dose-escalation study in C57BL/6J male mice to test effects of ad libitum 20 mg/mL SB and 50 mg/mL SB and their combinations with ABX in the DID procedure for 4 weeks. Effects of these SB concentrations on ethanol consumption and bodily parameters were determined for the duration of the treatments. At the end of study, blood, liver, and intestinal tissues were collected to study any potential adverse effects ad to measure blood ethanol concentrations.</p><p><strong>Results: </strong>Increasing SB concentrations in the drinking water caused a loss in the protective effect against ethanol consumption and produced adverse effects on body and liver weights, reduced overall liquid intake. The hypothesis that these effects were due to aversion to SB smell/taste at these high concentrations were further tested in a follow up proof-of-concept study with intragastric gavage administration of SB. The higher gavage dose (320 mg/kg) caused reduction in ethanol consumption without any adverse effects.</p><p><strong>Conclusion: </strong>Overall, these findings added more support for the therapeutic potential of SB in management of AUD, given a proper form of administration.</p><p><strong>Introduction: </strong>In our earlier efforts to establish gut-brain axis during alcohol use disorder (AUD), we have demonstrated that supplementation of C57BL/6J male mice with 8 mg/mL sodium butyrate, a major short-chain fatty acid, in drinking water reduced ethanol intake and neuroinflammatory response in antibiotic (ABX)-enhanced voluntary binge-like alcohol consumption model, drinking in the dark (DID).</p><p><strong>Methods: </strong>To further evaluate the preclinical potential of SB, we have set a dose-escalation study in C57BL/6J male mice to test effects of ad libitum 20 mg/mL SB and 50 mg/mL SB and their combinations with ABX in the DID procedure for 4 weeks. Effects of these SB concentrations on ethanol consumption and bodily parameters were determined for the duration of the treatments. At the end of study, blood, liver, and intestinal tissues were collected to study any potential adverse effects ad to measure blood ethanol concentrations.</p><p><strong>Results: </strong>Increasing SB concentrations in the drinking water caused a loss in the protective effect against ethanol consumption and produced adverse effects on body and liver weights, reduced overall liquid intake. The hypothesis that these effects were due to aversion to SB smell/taste at these","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"36-48"},"PeriodicalIF":2.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11794028/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141971702","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Semaglutide Ameliorates Hepatocyte Steatosis in a Cell Co-Culture System by Downregulating the IRE1α-XBP1-C/EBPα Signaling Pathway in Macrophages.","authors":"Qin Hu, Li Zhang, YiTing Tao, ShuangLin Xie, AiYun Wang, Caiying Luo, RenHua Yang, Zhiqiang Shen, Bo He, Yu Fang, Peng Chen","doi":"10.1159/000540654","DOIUrl":"10.1159/000540654","url":null,"abstract":"<p><strong>Introduction: </strong>Non-alcoholic fatty liver disease (NAFLD) is currently the most common type of chronic liver disease. Semaglutide is a glucose-lowering drug administered for the treatment of type 2 diabetes mellitus (T2DM) and is clinically effective in the treatment of NAFLD. X-box binding protein 1 (XBP1) is related to the pathogenesis of both NAFLD and T2DM. The aim of the present study was to demonstrate whether the underlying mechanism of semaglutide treatment for NAFLD is via downregulation of the inositol-requiring transmembrane kinase/endonuclease-1α (IRE1α)-XBP1-CCAAT/enhancer binding protein α (C/EBPα) signaling pathway in macrophages.</p><p><strong>Methods: </strong>In the present study, NAFLD cell modeling was induced by oleic acid (0.4 m<sc>m</sc>) and palmitic acid (0.2 m<sc>m</sc>). Hepatocytes (AML12) and macrophages (RAW264.7) were co-cultured in 6-well Transwell plates. Semaglutide (60 or 140 n<sc>m</sc>) was administrated for 24 h, while pioglitazone (2 μ<sc>m</sc>) and toyocamycin (200 n<sc>m</sc>) were used as a positive control drug and a XBP1 inhibitor, respectively. Autophagy and apoptosis of AML12 cells were detected by transmission electron microscopy and Western blotting (WB). Hepatocyte steatosis was evaluated by adopting total intracellular triglyceride determination, analysis of the relative expression of proteins and genes associated with lipid metabolism and hepatocyte Oil red O staining. Detection of inflammation factors was conducted by ELISA and WB. To explore the underlying mechanism of NAFLD treatment with semaglutide, the relative expression of related proteins and genes were tested.</p><p><strong>Results: </strong>Our study demonstrated that semaglutide treatment improved autophagy and inhibited apoptosis of hepatocytes, while notably ameliorating steatosis of hepatocytes. In addition, inflammation was attenuated in the NAFLD cell co-culture model after semaglutide administration. Semaglutide also significantly reduced the protein and gene expression levels of the IRE1α-XBP1-C/EBPα signaling pathway in macrophages.</p><p><strong>Conclusion: </strong>Semaglutide partially ameliorated NAFLD by downregulating the IRE1α-XBP1-C/EBPα signaling pathway in macrophages. These findings may provide a potential theoretical basis for semaglutide therapy for NAFLD.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"26-35"},"PeriodicalIF":2.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141875727","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":"Early Use of PCSK9 Inhibitors in the Prognosis of Patients with Acute Coronary Syndrome by Protecting Vascular Endothelial Function.","authors":"Linghao Xu, Yuanqi Wang, Yiqiong Wang, Liang Wang, Peizhao Du, Jing Cheng, Chunsheng Zhang, Tiantian Jiao, Lijian Xing, Md Sakibur Rahman Tapu, Haonan Jia, Jiming Li","doi":"10.1159/000540083","DOIUrl":"10.1159/000540083","url":null,"abstract":"<p><strong>Introduction: </strong>Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) has a protective effect on acute coronary syndrome (ACS). However, most studies have shown that this protective effect is based on a decrease in low-density lipoprotein cholesterol, while other mechanisms remain limited. This study aimed to determine whether PCSK9i can improve the prognosis of ACS patients by protecting endothelial function.</p><p><strong>Methods: </strong>A total of 113 ACS patients were enrolled and randomly assigned to PCSK9i group (PCSK9i combined with statins) and control group (statins only). Blood lipids and endothelial function indicators were measured and analyzed 6 weeks before and after treatment. The effect of PCSK9i on the expression and secretion of endothelial function indicators in vascular endothelial cells were studied by cell experiments.</p><p><strong>Results: </strong>After 6 weeks of treatment, endothelial function indicators such as nitric oxide (NO), thrombomodulin, intercellular cell adhesion molecule-1, endothelin-1, and flow-mediated vasodilation were significantly improved in PCSK9i group compared with control group. Only the changes of NO and von Willebrand factor were associated with blood lipid levels, whereas the changes of other endothelial function indicators were not significantly associated with blood lipid levels. PCSK9i reduced the incidence of major adverse cardiovascular events in patients with ACS compared to those in the control group. In cell experiments, PCSK9i treatment significantly ameliorated LPS induced endothelial injury in HUVECs.</p><p><strong>Conclusion: </strong>PCSK9i can protect vascular endothelial function partly independently of its lipid-lowering effect and ameliorate the prognosis of patients with ACS within 6 weeks. This mechanism may involve heat shock transcription factor 1/heat shock proteins -related signaling pathways. Early use of PCSK9i in patients with ACS should be strongly considered in clinical practice.</p>","PeriodicalId":20209,"journal":{"name":"Pharmacology","volume":" ","pages":"1-14"},"PeriodicalIF":2.9,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141535081","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}