首页 > 最新文献

Frontiers in Pharmacology最新文献

英文 中文
Impact of fluoxetine exposure on Lymnaea stagnalis and its developing eggs: integrating untargeted lipidomics, targeted metabolomics, and classical risk assessment. 氟西汀暴露对锚头鳋及其发育中卵子的影响:非靶向脂质组学、靶向代谢组学和经典风险评估的整合。
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-04 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1536438
Diana Ilyaskina, Yumi Nakadera, Marja H Lamoree, Joris M Koene, Pim E G Leonards

Pharmaceuticals such as selective serotonin reuptake inhibitors (SSRIs), are increasingly detected in aquatic environments, posing potential risks to non-target organisms, because many of those substances are widely shared neuromodulator. In this study, we investigated the effects of SSRI antidepressant, namely, fluoxetine, exposure on the freshwater snail L. stagnalis, focusing on egg development, neurochemical pathways, and lipid metabolism. Snails were exposed to a range of 51-434 µg fluoxetine L⁻1 for 7 days, followed by analysis of survival, feeding behaviour, reproduction, and metabolomic changes in the central nervous system (CNS), albumen gland, and eggs. Although no significant effects were observed on survival or fecundity, fluoxetine exposure significantly impaired egg development in a dose-dependent manner, reducing hatching rates with an EC50 of 126 µg fluoxetine L⁻1. Removal of eggs from the contaminated environment partially reversed these developmental effects, suggesting potential recovery if fluoxetine levels decrease. Molecular analysis revealed several neurochemical and lipidomic alterations. In the CNS, elevated levels of catecholamines, phosphatidylcholines (PC), and ceramides were linked to disruptions in neurotransmission, membrane integrity, and impaired embryo development. In the albumen gland, we detected a decrease of key lipid classes, including sphingomyelins and fatty acids, which can be linked with impaired egg quality. Additionally, a decrease in histamine in both the albumen gland and eggs suggested further disruption of egg development, potentially affecting metamorphosis success. Moreover, the dose-dependent increase in choline, along with PC and oxidized PC, indicated oxidative stress and lipid peroxidation in the CNS and exposed eggs of Lymnaea stagnalis. Our findings highlight the benefits of combining behavioral assessments with metabolomic profiling to better understand the mechanistic pathways underlying fluoxetine's adverse effects.

{"title":"Impact of fluoxetine exposure on <i>Lymnaea stagnalis</i> and its developing eggs: integrating untargeted lipidomics, targeted metabolomics, and classical risk assessment.","authors":"Diana Ilyaskina, Yumi Nakadera, Marja H Lamoree, Joris M Koene, Pim E G Leonards","doi":"10.3389/fphar.2025.1536438","DOIUrl":"10.3389/fphar.2025.1536438","url":null,"abstract":"<p><p>Pharmaceuticals such as selective serotonin reuptake inhibitors (SSRIs), are increasingly detected in aquatic environments, posing potential risks to non-target organisms, because many of those substances are widely shared neuromodulator. In this study, we investigated the effects of SSRI antidepressant, namely, fluoxetine, exposure on the freshwater snail <i>L. stagnalis</i>, focusing on egg development, neurochemical pathways, and lipid metabolism. Snails were exposed to a range of 51-434 µg fluoxetine L⁻<sup>1</sup> for 7 days, followed by analysis of survival, feeding behaviour, reproduction, and metabolomic changes in the central nervous system (CNS), albumen gland, and eggs. Although no significant effects were observed on survival or fecundity, fluoxetine exposure significantly impaired egg development in a dose-dependent manner, reducing hatching rates with an EC50 of 126 µg fluoxetine L⁻<sup>1</sup>. Removal of eggs from the contaminated environment partially reversed these developmental effects, suggesting potential recovery if fluoxetine levels decrease. Molecular analysis revealed several neurochemical and lipidomic alterations. In the CNS, elevated levels of catecholamines, phosphatidylcholines (PC), and ceramides were linked to disruptions in neurotransmission, membrane integrity, and impaired embryo development. In the albumen gland, we detected a decrease of key lipid classes, including sphingomyelins and fatty acids, which can be linked with impaired egg quality. Additionally, a decrease in histamine in both the albumen gland and eggs suggested further disruption of egg development, potentially affecting metamorphosis success. Moreover, the dose-dependent increase in choline, along with PC and oxidized PC, indicated oxidative stress and lipid peroxidation in the CNS and exposed eggs of <i>Lymnaea stagnalis</i>. Our findings highlight the benefits of combining behavioral assessments with metabolomic profiling to better understand the mechanistic pathways underlying fluoxetine's adverse effects.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1536438"},"PeriodicalIF":4.4,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11832466/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143448742","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computational and in vitro evaluation of sumac-derived ©Rutan compounds towards Sars-CoV-2 Mpro inhibition. 计算和体外评估苏木衍生的©Rutan化合物对 Sars-CoV-2 Mpro 的抑制作用。
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-04 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1518463
Muzaffar Kayumov, Parthiban Marimuthu, Jamoliddin Razzokov, Nurkhodja Mukhamedov, Akmal Asrorov, Nodir S Berdiev, Jamolitdin F Ziyavitdinov, Ansor Yashinov, Yuliya Oshchepkova, Shavkat Salikhov, Sharafitdin Mirzaakhmedov

The emergence of the SARS-CoV-2 virus caused the COVID-19 outbreak leading to a global pandemic. Natural substances started being screened for their antiviral activity by computational and in-vitro techniques. Here, we evaluated the anti-SARS-CoV-2 main protease (Mpro) efficacy of ©Rutan, which contains five polyphenols (R5, R6, R7, R7', and R8) extracted from sumac Rhus coriaria L. We obtained three fractions after large-scale purification: fraction 1 held R5, fraction 2 consisted of R6, R7 and R7', and fraction 3 held R8. In vitro results showed their anti-Mpro potential: IC50 values of R5 and R8 made 42.52 µM and 5.48 µM, respectively. Further, we studied Mpro-polyphenol interactions by in silico analysis to understand mechanistic extrapolation of Rutan binding nature with Mpro. We extensively incorporated a series of in silico techniques. Initially, for the docking protocol validation, redocking of the co-crystal ligand GC-376* to the binding pocket of Mpro was carried out. The representative docked complexes were subjected to long-range 500 ns molecular dynamics simulations. The binding free energy (BFE in kcal/mol) of components were calculated as follows: R8 (-104.636) > R6 (-93.754) > R7' (-92.113) > R5 (-81.115) > R7 (-67.243). In silico results of R5 and R8 correspond with their in vitro outcomes. Furthermore, the per-residue decomposition analysis showed C145, E166, and Q189 residues as the hotspot residues for components contributing to maximum BFE energies. All five components effectively interact with the catalytic pocket of Mpro and form stable complexes that allow the estimation of their inhibitory activity. Assay kit analyses revealed that Rutan and its components have effective anti-SARS-CoV-2 Mpro inhibitory activity.

{"title":"Computational and <i>in vitro</i> evaluation of sumac-derived <sup>©</sup>Rutan compounds towards Sars-CoV-2 M<sup>pro</sup> inhibition.","authors":"Muzaffar Kayumov, Parthiban Marimuthu, Jamoliddin Razzokov, Nurkhodja Mukhamedov, Akmal Asrorov, Nodir S Berdiev, Jamolitdin F Ziyavitdinov, Ansor Yashinov, Yuliya Oshchepkova, Shavkat Salikhov, Sharafitdin Mirzaakhmedov","doi":"10.3389/fphar.2025.1518463","DOIUrl":"10.3389/fphar.2025.1518463","url":null,"abstract":"<p><p>The emergence of the SARS-CoV-2 virus caused the COVID-19 outbreak leading to a global pandemic. Natural substances started being screened for their antiviral activity by computational and <i>in-vitro</i> techniques. Here, we evaluated the anti-SARS-CoV-2 main protease (M<sup>pro</sup>) efficacy of <sup>©</sup>Rutan, which contains five polyphenols (R5, R6, R7, R7', and R8) extracted from sumac <i>Rhus coriaria</i> L. We obtained three fractions after large-scale purification: fraction 1 held R5, fraction 2 consisted of R6, R7 and R7', and fraction 3 held R8. <i>In vitro</i> results showed their anti-M<sup>pro</sup> potential: IC<sub>50</sub> values of R5 and R8 made 42.52 µM and 5.48 µM, respectively. Further, we studied M<sup>pro</sup>-polyphenol interactions by <i>in silico</i> analysis to understand mechanistic extrapolation of Rutan binding nature with M<sup>pro</sup>. We extensively incorporated a series of <i>in silico</i> techniques. Initially, for the docking protocol validation, redocking of the co-crystal ligand GC-376* to the binding pocket of M<sup>pro</sup> was carried out. The representative docked complexes were subjected to long-range 500 ns molecular dynamics simulations. The binding free energy (BFE in kcal/mol) of components were calculated as follows: R8 (-104.636) > R6 (-93.754) > R7' (-92.113) > R5 (-81.115) > R7 (-67.243). <i>In silico</i> results of R5 and R8 correspond with their <i>in vitro</i> outcomes. Furthermore, the per-residue decomposition analysis showed C145, E166, and Q189 residues as the hotspot residues for components contributing to maximum BFE energies. All five components effectively interact with the catalytic pocket of M<sup>pro</sup> and form stable complexes that allow the estimation of their inhibitory activity. Assay kit analyses revealed that Rutan and its components have effective anti-SARS-CoV-2 M<sup>pro</sup> inhibitory activity.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1518463"},"PeriodicalIF":4.4,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11832515/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143448721","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effects of celastrol on the heart and liver galaninergic system expression in a mouse model of Western-type diet-induced obesity and metabolic dysfunction-associated steatotic liver disease and steatohepatitis. 西洋膳食诱发肥胖和代谢功能障碍相关脂肪性肝病和脂肪性肝炎小鼠模型中,塞拉司醇对心脏和肝脏加兰宁能系统表达的影响。
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-04 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1476994
Nikolina Canová, Jana Šípková, Mahak Arora, Zuzana Pavlíková, Tomáš Kučera, Ondřej Šeda, Tijana Šopin, Tomáš Vacík, Ondřej Slanař

Background: The complexity of the galaninergic system is still not fully understood, especially under specific pre-existing comorbidities related to metabolic dysfunction. A plant-derived triterpenoid celastrol was demonstrated to exert a complex effect on the galaninergic system and to have hepatoprotective and anti-obesity properties. However, the exact molecular mechanisms responsible for these effects remain unclear. Specifically, there are no data on the impact of celastrol on the heart and liver galaninergic system. Therefore, this study aimed to investigate the effects of celastrol on the galaninergic system expression in the heart and liver of mice suffering from diet-induced obesity and metabolic dysfunction-associated steatotic liver disease and steatohepatitis (MASLD/MASH).

Methods: The male mice C57BL/6J were fed a Western-type high-fat diet for 16 and 20 weeks to induce obesity and MASLD/MASH. Celastrol was administered along with a specific diet for the last 4 weeks to evaluate its impact on the progression of these conditions. Moreover, the inhibitor of sterol regulatory element-binding protein 1/2 (SREBP1/2), fatostatin, was also tested to compare its influence on the galaninergic system with celastrol.

Results: The study demonstrates that celastrol treatment was safe and led to a reduction in food and energy intake, body fat and liver weights, and MASLD-to-MASH progression and improved glucose tolerance, serum biochemistry markers, and hepatic lipid peroxidation in mice. Quantitative gene expression originally showed significant regulation of galanin and all three of its receptors (GalR1/2/3) in the heart ventricles and only GalR2 in the liver of obese mice. Celastrol influenced the gene expression of galanin receptors: it downregulated Galr1 in the heart and upregulated Galr2 in the liver and Galr3 in the heart ventricles, potentially affecting energy metabolism, oxidative stress, and inflammation. Fatostatin suppressed gene expression of all the detected members of the galaninergic system in the heart ventricles, depicting the role of SREBP in this process.

Conclusion: These findings suggest that celastrol may beneficially modulate the galaninergic system under obesity and MASLD-to-MASH progression, indicating its potential as a therapeutic agent for disorders associated with metabolic dysfunction.

{"title":"Effects of celastrol on the heart and liver galaninergic system expression in a mouse model of Western-type diet-induced obesity and metabolic dysfunction-associated steatotic liver disease and steatohepatitis.","authors":"Nikolina Canová, Jana Šípková, Mahak Arora, Zuzana Pavlíková, Tomáš Kučera, Ondřej Šeda, Tijana Šopin, Tomáš Vacík, Ondřej Slanař","doi":"10.3389/fphar.2025.1476994","DOIUrl":"10.3389/fphar.2025.1476994","url":null,"abstract":"<p><strong>Background: </strong>The complexity of the galaninergic system is still not fully understood, especially under specific pre-existing comorbidities related to metabolic dysfunction. A plant-derived triterpenoid celastrol was demonstrated to exert a complex effect on the galaninergic system and to have hepatoprotective and anti-obesity properties. However, the exact molecular mechanisms responsible for these effects remain unclear. Specifically, there are no data on the impact of celastrol on the heart and liver galaninergic system. Therefore, this study aimed to investigate the effects of celastrol on the galaninergic system expression in the heart and liver of mice suffering from diet-induced obesity and metabolic dysfunction-associated steatotic liver disease and steatohepatitis (MASLD/MASH).</p><p><strong>Methods: </strong>The male mice C57BL/6J were fed a Western-type high-fat diet for 16 and 20 weeks to induce obesity and MASLD/MASH. Celastrol was administered along with a specific diet for the last 4 weeks to evaluate its impact on the progression of these conditions. Moreover, the inhibitor of sterol regulatory element-binding protein 1/2 (SREBP1/2), fatostatin, was also tested to compare its influence on the galaninergic system with celastrol.</p><p><strong>Results: </strong>The study demonstrates that celastrol treatment was safe and led to a reduction in food and energy intake, body fat and liver weights, and MASLD-to-MASH progression and improved glucose tolerance, serum biochemistry markers, and hepatic lipid peroxidation in mice. Quantitative gene expression originally showed significant regulation of galanin and all three of its receptors (GalR1/2/3) in the heart ventricles and only GalR2 in the liver of obese mice. Celastrol influenced the gene expression of galanin receptors: it downregulated <i>Galr1</i> in the heart and upregulated <i>Galr2</i> in the liver and <i>Galr3</i> in the heart ventricles, potentially affecting energy metabolism, oxidative stress, and inflammation. Fatostatin suppressed gene expression of all the detected members of the galaninergic system in the heart ventricles, depicting the role of SREBP in this process.</p><p><strong>Conclusion: </strong>These findings suggest that celastrol may beneficially modulate the galaninergic system under obesity and MASLD-to-MASH progression, indicating its potential as a therapeutic agent for disorders associated with metabolic dysfunction.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1476994"},"PeriodicalIF":4.4,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11832397/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143448724","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring the anti-metastatic potential of sunitinib and novel analogs in colorectal cancer: insights into HIF-1α mediated metastasis.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-04 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1520881
Fatemeh B Rassouli, Maryam M Matin, Farzin Hadizadeh, Masoud Nejabat, Hossein Allahverdizadeh, Hamidreza Jamali, Shahin Gharedaghi, Halimeh Hassanzadeh

Introduction: Colorectal cancer (CRC) is a prevalent malignancy worldwide with high mortality rate. Metastasis, the primary cause of cancer-related deaths, is attributed to various factors including tumor hypoxia. Due to the urgent demand for potent anti-metastatic agents, we aimed to determine the effects of sunitinib and novel analogs on the metastatic behavior of human CRC cells in hypoxic condition for the first time.

Methods: For in silico analyses, pathogenic targets of metastatic CRC were identified, PPI network was constructed and KEGG pathway enrichment analysis was conducted. The expression of HIF1A was evaluated in seven CRC cell lines, and computational modeling was carried out to define the interaction of sunitinib with HIF-1α. For in vitro studies, analogs of sunitinib were synthesized, and cells were assessed for viability, migration, invasion, MMPs activity and gene expression in hypoxic condition.

Results and discussion: Computational analyses highlighted the importance of HIF-1α as a crucial mediator of metastasis in CRC. Molecular docking and dynamics simulations demonstrated favorable and stable interaction of sunitinib and three novel analogs with HIF-1α PAS-B domain. Volcano plots indicated upregulation of HIF1A in LoVo cells compared to six other CRC cell lines. Findings of in vitro studies revealed considerable inhibitory effects of sunitinib and analogs on LoVo cell migration and invasion in hypoxic condition. Gelatin zymography and qPCR analysis indicated decreased activity of MMP-2 and MMP-9, along with downregulation of EMT transcription factors in hypoxic condition. Current study reports promising anti-metastatic effects of sunitinib and novel analogs on CRC cells, providing foundation for further investigation to combat cancer metastasis.

{"title":"Exploring the anti-metastatic potential of sunitinib and novel analogs in colorectal cancer: insights into HIF-1α mediated metastasis.","authors":"Fatemeh B Rassouli, Maryam M Matin, Farzin Hadizadeh, Masoud Nejabat, Hossein Allahverdizadeh, Hamidreza Jamali, Shahin Gharedaghi, Halimeh Hassanzadeh","doi":"10.3389/fphar.2025.1520881","DOIUrl":"10.3389/fphar.2025.1520881","url":null,"abstract":"<p><strong>Introduction: </strong>Colorectal cancer (CRC) is a prevalent malignancy worldwide with high mortality rate. Metastasis, the primary cause of cancer-related deaths, is attributed to various factors including tumor hypoxia. Due to the urgent demand for potent anti-metastatic agents, we aimed to determine the effects of sunitinib and novel analogs on the metastatic behavior of human CRC cells in hypoxic condition for the first time.</p><p><strong>Methods: </strong>For <i>in silico</i> analyses, pathogenic targets of metastatic CRC were identified, PPI network was constructed and KEGG pathway enrichment analysis was conducted. The expression of <i>HIF1A</i> was evaluated in seven CRC cell lines, and computational modeling was carried out to define the interaction of sunitinib with HIF-1α. For <i>in vitro</i> studies, analogs of sunitinib were synthesized, and cells were assessed for viability, migration, invasion, MMPs activity and gene expression in hypoxic condition.</p><p><strong>Results and discussion: </strong>Computational analyses highlighted the importance of HIF-1α as a crucial mediator of metastasis in CRC. Molecular docking and dynamics simulations demonstrated favorable and stable interaction of sunitinib and three novel analogs with HIF-1α PAS-B domain. Volcano plots indicated upregulation of <i>HIF1A</i> in LoVo cells compared to six other CRC cell lines. Findings of <i>in vitro</i> studies revealed considerable inhibitory effects of sunitinib and analogs on LoVo cell migration and invasion in hypoxic condition. Gelatin zymography and qPCR analysis indicated decreased activity of MMP-2 and MMP-9, along with downregulation of EMT transcription factors in hypoxic condition. Current study reports promising anti-metastatic effects of sunitinib and novel analogs on CRC cells, providing foundation for further investigation to combat cancer metastasis.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1520881"},"PeriodicalIF":4.4,"publicationDate":"2025-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11832664/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143448740","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Angel or devil: the dual roles of 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucopyranoside in the development of liver injury based on integrating pharmacological techniques: a systematic review.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1523713
Jiajie Jiang, Qixiu Wang, Qiang Wu, Bobin Deng, Cui Guo, Jie Chen, Jinhao Zeng, Yaoguang Guo, Xiao Ma

Background and purpose: 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (TSG) exhibits a dualistic pharmacological profile, acting as both a hepatoprotective and hepatotoxic agent, which is intricately linked to its interaction with multiple signaling pathways and its stereoisomeric forms, namely, cis-SG and trans-SG. The purpose of this study is to evaluate both the hepatoprotective and hepatotoxic effects of TSG and give therapeutic guidance.

Methods: This study performed a systematic search of eight databases to identify preclinical literature up until March 2024. The CAMARADES system evaluated evidence quality and bias. STATA and Python were used for statistical analysis, including dose-effect maps, 3D maps and radar charts to show the dose-time-effect relationship of TSG on hepatoprotection and hepatotoxicity.

Results: After a rigorous screening process, a total of 24 studies encompassing 564 rodents were selected for inclusion in this study. The findings revealed that TSG exhibited bidirectional effects on the levels of ALT and AST, while also regulating the levels of ALT, AST, TNF-α, IL-6, serum TG, serum TC, SOD, MDA, IFN-γ, and apoptosis rate. The histological analysis of liver tissue confirmed the regulatory effects of TSG, and a comprehensive analysis revealed the optimal protective dosage range was 27.27-38.81 mg/kg/d and the optimal toxic dosage range was 51.93-76.07 mg/kg/d. TSG exerts the dual effects on liver injury (LI) through the network of Keap1/Nrf2/HO-1/NQO1, NF-κB, PPAR, PI3K/Akt, JAK/STAT and TGF-β pathways.

Conclusion: TSG could mediate the pathways of oxidation, inflammation, and metabolism to result in hepatoprotection (27.27-38.81 mg/kg/d) and hepatotoxicity (51.93-76.07 mg/kg/d).

{"title":"Angel or devil: the dual roles of 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucopyranoside in the development of liver injury based on integrating pharmacological techniques: a systematic review.","authors":"Jiajie Jiang, Qixiu Wang, Qiang Wu, Bobin Deng, Cui Guo, Jie Chen, Jinhao Zeng, Yaoguang Guo, Xiao Ma","doi":"10.3389/fphar.2025.1523713","DOIUrl":"10.3389/fphar.2025.1523713","url":null,"abstract":"<p><strong>Background and purpose: </strong>2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucoside (TSG) exhibits a dualistic pharmacological profile, acting as both a hepatoprotective and hepatotoxic agent, which is intricately linked to its interaction with multiple signaling pathways and its stereoisomeric forms, namely, cis-SG and trans-SG. The purpose of this study is to evaluate both the hepatoprotective and hepatotoxic effects of TSG and give therapeutic guidance.</p><p><strong>Methods: </strong>This study performed a systematic search of eight databases to identify preclinical literature up until March 2024. The CAMARADES system evaluated evidence quality and bias. STATA and Python were used for statistical analysis, including dose-effect maps, 3D maps and radar charts to show the dose-time-effect relationship of TSG on hepatoprotection and hepatotoxicity.</p><p><strong>Results: </strong>After a rigorous screening process, a total of 24 studies encompassing 564 rodents were selected for inclusion in this study. The findings revealed that TSG exhibited bidirectional effects on the levels of ALT and AST, while also regulating the levels of ALT, AST, TNF-α, IL-6, serum TG, serum TC, SOD, MDA, IFN-γ, and apoptosis rate. The histological analysis of liver tissue confirmed the regulatory effects of TSG, and a comprehensive analysis revealed the optimal protective dosage range was 27.27-38.81 mg/kg/d and the optimal toxic dosage range was 51.93-76.07 mg/kg/d. TSG exerts the dual effects on liver injury (LI) through the network of Keap1/Nrf2/HO-1/NQO1, NF-κB, PPAR, PI3K/Akt, JAK/STAT and TGF-β pathways.</p><p><strong>Conclusion: </strong>TSG could mediate the pathways of oxidation, inflammation, and metabolism to result in hepatoprotection (27.27-38.81 mg/kg/d) and hepatotoxicity (51.93-76.07 mg/kg/d).</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1523713"},"PeriodicalIF":4.4,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830817/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440495","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Postpartum changes in maternal physiology and milk composition: a comprehensive database for developing lactation physiologically-based pharmacokinetic models.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1517069
Neel Deferm, Jean Dinh, Amita Pansari, Masoud Jamei, Khaled Abduljalil

Introduction: Pharmacotherapy during lactation often lacks reliable drug safety data, resulting in delayed treatment or early cessation of breastfeeding. In silico tools, such as physiologically-based pharmacokinetic (PBPK) models, can help to bridge this knowledge gap. To increase the accuracy of these models, it is essential to account for the physiological changes that occur throughout the postpartum period.

Methods: This study aimed to collect and analyze data on the longitudinal changes in various physiological parameters that can affect drug distribution into breast milk during lactation. Following meta-analysis of the collated data, mathematical functions were fitted to the available data for each parameter. The best-performing functions were selected through numerical and visual diagnostics.

Results and discussion: The literature search identified 230 studies, yielding a dataset of 36,689 data points from 20,801 postpartum women, covering data from immediately after childbirth to 12 months postpartum. Sufficient data were obtained to describe postpartum changes in maternal plasma volume, breast volume, cardiac output, glomerular filtration rate, haematocrit, human serum albumin, alpha-1-acid glycoprotein, milk pH, milk volume, milk fat, milk protein, milk water content, and daily infant milk intake. Although data beyond 7 months postpartum were limited for some parameters, mathematical functions were generated for all parameters. These functions can be integrated into lactation PBPK models to increase their predictive power and better inform medication efficacy and safety for breastfeeding women.

{"title":"Postpartum changes in maternal physiology and milk composition: a comprehensive database for developing lactation physiologically-based pharmacokinetic models.","authors":"Neel Deferm, Jean Dinh, Amita Pansari, Masoud Jamei, Khaled Abduljalil","doi":"10.3389/fphar.2025.1517069","DOIUrl":"10.3389/fphar.2025.1517069","url":null,"abstract":"<p><strong>Introduction: </strong>Pharmacotherapy during lactation often lacks reliable drug safety data, resulting in delayed treatment or early cessation of breastfeeding. <i>In silico</i> tools, such as physiologically-based pharmacokinetic (PBPK) models, can help to bridge this knowledge gap. To increase the accuracy of these models, it is essential to account for the physiological changes that occur throughout the postpartum period.</p><p><strong>Methods: </strong>This study aimed to collect and analyze data on the longitudinal changes in various physiological parameters that can affect drug distribution into breast milk during lactation. Following meta-analysis of the collated data, mathematical functions were fitted to the available data for each parameter. The best-performing functions were selected through numerical and visual diagnostics.</p><p><strong>Results and discussion: </strong>The literature search identified 230 studies, yielding a dataset of 36,689 data points from 20,801 postpartum women, covering data from immediately after childbirth to 12 months postpartum. Sufficient data were obtained to describe postpartum changes in maternal plasma volume, breast volume, cardiac output, glomerular filtration rate, haematocrit, human serum albumin, alpha-1-acid glycoprotein, milk pH, milk volume, milk fat, milk protein, milk water content, and daily infant milk intake. Although data beyond 7 months postpartum were limited for some parameters, mathematical functions were generated for all parameters. These functions can be integrated into lactation PBPK models to increase their predictive power and better inform medication efficacy and safety for breastfeeding women.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1517069"},"PeriodicalIF":4.4,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830814/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Leveraging AI to automate detection and quantification of extrachromosomal DNA to decode drug responses.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 eCollection Date: 2024-01-01 DOI: 10.3389/fphar.2024.1516621
Kohen Goble, Aarav Mehta, Damien Guilbaud, Jacob Fessler, Jingting Chen, William Nenad, Christina G Ford, Oliver Cope, Darby Cheng, William Dennis, Nithya Gurumurthy, Yue Wang, Kriti Shukla, Elizabeth Brunk

Introduction: Traditional drug discovery efforts primarily target rapid, reversible protein-mediated adaptations to counteract cancer cell resistance. However, cancer cells also utilize DNA-based strategies, often perceived as slow, irreversible changes like point mutations or drug-resistant clone selection. Extrachromosomal DNA (ecDNA), in contrast, represents a rapid, reversible, and predictable DNA alteration critical for cancer's adaptive response.

Methods: In this study, we developed a novel post-processing pipeline for automated detection and quantification of ecDNA in metaphase Fluorescence in situ Hybridization (FISH) images, leveraging the Microscopy Image Analyzer (MIA) tool. This pipeline is tailored to monitor ecDNA dynamics during drug treatment.

Results: Our approach effectively quantified ecDNA changes, providing a robust framework for analyzing the adaptive responses of cancer cells under therapeutic pressure.

Discussion: The pipeline not only serves as a valuable resource for automating ecDNA detection in metaphase FISH images but also highlights the role of ecDNA in facilitating swift and reversible adaptation to epigenetic remodeling agents such as JQ1.

{"title":"Leveraging AI to automate detection and quantification of extrachromosomal DNA to decode drug responses.","authors":"Kohen Goble, Aarav Mehta, Damien Guilbaud, Jacob Fessler, Jingting Chen, William Nenad, Christina G Ford, Oliver Cope, Darby Cheng, William Dennis, Nithya Gurumurthy, Yue Wang, Kriti Shukla, Elizabeth Brunk","doi":"10.3389/fphar.2024.1516621","DOIUrl":"10.3389/fphar.2024.1516621","url":null,"abstract":"<p><strong>Introduction: </strong>Traditional drug discovery efforts primarily target rapid, reversible protein-mediated adaptations to counteract cancer cell resistance. However, cancer cells also utilize DNA-based strategies, often perceived as slow, irreversible changes like point mutations or drug-resistant clone selection. Extrachromosomal DNA (ecDNA), in contrast, represents a rapid, reversible, and predictable DNA alteration critical for cancer's adaptive response.</p><p><strong>Methods: </strong>In this study, we developed a novel post-processing pipeline for automated detection and quantification of ecDNA in metaphase Fluorescence in situ Hybridization (FISH) images, leveraging the Microscopy Image Analyzer (MIA) tool. This pipeline is tailored to monitor ecDNA dynamics during drug treatment.</p><p><strong>Results: </strong>Our approach effectively quantified ecDNA changes, providing a robust framework for analyzing the adaptive responses of cancer cells under therapeutic pressure.</p><p><strong>Discussion: </strong>The pipeline not only serves as a valuable resource for automating ecDNA detection in metaphase FISH images but also highlights the role of ecDNA in facilitating swift and reversible adaptation to epigenetic remodeling agents such as JQ1.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"15 ","pages":"1516621"},"PeriodicalIF":4.4,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830698/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440493","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rutaecarpine alleviates hepatic ischemia‒reperfusion injury in liver transplantation by inhibiting inflammatory response and oxidative stress.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1539744
Yan Liu, Feng Qi, Lun-Jian Xiang, Zhu-Jun Yi, Sheng-Wei Li

Background: Donation after circulatory death (DCD) livers are limited by mandatory warm ischemia and are more susceptible to ischemia‒reperfusion injury (IRI). Inflammation and oxidative stress play key roles in the development of hepatic IRI, and Rutaecarpine (Rut) has anti-inflammatory and anti-oxidative stress effects. The aim of this study was to investigate whether Rut can alleviate hepatic IRI in liver transplantation (LT) and to explore the underlying mechanisms.

Methods: Rat DCD LT and oxygen-glucose deprivation/reoxygenation (OGD/R) cell models were established to clarify the effect of Rut on hepatic IRI. The key molecules involved in the hepatoprotective effects of Rut were identified through joint analysis of data from LT patients and drug targets. The target was further validated by in silico, in vivo and in vitro experiments.

Results: Rut significantly alleviated liver dysfunction, pathological injury, and apoptosis and improved the survival rate of the rats subjected to LT. In addition, Rut significantly inhibited inflammatory response and oxidative stress. Rut also had similar effects on OGD/R-induced hepatocyte injury. Mechanistically, bioinformatics analysis and in vivo and in vitro experiments revealed that PDE4B may be a key target by which Rut exerts its protective effect, and molecular docking and cellular thermal shift assay confirmed this result. The function of PDE4B was studied via gene intervention technology, and the results showed that PDE4B can aggravate hepatic IRI. Furthermore, PDE4B overexpression abrogated the protective effect of Rut on the liver in LT.

Conclusion: Rut alleviates hepatic IRI by targeting PDE4B to inhibit inflammation and oxidative stress. These findings highlight the potential of Rut as a drug candidate for the treatment of patients undergoing LT.

{"title":"Rutaecarpine alleviates hepatic ischemia‒reperfusion injury in liver transplantation by inhibiting inflammatory response and oxidative stress.","authors":"Yan Liu, Feng Qi, Lun-Jian Xiang, Zhu-Jun Yi, Sheng-Wei Li","doi":"10.3389/fphar.2025.1539744","DOIUrl":"10.3389/fphar.2025.1539744","url":null,"abstract":"<p><strong>Background: </strong>Donation after circulatory death (DCD) livers are limited by mandatory warm ischemia and are more susceptible to ischemia‒reperfusion injury (IRI). Inflammation and oxidative stress play key roles in the development of hepatic IRI, and Rutaecarpine (Rut) has anti-inflammatory and anti-oxidative stress effects. The aim of this study was to investigate whether Rut can alleviate hepatic IRI in liver transplantation (LT) and to explore the underlying mechanisms.</p><p><strong>Methods: </strong>Rat DCD LT and oxygen-glucose deprivation/reoxygenation (OGD/R) cell models were established to clarify the effect of Rut on hepatic IRI. The key molecules involved in the hepatoprotective effects of Rut were identified through joint analysis of data from LT patients and drug targets. The target was further validated by <i>in silico</i>, <i>in vivo</i> and <i>in vitro</i> experiments.</p><p><strong>Results: </strong>Rut significantly alleviated liver dysfunction, pathological injury, and apoptosis and improved the survival rate of the rats subjected to LT. In addition, Rut significantly inhibited inflammatory response and oxidative stress. Rut also had similar effects on OGD/R-induced hepatocyte injury. Mechanistically, bioinformatics analysis and <i>in vivo</i> and <i>in vitro</i> experiments revealed that PDE4B may be a key target by which Rut exerts its protective effect, and molecular docking and cellular thermal shift assay confirmed this result. The function of PDE4B was studied <i>via</i> gene intervention technology, and the results showed that PDE4B can aggravate hepatic IRI. Furthermore, PDE4B overexpression abrogated the protective effect of Rut on the liver in LT.</p><p><strong>Conclusion: </strong>Rut alleviates hepatic IRI by targeting PDE4B to inhibit inflammation and oxidative stress. These findings highlight the potential of Rut as a drug candidate for the treatment of patients undergoing LT.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1539744"},"PeriodicalIF":4.4,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830625/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440537","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of novel and highly potent small molecule inhibitors targeting FLT3-ITD for the treatment of acute myeloid leukemia using structure-based virtual screening and biological evaluation.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1511257
Kun Shi, Ye Hong, Huajing Liu, Xiaotian Yang, Fengzhen Wang, Yanming Zhang

Considering the essential role of FLT3-ITD mutations in the development of acute myeloid leukemia (AML), the research and development of FLT3 inhibitors hold significant therapeutic potential. In this study, we identified a novel, highly potent small molecule inhibitor, FLIN-4, targeting FLT3 through structure-based virtual screening. Notably, FLIN-4 showed exceptional inhibitory effects in kinase activity inhibition assays, exhibiting a potent inhibitory effect against FLT3 (IC50 = 1.07 ± 0.04 nM). This potency was significantly superior to that of the known positive inhibitor Midostaurin, showing approximately 27 times higher inhibitory potency. Molecular dynamics simulations have confirmed the stable interaction between FLIN-4 and FLT3. Furthermore, cytotoxicity assays revealed that FLIN-4 has significant anti-proliferative activity against the AML cell line MV4-11 (IC50 = 1.31 ± 0.06 nM). Overall, these data suggest that FLIN-4, as a potential therapeutic candidate for AML, is valuable for further research and development.

{"title":"Discovery of novel and highly potent small molecule inhibitors targeting FLT3-ITD for the treatment of acute myeloid leukemia using structure-based virtual screening and biological evaluation.","authors":"Kun Shi, Ye Hong, Huajing Liu, Xiaotian Yang, Fengzhen Wang, Yanming Zhang","doi":"10.3389/fphar.2025.1511257","DOIUrl":"10.3389/fphar.2025.1511257","url":null,"abstract":"<p><p>Considering the essential role of FLT3-ITD mutations in the development of acute myeloid leukemia (AML), the research and development of FLT3 inhibitors hold significant therapeutic potential. In this study, we identified a novel, highly potent small molecule inhibitor, FLIN-4, targeting FLT3 through structure-based virtual screening. Notably, FLIN-4 showed exceptional inhibitory effects in kinase activity inhibition assays, exhibiting a potent inhibitory effect against FLT3 (IC<sub>50</sub> = 1.07 ± 0.04 nM). This potency was significantly superior to that of the known positive inhibitor Midostaurin, showing approximately 27 times higher inhibitory potency. Molecular dynamics simulations have confirmed the stable interaction between FLIN-4 and FLT3. Furthermore, cytotoxicity assays revealed that FLIN-4 has significant anti-proliferative activity against the AML cell line MV4-11 (IC<sub>50</sub> = 1.31 ± 0.06 nM). Overall, these data suggest that FLIN-4, as a potential therapeutic candidate for AML, is valuable for further research and development.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1511257"},"PeriodicalIF":4.4,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830721/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440516","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bioprinted hydrogels in bone regeneration: a bibliometric analysis.
IF 4.4 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-03 eCollection Date: 2025-01-01 DOI: 10.3389/fphar.2025.1532629
Huijie Zhang, Xiaoyu Li, Zhenyu Jia, Kun Jiao, Chen Liu, Zixiang Deng, Yushu Bai, Xianzhao Wei, Xiaoyi Zhou

Background: The application of bioprinted hydrogels in the field of bone regeneration is garnering increasing attention. The objective of this study is to provide a comprehensive overview of the current research status, hotspots and research directions in this field through bibliometric methods, and to predict the development trend of this field.

Methods: A search was conducted on 27 December 2024, for papers published on the Web of Science from 2010 to 2025. We used the bibliometrix package in the software program R to analyze the retrieved data and VOSviewer and CiteSpace to visualize hotspots and research trends in bioprinted hydrogels for bone regeneration.

Results: We identified and reviewed 684 articles published in this field between 2010 and 2025. A total of 811 institutions and 1,166 researchers from 41 countries/regions contributed to these publications. Among them, China led in terms of the number of articles published, single-country publications (SCP), and multi-country publications (MCP). Our bibliometric-based visualization analysis revealed that the mechanical properties and osteogenic differentiation capacity of biomaterials have been a focal research topic over the past decade, while emerging research has also concentrated on the in vitro fabrication of stem cells for bone regeneration and osteogenic differentiation, particularly the precise application of in situ stem cell-loaded bioprinted organoids.

Conclusion: This study provides an in-depth analysis of the research trajectory in the application of bioprinted hydrogels for bone regeneration. The number of research papers in this field is increasing annually, and the main research hotspots include bone regeneration, 3D printing, scaffolds, and hydrogels. Future research directions may focus on gelatin, additive manufacturing, and growth factors. Additionally, international collaboration is essential to enhance the effectiveness of bioprinted hydrogels in bone regeneration applications.

{"title":"Bioprinted hydrogels in bone regeneration: a bibliometric analysis.","authors":"Huijie Zhang, Xiaoyu Li, Zhenyu Jia, Kun Jiao, Chen Liu, Zixiang Deng, Yushu Bai, Xianzhao Wei, Xiaoyi Zhou","doi":"10.3389/fphar.2025.1532629","DOIUrl":"10.3389/fphar.2025.1532629","url":null,"abstract":"<p><strong>Background: </strong>The application of bioprinted hydrogels in the field of bone regeneration is garnering increasing attention. The objective of this study is to provide a comprehensive overview of the current research status, hotspots and research directions in this field through bibliometric methods, and to predict the development trend of this field.</p><p><strong>Methods: </strong>A search was conducted on 27 December 2024, for papers published on the Web of Science from 2010 to 2025. We used the bibliometrix package in the software program R to analyze the retrieved data and VOSviewer and CiteSpace to visualize hotspots and research trends in bioprinted hydrogels for bone regeneration.</p><p><strong>Results: </strong>We identified and reviewed 684 articles published in this field between 2010 and 2025. A total of 811 institutions and 1,166 researchers from 41 countries/regions contributed to these publications. Among them, China led in terms of the number of articles published, single-country publications (SCP), and multi-country publications (MCP). Our bibliometric-based visualization analysis revealed that the mechanical properties and osteogenic differentiation capacity of biomaterials have been a focal research topic over the past decade, while emerging research has also concentrated on the <i>in vitro</i> fabrication of stem cells for bone regeneration and osteogenic differentiation, particularly the precise application of <i>in situ</i> stem cell-loaded bioprinted organoids.</p><p><strong>Conclusion: </strong>This study provides an in-depth analysis of the research trajectory in the application of bioprinted hydrogels for bone regeneration. The number of research papers in this field is increasing annually, and the main research hotspots include bone regeneration, 3D printing, scaffolds, and hydrogels. Future research directions may focus on gelatin, additive manufacturing, and growth factors. Additionally, international collaboration is essential to enhance the effectiveness of bioprinted hydrogels in bone regeneration applications.</p>","PeriodicalId":12491,"journal":{"name":"Frontiers in Pharmacology","volume":"16 ","pages":"1532629"},"PeriodicalIF":4.4,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143440498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Frontiers in Pharmacology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1