首页 > 最新文献

Pharmacological research最新文献

英文 中文
An in vitro pharmacogenomic approach reveals subtype-specific therapeutic vulnerabilities in atypical teratoid/rhabdoid tumors (AT/RT).
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-15 DOI: 10.1016/j.phrs.2025.107660
David Pauck, Daniel Picard, Mara Maue, Kübra Taban, Viktoria Marquardt, Lena Blümel, Jasmin Bartl, Nan Qin, Nadezhda Kubon, Dominik Schöndorf, Frauke-Dorothee Meyer, Johanna Theruvath, Siddhartha Mitra, Martin Hasselblatt, Michael C Frühwald, Guido Reifenberger, Marc Remke

Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant embryonal brain tumor driven by genetic alterations inactivating the SMARCB1 or, less commonly, the SMARCA4 gene. Large-scale molecular profiling studies have identified distinct molecular subtypes termed AT/RT-TYR, -SHH and -MYC. Despite the increasing knowledge of AT/RT biology, curative treatment options are still lacking for certain risk groups and outcomes of these patients remain poor. We performed an in vitro high-throughput drug screen of 768 small molecule drugs covering conventional chemotherapeutic agents and late-stage developmental drugs in 13 AT/RT cell lines and determined intra- and inter-entity differential responses to unravel specific vulnerabilities. Our data demonstrated in vitro preferential activity of mitogen-activated protein kinase kinase (MEK) and mouse double minute 2 homolog (MDM2) inhibitors in AT/RT cell lines compared to other high-grade brain tumor cell lines including medulloblastoma and malignant glioma models. Moreover, we were able to link distinct drug response patterns to AT/RT molecular subtypes through integration of drug response data with large-scale DNA methylation and RNASeq-based expression profiles. Subtype-dependent drug response profiles demonstrated sensitivity of AT/RT-SHH cell lines to B-cell lymphoma 2 (BCL2) and heat shock protein 90 (HSP90) inhibitors, and increased activity of microtubule inhibitors, kinesin spindle protein (KSP) inhibitors, and the eukaryotic translation initiation factor 4E (eIF4E) inhibitor briciclib in a subset of AT/RT-MYC cell lines. In summary, our in vitro pharmacogenomic approach revealed preclinical evidence of tumor type- and subtype-specific therapeutic vulnerabilities in AT/RT cell lines that may inform future in vivo and clinical evaluations of novel pharmacological strategies.

{"title":"An in vitro pharmacogenomic approach reveals subtype-specific therapeutic vulnerabilities in atypical teratoid/rhabdoid tumors (AT/RT).","authors":"David Pauck, Daniel Picard, Mara Maue, Kübra Taban, Viktoria Marquardt, Lena Blümel, Jasmin Bartl, Nan Qin, Nadezhda Kubon, Dominik Schöndorf, Frauke-Dorothee Meyer, Johanna Theruvath, Siddhartha Mitra, Martin Hasselblatt, Michael C Frühwald, Guido Reifenberger, Marc Remke","doi":"10.1016/j.phrs.2025.107660","DOIUrl":"https://doi.org/10.1016/j.phrs.2025.107660","url":null,"abstract":"<p><p>Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant embryonal brain tumor driven by genetic alterations inactivating the SMARCB1 or, less commonly, the SMARCA4 gene. Large-scale molecular profiling studies have identified distinct molecular subtypes termed AT/RT-TYR, -SHH and -MYC. Despite the increasing knowledge of AT/RT biology, curative treatment options are still lacking for certain risk groups and outcomes of these patients remain poor. We performed an in vitro high-throughput drug screen of 768 small molecule drugs covering conventional chemotherapeutic agents and late-stage developmental drugs in 13 AT/RT cell lines and determined intra- and inter-entity differential responses to unravel specific vulnerabilities. Our data demonstrated in vitro preferential activity of mitogen-activated protein kinase kinase (MEK) and mouse double minute 2 homolog (MDM2) inhibitors in AT/RT cell lines compared to other high-grade brain tumor cell lines including medulloblastoma and malignant glioma models. Moreover, we were able to link distinct drug response patterns to AT/RT molecular subtypes through integration of drug response data with large-scale DNA methylation and RNASeq-based expression profiles. Subtype-dependent drug response profiles demonstrated sensitivity of AT/RT-SHH cell lines to B-cell lymphoma 2 (BCL2) and heat shock protein 90 (HSP90) inhibitors, and increased activity of microtubule inhibitors, kinesin spindle protein (KSP) inhibitors, and the eukaryotic translation initiation factor 4E (eIF4E) inhibitor briciclib in a subset of AT/RT-MYC cell lines. In summary, our in vitro pharmacogenomic approach revealed preclinical evidence of tumor type- and subtype-specific therapeutic vulnerabilities in AT/RT cell lines that may inform future in vivo and clinical evaluations of novel pharmacological strategies.</p>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":" ","pages":"107660"},"PeriodicalIF":9.1,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143441609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lactobacillus vaginalis alleviates DSS induced colitis by regulating the gut microbiota and increasing the production of 3-indoleacrylic acid.
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-15 DOI: 10.1016/j.phrs.2025.107663
Zhuoya Wang, Tian Liu, Li Liu, Jian Xie, Furui Tang, Yimin Pi, Yuchun Zhong, Zhidong He, Wenming Zhang, Cihua Zheng

Ulcerative colitis (UC) is a chronic inflammatory disorder, and its incidence is experiencing an upward trend worldwide. UC can result in gut microbiota dysbiosis, impaired intestinal epithelial barrier, and systemic inflammation, for all of which there is presently no definitive treatment available. Lactobacillus is known to regulate gut microbiota and related metabolites to intervene in the development of UC. The objective of this study was to explore the underlying mechanism through which a novel probiotic, Lactobacillus vaginalis, alleviates DSS-induced colitis. Specifically, L. vaginalis were found to ameliorate the DSS-induced UC phenotype, restore intestinal microbiota balance and intestinal barrier function, and elevate the levels of 3-indoleacrylic acid (IAA) in mouse feces. Furthermore, fecal microbiota transplantation and fecal filtrate transplantation provide additional evidence that L. vaginalis alleviate DSS-induced colitis through metabolic products. Additionally, IAA has been shown to alleviate DSS-induced colitis symptoms, decrease inflammatory responses, and enhance intestinal barrier function. Finally, our findings confirm that L. vaginal and metabolites possess the capability to regulate the immune microenvironment in mice with colitis. And the RNA-seq analysis suggests that L. vaginal may play a pivotal role in alleviating colitis by modulating the PPAR signaling pathway. In conclusion, our findings suggest that oral administration of L. vaginalis alleviates DSS induced colonic inflammation by increasing the levels of IAA. L. vaginalis, as an emerging probiotic, provides a potential therapeutic strategy for clinical UC.

{"title":"Lactobacillus vaginalis alleviates DSS induced colitis by regulating the gut microbiota and increasing the production of 3-indoleacrylic acid.","authors":"Zhuoya Wang, Tian Liu, Li Liu, Jian Xie, Furui Tang, Yimin Pi, Yuchun Zhong, Zhidong He, Wenming Zhang, Cihua Zheng","doi":"10.1016/j.phrs.2025.107663","DOIUrl":"https://doi.org/10.1016/j.phrs.2025.107663","url":null,"abstract":"<p><p>Ulcerative colitis (UC) is a chronic inflammatory disorder, and its incidence is experiencing an upward trend worldwide. UC can result in gut microbiota dysbiosis, impaired intestinal epithelial barrier, and systemic inflammation, for all of which there is presently no definitive treatment available. Lactobacillus is known to regulate gut microbiota and related metabolites to intervene in the development of UC. The objective of this study was to explore the underlying mechanism through which a novel probiotic, Lactobacillus vaginalis, alleviates DSS-induced colitis. Specifically, L. vaginalis were found to ameliorate the DSS-induced UC phenotype, restore intestinal microbiota balance and intestinal barrier function, and elevate the levels of 3-indoleacrylic acid (IAA) in mouse feces. Furthermore, fecal microbiota transplantation and fecal filtrate transplantation provide additional evidence that L. vaginalis alleviate DSS-induced colitis through metabolic products. Additionally, IAA has been shown to alleviate DSS-induced colitis symptoms, decrease inflammatory responses, and enhance intestinal barrier function. Finally, our findings confirm that L. vaginal and metabolites possess the capability to regulate the immune microenvironment in mice with colitis. And the RNA-seq analysis suggests that L. vaginal may play a pivotal role in alleviating colitis by modulating the PPAR signaling pathway. In conclusion, our findings suggest that oral administration of L. vaginalis alleviates DSS induced colonic inflammation by increasing the levels of IAA. L. vaginalis, as an emerging probiotic, provides a potential therapeutic strategy for clinical UC.</p>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":" ","pages":"107663"},"PeriodicalIF":9.1,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143441611","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neuronal PCSK9 regulates cognitive performances via the modulation of ApoER2 synaptic localization
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-12 DOI: 10.1016/j.phrs.2025.107652
Silvia Pelucchi , Lorenzo Da Dalt , Giulia De Cesare , Ramona Stringhi , Laura D’Andrea , Filippo La Greca , Clara Cambria , Lina Vandermeulen , Elisa Zianni , Stefano Musardo , Silvia Roda , Fabrizia Bonacina , Sofia Nasini , Maria Giovanna Lupo , Nicola Ferri , Stefano Comai , Fabrizio Gardoni , Flavia Antonucci , Diego Scheggia , Monica Di Luca , Elena Marcello
PCSK9 promotes the degradation of the low-density lipoprotein receptors and its inhibition by monoclonal antibodies or gene silencing approaches results in the reduction of plasma cholesterol levels coupled to that of cardiovascular events. Notably, while the liver is the primary source of circulating PCSK9, this protein is also abundantly expressed in the brain. However, its specific functions in the brain remain poorly understood. Here, we demonstrate that neuron-specific PCSK9 knockout mice exhibit impaired cognitive function, driven by alterations in hippocampal synapse morphology and synaptic plasticity mechanisms, coupled to spatial memory deficits. Among PCSK9 targets, we identified ApoER2 as the primary mediator of PCSK9-dependent effects on synaptic function. In neuronal cultures, PCSK9 downregulation affects ApoER2 synaptic membrane localization and lipid droplets abundance. In conclusion, our results highlight the critical role of neuronal PCSK9 in modulating synaptic ApoER2 and reveal the detrimental effects of its deficiency on synaptic function and cognitive performance. Our results shed light on the complex biology of PCSK9, crucial for evaluating side effects of PCSK9 inhibition and for developing new therapies targeting PCSK9 for brain disorders.
{"title":"Neuronal PCSK9 regulates cognitive performances via the modulation of ApoER2 synaptic localization","authors":"Silvia Pelucchi ,&nbsp;Lorenzo Da Dalt ,&nbsp;Giulia De Cesare ,&nbsp;Ramona Stringhi ,&nbsp;Laura D’Andrea ,&nbsp;Filippo La Greca ,&nbsp;Clara Cambria ,&nbsp;Lina Vandermeulen ,&nbsp;Elisa Zianni ,&nbsp;Stefano Musardo ,&nbsp;Silvia Roda ,&nbsp;Fabrizia Bonacina ,&nbsp;Sofia Nasini ,&nbsp;Maria Giovanna Lupo ,&nbsp;Nicola Ferri ,&nbsp;Stefano Comai ,&nbsp;Fabrizio Gardoni ,&nbsp;Flavia Antonucci ,&nbsp;Diego Scheggia ,&nbsp;Monica Di Luca ,&nbsp;Elena Marcello","doi":"10.1016/j.phrs.2025.107652","DOIUrl":"10.1016/j.phrs.2025.107652","url":null,"abstract":"<div><div>PCSK9 promotes the degradation of the low-density lipoprotein receptors and its inhibition by monoclonal antibodies or gene silencing approaches results in the reduction of plasma cholesterol levels coupled to that of cardiovascular events. Notably, while the liver is the primary source of circulating PCSK9, this protein is also abundantly expressed in the brain. However, its specific functions in the brain remain poorly understood. Here, we demonstrate that neuron-specific PCSK9 knockout mice exhibit impaired cognitive function, driven by alterations in hippocampal synapse morphology and synaptic plasticity mechanisms, coupled to spatial memory deficits. Among PCSK9 targets, we identified ApoER2 as the primary mediator of PCSK9-dependent effects on synaptic function. In neuronal cultures, PCSK9 downregulation affects ApoER2 synaptic membrane localization and lipid droplets abundance. In conclusion, our results highlight the critical role of neuronal PCSK9 in modulating synaptic ApoER2 and reveal the detrimental effects of its deficiency on synaptic function and cognitive performance. Our results shed light on the complex biology of PCSK9, crucial for evaluating side effects of PCSK9 inhibition and for developing new therapies targeting PCSK9 for brain disorders.</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107652"},"PeriodicalIF":9.1,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143426049","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
Quality and composition control of complex TCM preparations through a novel “Herbs-in vivo Compounds-Targets-Pathways” network methodology: The case of Lianhuaqingwen capsules
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-12 DOI: 10.1016/j.phrs.2025.107650
Simian Chen , Hui Qi , Chunyan Zhu , Yiheng Zhao , Bo Jiao , Yun Tan , Yuexin Yang , Tongxing Wang , Yunlong Hou , Binxin Dai , Dandan Zhang , Hairong Zhang , Junyu Zhang , Xiaojuan Jiang , Xiaodan Guo , Xiaoyu Qian , Caixia Yuan , Xue Bai , Jiayun Chen , Suping Wang , Caisheng Wu
Lianhuaqingwen (LHQW) capsules, a Chinese patent medicine composed of 13 herbal ingredients, are widely used for respiratory diseases. However, the complex composition of LHQW poses challenges in assessing its quality and consistency. In this study, a comprehensive network of LHQW was constructed by integrating Digital RNA with pertUrbation of Genes (DRUG)-seq, RNA sequencing, and pharmacodynamic data. This approach enables rapid and systematic screening of compounds in LHQW that exhibit high-exposure in vivo and significant activity potential, serving as potential quality control markers. Specifically, DRUG-seq was employed to evaluate gene expression alterations in peripheral blood mononuclear cells derived from healthy volunteers. Ultrahigh-performance liquid chromatography coupled with high-resolution mass spectrometry (UPLC-HRMS) identified 505 compounds in LHQW-treated rats. Additionally, absorption, distribution, metabolism, and excretion (ADME) profiles were plotted for 27 primary components of LHQW. Furthermore, an HPLC-MS/MS method quantified 46 compounds from LHQW, with 15 of them identified as potential quality markers with high exposure levels. These markers exhibited significant inhibitory effects on lipopolysaccharide (LPS)-induced pneumonia in mice, with mechanisms predicted by RNA-seq and verified by RT-qPCR. In summary, this study successfully constructed an "Herbs- in vivo Compounds-targets-pathways" network, offering novel insights into the mechanisms of LHQW and establishing a foundation for enhancing quality control measures.
{"title":"Quality and composition control of complex TCM preparations through a novel “Herbs-in vivo Compounds-Targets-Pathways” network methodology: The case of Lianhuaqingwen capsules","authors":"Simian Chen ,&nbsp;Hui Qi ,&nbsp;Chunyan Zhu ,&nbsp;Yiheng Zhao ,&nbsp;Bo Jiao ,&nbsp;Yun Tan ,&nbsp;Yuexin Yang ,&nbsp;Tongxing Wang ,&nbsp;Yunlong Hou ,&nbsp;Binxin Dai ,&nbsp;Dandan Zhang ,&nbsp;Hairong Zhang ,&nbsp;Junyu Zhang ,&nbsp;Xiaojuan Jiang ,&nbsp;Xiaodan Guo ,&nbsp;Xiaoyu Qian ,&nbsp;Caixia Yuan ,&nbsp;Xue Bai ,&nbsp;Jiayun Chen ,&nbsp;Suping Wang ,&nbsp;Caisheng Wu","doi":"10.1016/j.phrs.2025.107650","DOIUrl":"10.1016/j.phrs.2025.107650","url":null,"abstract":"<div><div>Lianhuaqingwen (LHQW) capsules, a Chinese patent medicine composed of 13 herbal ingredients, are widely used for respiratory diseases. However, the complex composition of LHQW poses challenges in assessing its quality and consistency. In this study, a comprehensive network of LHQW was constructed by integrating Digital RNA with pertUrbation of Genes (DRUG)-seq, RNA sequencing, and pharmacodynamic data. This approach enables rapid and systematic screening of compounds in LHQW that exhibit high-exposure <em>in vivo</em> and significant activity potential, serving as potential quality control markers. Specifically, DRUG-seq was employed to evaluate gene expression alterations in peripheral blood mononuclear cells derived from healthy volunteers. Ultrahigh-performance liquid chromatography coupled with high-resolution mass spectrometry (UPLC-HRMS) identified 505 compounds in LHQW-treated rats. Additionally, absorption, distribution, metabolism, and excretion (ADME) profiles were plotted for 27 primary components of LHQW. Furthermore, an HPLC-MS/MS method quantified 46 compounds from LHQW, with 15 of them identified as potential quality markers with high exposure levels. These markers exhibited significant inhibitory effects on lipopolysaccharide (LPS)-induced pneumonia in mice, with mechanisms predicted by RNA-seq and verified by RT-qPCR. In summary, this study successfully constructed an \"Herbs- <em>in vivo</em> Compounds-targets-pathways\" network, offering novel insights into the mechanisms of LHQW and establishing a foundation for enhancing quality control measures.</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107650"},"PeriodicalIF":9.1,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143414770","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
Gut microbiome-derived indole-3-carboxaldehyde regulates stress vulnerability in chronic restraint stress by activating aryl hydrocarbon receptors
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-12 DOI: 10.1016/j.phrs.2025.107654
Congcong Chen , Qiang Xiao , Zhaoyi Wen , Fenfang Gong , Huang Zhan , Jian Liu , Hui Li , Yukun Jiao
Chronic stress constitutes a major precipitating factor for Major Depressive Disorder (MDD), and comprehending individual differences in stress responses is crucial for the development of effective intervention strategies for MDD. Recent studies indicate that an individual's vulnerability to chronic stress is closely associated with gut microbiota composition, but the underlying mechanisms remain unclear. This study aims to investigate whether the gut microbiota and its metabolites can serve as gut-brain signaling molecules and explores how the gut microbiota affects stress sensitivity. Here, we showed that gut microbiome-derived indole-3-carboxaldehyde (I3C) can act as a gut-brain signaling molecule that links tryptophan metabolism by gut microbes to stress vulnerability in the host. First, we identified a specific reduction in gut microbiome-derived I3C levels in the hippocampus and colon through untargeted and targeted metabolomic analyses. Then, the study of gut microbiota suggested that the relative abundance of lactobacillus was reduced significantly in stress-susceptible rats, whereas fecal microbiota transplantation regulates stress vulnerability. Furthermore, supplementation with I3C and the representative I3C-producing strain, Lactobacillus reuteri, was shown to alleviate depression-like behaviors induced by chronic stress. Further research confirms that I3C can inhibit neuroinflammation and promote hippocampal neurogenesis through the aryl hydrocarbon receptors (AhR) signal pathway, thereby mitigating the host's susceptibility to stress. In conclusion, our findings elucidate that the gut microbiome-derived-I3C can help buffer the host's stress through the AhR/SOCS2/NF-κB/NLRP3 pathway, providing a gut-brain signaling basis for emotional behavior.
{"title":"Gut microbiome-derived indole-3-carboxaldehyde regulates stress vulnerability in chronic restraint stress by activating aryl hydrocarbon receptors","authors":"Congcong Chen ,&nbsp;Qiang Xiao ,&nbsp;Zhaoyi Wen ,&nbsp;Fenfang Gong ,&nbsp;Huang Zhan ,&nbsp;Jian Liu ,&nbsp;Hui Li ,&nbsp;Yukun Jiao","doi":"10.1016/j.phrs.2025.107654","DOIUrl":"10.1016/j.phrs.2025.107654","url":null,"abstract":"<div><div>Chronic stress constitutes a major precipitating factor for Major Depressive Disorder (MDD), and comprehending individual differences in stress responses is crucial for the development of effective intervention strategies for MDD. Recent studies indicate that an individual's vulnerability to chronic stress is closely associated with gut microbiota composition, but the underlying mechanisms remain unclear. This study aims to investigate whether the gut microbiota and its metabolites can serve as gut-brain signaling molecules and explores how the gut microbiota affects stress sensitivity. Here, we showed that gut microbiome-derived indole-3-carboxaldehyde (I3C) can act as a gut-brain signaling molecule that links tryptophan metabolism by gut microbes to stress vulnerability in the host. First, we identified a specific reduction in gut microbiome-derived I3C levels in the hippocampus and colon through untargeted and targeted metabolomic analyses. Then, the study of gut microbiota suggested that the relative abundance of <em>lactobacillus</em> was reduced significantly in stress-susceptible rats, whereas fecal microbiota transplantation regulates stress vulnerability. Furthermore, supplementation with I3C and the representative I3C-producing strain, <em>Lactobacillus reuteri</em>, was shown to alleviate depression-like behaviors induced by chronic stress. Further research confirms that I3C can inhibit neuroinflammation and promote hippocampal neurogenesis through the aryl hydrocarbon receptors (AhR) signal pathway, thereby mitigating the host's susceptibility to stress. In conclusion, our findings elucidate that the gut microbiome-derived-I3C can help buffer the host's stress through the AhR/SOCS2/NF-κB/NLRP3 pathway, providing a gut-brain signaling basis for emotional behavior.</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107654"},"PeriodicalIF":9.1,"publicationDate":"2025-02-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143394383","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
The role of HIF-1α in hypoxic metabolic reprogramming in osteoarthritis
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-11 DOI: 10.1016/j.phrs.2025.107649
Jie Zhang , Peng Gao , Wei-Rong Chang , Jia-Yi Song , Fang-Yu An , Yu-Jie Wang , Zhi-Pan Xiao , Hua Jin , Xu-Hui Zhang , Chun-Lu Yan
The joint dysfunction caused by osteoarthritis (OA) is increasingly becoming a major challenge in global healthcare, and there is currently no effective strategy to prevent the progression of OA. Therefore, better elucidating the relevant mechanisms of OA occurrence and development will provide theoretical basis for formulating new prevention and control strategies. Due to long-term exposure of cartilage tissue to the hypoxic microenvironment of joints, metabolic reprogramming changes occur. Hypoxia-inducible factor-1alpha (HIF-1α), as a core gene regulating hypoxia response in vivo, plays an important regulatory role in the hypoxic metabolism of chondrocytes. HIF-1α adapts to the hypoxic microenvironment by regulating metabolic reprogramming changes such as glycolysis, oxidative phosphorylation (OXPHOS), amino acid metabolism, and lipid metabolism in OA chondrocytes. In addition, HIF-1α also regulates macrophage polarization and synovial inflammation, chondrocytes degeneration and extracellular matrix (ECM) degradation, subchondral bone remodeling and angiogenesis in the hypoxic microenvironment of OA, and affects the pathophysiological progression of OA. Consequently, the regulation of chondrocytes metabolic reprogramming by HIF-1α has become an important therapeutic target for OA. Therefore, this article reviews the mechanism of hypoxia affecting chondrocyte metabolic reprogramming, focusing on the regulatory mechanism of HIF-1α on chondrocyte metabolic reprogramming, and summarizes potential effective ingredients or targets targeting chondrocyte metabolic reprogramming, in order to provide more beneficial basis for the prevention and treatment of clinical OA and the development of effective drugs.
{"title":"The role of HIF-1α in hypoxic metabolic reprogramming in osteoarthritis","authors":"Jie Zhang ,&nbsp;Peng Gao ,&nbsp;Wei-Rong Chang ,&nbsp;Jia-Yi Song ,&nbsp;Fang-Yu An ,&nbsp;Yu-Jie Wang ,&nbsp;Zhi-Pan Xiao ,&nbsp;Hua Jin ,&nbsp;Xu-Hui Zhang ,&nbsp;Chun-Lu Yan","doi":"10.1016/j.phrs.2025.107649","DOIUrl":"10.1016/j.phrs.2025.107649","url":null,"abstract":"<div><div>The joint dysfunction caused by osteoarthritis (OA) is increasingly becoming a major challenge in global healthcare, and there is currently no effective strategy to prevent the progression of OA. Therefore, better elucidating the relevant mechanisms of OA occurrence and development will provide theoretical basis for formulating new prevention and control strategies. Due to long-term exposure of cartilage tissue to the hypoxic microenvironment of joints, metabolic reprogramming changes occur. Hypoxia-inducible factor-1alpha (HIF-1α), as a core gene regulating hypoxia response in vivo, plays an important regulatory role in the hypoxic metabolism of chondrocytes. HIF-1α adapts to the hypoxic microenvironment by regulating metabolic reprogramming changes such as glycolysis, oxidative phosphorylation (OXPHOS), amino acid metabolism, and lipid metabolism in OA chondrocytes. In addition, HIF-1α also regulates macrophage polarization and synovial inflammation, chondrocytes degeneration and extracellular matrix (ECM) degradation, subchondral bone remodeling and angiogenesis in the hypoxic microenvironment of OA, and affects the pathophysiological progression of OA. Consequently, the regulation of chondrocytes metabolic reprogramming by HIF-1α has become an important therapeutic target for OA. Therefore, this article reviews the mechanism of hypoxia affecting chondrocyte metabolic reprogramming, focusing on the regulatory mechanism of HIF-1α on chondrocyte metabolic reprogramming, and summarizes potential effective ingredients or targets targeting chondrocyte metabolic reprogramming, in order to provide more beneficial basis for the prevention and treatment of clinical OA and the development of effective drugs.</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107649"},"PeriodicalIF":9.1,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143414774","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
Targeting TIPARP in ischemic stroke: A promising therapeutic strategy and future directions
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-11 DOI: 10.1016/j.phrs.2025.107651
Yinuo Pan, Xiande Ma
{"title":"Targeting TIPARP in ischemic stroke: A promising therapeutic strategy and future directions","authors":"Yinuo Pan,&nbsp;Xiande Ma","doi":"10.1016/j.phrs.2025.107651","DOIUrl":"10.1016/j.phrs.2025.107651","url":null,"abstract":"","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107651"},"PeriodicalIF":9.1,"publicationDate":"2025-02-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143394384","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
The efficacy of sodium-glucose transporter 2 inhibitors in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-08 DOI: 10.1016/j.phrs.2025.107647
Hongsheng Li , Yanli Hou , Wenyong Xin , Lina Ding , Ying Yang , Yikun Zhang , Wenqi Wu , Zhibin Wang , Wenyu Ding
The efficacy of sodium-glucose transporter 2 (SGLT-2) inhibitors for nonalcoholic fatty liver disease (NAFLD) is unclear. Therefore, we conducted a systematic review and meta-analysis to evaluate SGLT-2 inhibitors efficacy for NAFLD treatment. We systematically searched major electronic databases (PubMed, Cochrane Library, Web of Science, Embase) from inception until 11/2023, identifying randomized controlled trials (RCTs) of SGLT-2 inhibitors treatment for patients with NAFLD. The mean differences (MD or SMD) and 95 % confidence intervals (CIs) were calculated via random-effects models. Eleven articles (n = 805 patients with NAFLD) were included in this study. Of these, 408 participants received SGLT-2 inhibitors, while 397 participants were in the control group. SGLT-2 inhibitors significantly reduced liver enzyme levels, including aspartate alanine aminotransferase (ALT) (MD [95 % CI]; −9.31 U/L [-13.41, −5.21], p < 0.00001), aspartate aminotransferase (AST) (MD [95 % CI]; −6.06 U/L [-10.98, −1.15], p = 0.02), and gamma-glutamyltransferase (GGT) (MD [95 % CI]; −11.72 U/L [-15.65, −7.80], p < 0.00001). SGLT-2 inhibitors intervention was also associated with significant reductions in body weight (MD [95 % CI]; −2.72 kg [-3.49, −1.95], p < 0.00001) and BMI (MD [95 % CI]; −1.11 kg/m2 [-1.39, −0.82], p < 0.00001) and improvements in glycaemic indices, triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C). However, no significant changes in total cholesterol (TC) or low-density lipoprotein cholesterol (LDL-C) were observed. The meta-analysis revealed a beneficial effect of SGLT-2 inhibitors on liver functions and body weight, BMI, TG, HDL-C, and glucose homeostasis in patients with NAFLD, indicating that SGLT-2 inhibitors might be a clinical therapeutic strategy for these patients, especially individuals with concurrent type 2 diabetes mellitus (T2DM).
{"title":"The efficacy of sodium-glucose transporter 2 inhibitors in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis","authors":"Hongsheng Li ,&nbsp;Yanli Hou ,&nbsp;Wenyong Xin ,&nbsp;Lina Ding ,&nbsp;Ying Yang ,&nbsp;Yikun Zhang ,&nbsp;Wenqi Wu ,&nbsp;Zhibin Wang ,&nbsp;Wenyu Ding","doi":"10.1016/j.phrs.2025.107647","DOIUrl":"10.1016/j.phrs.2025.107647","url":null,"abstract":"<div><div>The efficacy of sodium-glucose transporter 2 (SGLT-2) inhibitors for nonalcoholic fatty liver disease (NAFLD) is unclear. Therefore, we conducted a systematic review and meta-analysis to evaluate SGLT-2 inhibitors efficacy for NAFLD treatment. We systematically searched major electronic databases (PubMed, Cochrane Library, Web of Science, Embase) from inception until 11/2023, identifying randomized controlled trials (RCTs) of SGLT-2 inhibitors treatment for patients with NAFLD. The mean differences (MD or SMD) and 95 % confidence intervals (<em>CIs</em>) were calculated via random-effects models. Eleven articles (n = 805 patients with NAFLD) were included in this study. Of these, 408 participants received SGLT-2 inhibitors, while 397 participants were in the control group. SGLT-2 inhibitors significantly reduced liver enzyme levels, including aspartate alanine aminotransferase (ALT) (MD [95 % <em>CI</em>]; −9.31 U/L [-13.41, −5.21], <em>p</em> &lt; 0.00001), aspartate aminotransferase (AST) (MD [95 % <em>CI</em>]; −6.06 U/L [-10.98, −1.15], <em>p</em> = 0.02), and gamma-glutamyltransferase (GGT) (MD [95 % <em>CI</em>]; −11.72 U/L [-15.65, −7.80], <em>p</em> &lt; 0.00001). SGLT-2 inhibitors intervention was also associated with significant reductions in body weight (MD [95 % <em>CI</em>]; −2.72 kg [-3.49, −1.95], <em>p</em> &lt; 0.00001) and BMI (MD [95 % <em>CI</em>]; −1.11 kg/m<sup>2</sup> [-1.39, −0.82], <em>p</em> &lt; 0.00001) and improvements in glycaemic indices, triglyceride (TG) and high-density lipoprotein cholesterol (HDL-C). However, no significant changes in total cholesterol (TC) or low-density lipoprotein cholesterol (LDL-C) were observed. The meta-analysis revealed a beneficial effect of SGLT-2 inhibitors on liver functions and body weight, BMI, TG, HDL-C, and glucose homeostasis in patients with NAFLD, indicating that SGLT-2 inhibitors might be a clinical therapeutic strategy for these patients, especially individuals with concurrent type 2 diabetes mellitus (T2DM).</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107647"},"PeriodicalIF":9.1,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143377290","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
The essential role of sphingolipids in TRPC5 ion channel localization and functionality within lipid rafts
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-07 DOI: 10.1016/j.phrs.2025.107648
Junliang Wan , Zhenying Hu , Huaiyi Zhu , Jingyi Li , Ziyuan Zheng , Zhitao Deng , Junyan Lu , Yu Chen , Gui-Lan Chen , Bo Zeng , Jin Zhang , Jingjing Duan
Sphingolipids are critical components of cellular membranes that play a pivotal role in modulating ion channel function by forming lipid rafts that stabilize and localize these channels. These lipids regulate membrane fluidity and protein-lipid interactions, directly influencing ion channel activity, trafficking, and signaling pathways essential for maintaining cellular homeostasis. Despite their fundamental role, the impact of sphingolipids on ion channel functionality, particularly within the nervous system, remains insufficiently understood. This study addresses this gap by examining the influence of sphingolipids on transient receptor potential canonical 5 (TRPC5), a key brain ion channel involved in sensory transduction and linked to conditions such as obesity, anxiety, and postpartum depression when disrupted. In this study, we demonstrate that TRPC5 is localized within lipid rafts. Inhibition of sphingolipid synthesis through myrioncin (Myr), the sphingomyelin synthase 2 inhibitor Ly93, or D,L-erythro-PDMP hydrochloride (PMDP) significantly disrupts TRPC5 localization at the plasma membrane. Treatment with lipid raft disruptors methyl-β-cyclodextrin (MCD) or sphingomyelin phosphodiesterase 3 (SMPD3), in conjunction with sphingolipid synthesis inhibitors, led to decreased TRPC5-mediated calcium flux and currents. This highlights the critical importance of TRPC5 localization in lipid rafts for its functionality. Furthermore, LC-MS/MS-based sphingolipidomics has shown that a balanced sphingolipid profile is crucial for channel function. Alterations in sphingolipid metabolism, especially the deficiency of sphingomyelin and glycosphingolipids, may primarily disrupt lipid raft structure. Interactions between amino acid residues with phenyl ring side chains and lipids at the inner and outer plasma membrane edges serve as ‘fixators’, anchoring TRPC5 channels within lipid rafts. Given the structural similarities among TRP channels, we propose that sphingolipid metabolic homeostasis may universally influence TRP channel activity, potentially explaining diverse neurological disorder phenotypes associated with sphingolipid metabolism disruptions.
{"title":"The essential role of sphingolipids in TRPC5 ion channel localization and functionality within lipid rafts","authors":"Junliang Wan ,&nbsp;Zhenying Hu ,&nbsp;Huaiyi Zhu ,&nbsp;Jingyi Li ,&nbsp;Ziyuan Zheng ,&nbsp;Zhitao Deng ,&nbsp;Junyan Lu ,&nbsp;Yu Chen ,&nbsp;Gui-Lan Chen ,&nbsp;Bo Zeng ,&nbsp;Jin Zhang ,&nbsp;Jingjing Duan","doi":"10.1016/j.phrs.2025.107648","DOIUrl":"10.1016/j.phrs.2025.107648","url":null,"abstract":"<div><div>Sphingolipids are critical components of cellular membranes that play a pivotal role in modulating ion channel function by forming lipid rafts that stabilize and localize these channels. These lipids regulate membrane fluidity and protein-lipid interactions, directly influencing ion channel activity, trafficking, and signaling pathways essential for maintaining cellular homeostasis. Despite their fundamental role, the impact of sphingolipids on ion channel functionality, particularly within the nervous system, remains insufficiently understood. This study addresses this gap by examining the influence of sphingolipids on transient receptor potential canonical 5 (TRPC5), a key brain ion channel involved in sensory transduction and linked to conditions such as obesity, anxiety, and postpartum depression when disrupted. In this study, we demonstrate that TRPC5 is localized within lipid rafts. Inhibition of sphingolipid synthesis through myrioncin (Myr), the sphingomyelin synthase 2 inhibitor Ly93, or D,L-erythro-PDMP hydrochloride (PMDP) significantly disrupts TRPC5 localization at the plasma membrane. Treatment with lipid raft disruptors methyl-<em>β</em>-cyclodextrin (MCD) or sphingomyelin phosphodiesterase 3 (SMPD3), in conjunction with sphingolipid synthesis inhibitors, led to decreased TRPC5-mediated calcium flux and currents. This highlights the critical importance of TRPC5 localization in lipid rafts for its functionality. Furthermore, LC-MS/MS-based sphingolipidomics has shown that a balanced sphingolipid profile is crucial for channel function. Alterations in sphingolipid metabolism, especially the deficiency of sphingomyelin and glycosphingolipids, may primarily disrupt lipid raft structure. Interactions between amino acid residues with phenyl ring side chains and lipids at the inner and outer plasma membrane edges serve as ‘fixators’, anchoring TRPC5 channels within lipid rafts. Given the structural similarities among TRP channels, we propose that sphingolipid metabolic homeostasis may universally influence TRP channel activity, potentially explaining diverse neurological disorder phenotypes associated with sphingolipid metabolism disruptions.</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107648"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143382925","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
Pharmacological regression of atherosclerotic plaque in patients with type 2 diabetes
IF 9.1 2区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-02-05 DOI: 10.1016/j.phrs.2025.107635
Loredana Bucciarelli , Daniele Andreini , Giulio Stefanini , Roberta Maria Fiorina , Marco Francone , Federica Catapano , Maria Elena Lunati , Edoardo Conte , Davide Marchetti , Paolo Fiorina
Atherosclerosis of the coronary arteries continues to be one of the major global health burdens and acute coronary syndrome is responsible annually for at least 30 % of all deaths globally. Acute coronary syndrome may be the consequence of thrombus formation after erosion or rupture of obstructive or non-obstructive atherosclerotic plaque. The rupture of plaques is mostly caused by mechanical stress usually called cap fatigue. Vulnerable plaques are characterized by a softer atheromatous core and a thinner fibrous cap, with inflammation and hypercholesterolemia playing a crucial role in the atherothrombotic process. Based on animal studies that extend back to the 1920s, regression of atherosclerotic plaques in humans has just started to be considered and pursued. The idea that the human atherosclerotic plaques could regress at all met an important resistance over the decades; indeed, advanced plaques contain components, such as necrosis, calcification and fibrosis, which are hard to be removed. However, new animal models and imaging technics allowed a more complete and accurate quantitative assessment of plaque volume and are shedding new light on atherosclerosis regression. In this review, we are revisiting the existence of atherosclerosis regression in preclinical and clinical studies, with a focus on the latest mechanistic insights and on the newest pharmacological agents, particularly in patients with diabetes. Interestingly, we suggested that based on literature insights and preclinical studies, a combination of drugs to target hyperglycemia, dyslipidemia and inflammation may be desirable for a fast-track Pharmacological regression of atherosclerotic plaque in patients with type 2 diabetes.
{"title":"Pharmacological regression of atherosclerotic plaque in patients with type 2 diabetes","authors":"Loredana Bucciarelli ,&nbsp;Daniele Andreini ,&nbsp;Giulio Stefanini ,&nbsp;Roberta Maria Fiorina ,&nbsp;Marco Francone ,&nbsp;Federica Catapano ,&nbsp;Maria Elena Lunati ,&nbsp;Edoardo Conte ,&nbsp;Davide Marchetti ,&nbsp;Paolo Fiorina","doi":"10.1016/j.phrs.2025.107635","DOIUrl":"10.1016/j.phrs.2025.107635","url":null,"abstract":"<div><div>Atherosclerosis of the coronary arteries continues to be one of the major global health burdens and acute coronary syndrome is responsible annually for at least 30 % of all deaths globally. Acute coronary syndrome may be the consequence of thrombus formation after erosion or rupture of obstructive or non-obstructive atherosclerotic plaque. The rupture of plaques is mostly caused by mechanical stress usually called cap fatigue. Vulnerable plaques are characterized by a softer atheromatous core and a thinner fibrous cap, with inflammation and hypercholesterolemia playing a crucial role in the atherothrombotic process. Based on animal studies that extend back to the 1920s, regression of atherosclerotic plaques in humans has just started to be considered and pursued. The idea that the human atherosclerotic plaques could regress at all met an important resistance over the decades; indeed, advanced plaques contain components, such as necrosis, calcification and fibrosis, which are hard to be removed. However, new animal models and imaging technics allowed a more complete and accurate quantitative assessment of plaque volume and are shedding new light on atherosclerosis regression. In this review, we are revisiting the existence of atherosclerosis regression in preclinical and clinical studies, with a focus on the latest mechanistic insights and on the newest pharmacological agents, particularly in patients with diabetes. Interestingly, we suggested that based on literature insights and preclinical studies, a combination of drugs to target hyperglycemia, dyslipidemia and inflammation may be desirable for a fast-track Pharmacological regression of atherosclerotic plaque in patients with type 2 diabetes.</div></div>","PeriodicalId":19918,"journal":{"name":"Pharmacological research","volume":"213 ","pages":"Article 107635"},"PeriodicalIF":9.1,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143348348","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
Pharmacological research
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