首页 > 最新文献

Journal of Neurochemistry最新文献

英文 中文
Careful Examination of a Novel Azobenzene Paroxetine Derivative and Its Interactions With Biogenic Amine Transporters 仔细研究一种新型偶氮苯帕罗西汀衍生物及其与生物胺转运体的相互作用
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-24 DOI: 10.1111/jnc.70068
Dominik Dreier, Oliver John V. Belleza, Katharina Schlögl, Stefanie Kickinger, Eva Hellsberg, Felix P. Mayer, Walter Sandtner, Philipp Mikšovsky, Matthias Schittmayer, Yuntao Hu, Kathrin Jäntsch, Marion Holy, Gerhard F. Ecker, Harald H. Sitte, Marko D. Mihovilovic

The serotonin transporter (SERT) belongs to the family of neurotransmitter sodium symporters (NSS), together with other neurotransmitter transporters for norepinephrine, dopamine, glycine, and GABA. The main physiological role of SERT is the retrieval of previously released serotonin from the synaptic cleft. Thereby, SERT plays an important role in regulating the extracellular serotonin concentration and maintaining serotonergic neurotransmission. This process can be influenced by molecules acting as serotonin uptake inhibitors, like paroxetine. Here, we report the development of a novel photoswitchable paroxetine derivative and its pharmacological interaction profile with SERT as a tool compound for the light-induced control of SERT. Based on the azo-extension strategy, the photoswitchable moiety was formed at the former position of the fluoro substituent in paroxetine. The resulting azo-paroxetine (9) was easily and reversibly switched between active (Z) and inactive (E) configurations and remained stable between these configurations: serotonin uptake was inhibited more than 12 times more potently by the active (Z)-configuration having a sub μM IC50 value. This was supported by electrophysiological patch-clamp recordings in the whole-cell configuration and docking studies. No significant toxic impact of azo-paroxetine (9) and no off-target activity at the norepinephrine transporter (NET), human GABA transporter subtypes 1 and 3, and rat GAT1 were observed. Our results demonstrate that the activity of SERT can be reversibly manipulated by the optopharmacological agent azo-paroxetine (9). This compound can thus be applied as a tool for the selective manipulation of SERT in central or peripheral investigations, further benefiting from its low probability for compound-related off-target effects.

{"title":"Careful Examination of a Novel Azobenzene Paroxetine Derivative and Its Interactions With Biogenic Amine Transporters","authors":"Dominik Dreier,&nbsp;Oliver John V. Belleza,&nbsp;Katharina Schlögl,&nbsp;Stefanie Kickinger,&nbsp;Eva Hellsberg,&nbsp;Felix P. Mayer,&nbsp;Walter Sandtner,&nbsp;Philipp Mikšovsky,&nbsp;Matthias Schittmayer,&nbsp;Yuntao Hu,&nbsp;Kathrin Jäntsch,&nbsp;Marion Holy,&nbsp;Gerhard F. Ecker,&nbsp;Harald H. Sitte,&nbsp;Marko D. Mihovilovic","doi":"10.1111/jnc.70068","DOIUrl":"https://doi.org/10.1111/jnc.70068","url":null,"abstract":"<p>The serotonin transporter (SERT) belongs to the family of neurotransmitter sodium symporters (NSS), together with other neurotransmitter transporters for norepinephrine, dopamine, glycine, and GABA. The main physiological role of SERT is the retrieval of previously released serotonin from the synaptic cleft. Thereby, SERT plays an important role in regulating the extracellular serotonin concentration and maintaining serotonergic neurotransmission. This process can be influenced by molecules acting as serotonin uptake inhibitors, like paroxetine. Here, we report the development of a novel photoswitchable paroxetine derivative and its pharmacological interaction profile with SERT as a tool compound for the light-induced control of SERT. Based on the azo-extension strategy, the photoswitchable moiety was formed at the former position of the fluoro substituent in paroxetine. The resulting azo-paroxetine (<b>9</b>) was easily and reversibly switched between active (<i>Z</i>) and inactive (<i>E</i>) configurations and remained stable between these configurations: serotonin uptake was inhibited more than 12 times more potently by the active (<i>Z</i>)-configuration having a sub μM IC<sub>50</sub> value. This was supported by electrophysiological patch-clamp recordings in the whole-cell configuration and docking studies. No significant toxic impact of azo-paroxetine (<b>9</b>) and no off-target activity at the norepinephrine transporter (NET), human GABA transporter subtypes 1 and 3, and rat GAT1 were observed. Our results demonstrate that the activity of SERT can be reversibly manipulated by the optopharmacological agent azo-paroxetine (<b>9</b>). This compound can thus be applied as a tool for the selective manipulation of SERT in central or peripheral investigations, further benefiting from its low probability for compound-related off-target effects.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70068","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143866051","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Inhibition of cGMP-Signalling Rescues Retinal Ganglion Cells From Axotomy-Induced Degeneration
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-24 DOI: 10.1111/jnc.70072
Katia Ihadadene, Azdah Hamed A Fallatah, Yu Zhu, Arianna Tolone, François Paquet-Durand

The axons of retinal ganglion cells (RGCs) form the optic nerve, which relays visual information to the brain. RGC degeneration is the root cause of a variety of blinding diseases linked to optic nerve damage, including glaucoma, the second leading cause of blindness worldwide. The underlying cellular mechanisms of RGC degeneration are largely unclear; yet, they have been connected to excessive production of the signalling molecule nitric oxide (NO) by nitric oxide synthase (NOS). NO activates soluble guanylate cyclase (sGC), which subsequently produces the second messenger cyclic guanosine monophosphate (cGMP). This, in turn, activates protein kinase G (PKG), which can phosphorylate downstream protein targets. To study the role of NO/cGMP/PKG signalling in RGC degeneration, we used organotypic retinal explant cultures in which the optic nerve had been severed. We assessed the activity of NOS, RGC death and survival at different times after optic nerve transection. While NOS activity was high right after optic nerve transection, significant RGC loss occurred with a 24–48-h delay. We then treated retinal explants with inhibitors selectively targeting either NOS, sGC, PKG, or Kv1.3 and Kv1.6 voltage-gated potassium channels. While all four treatments reduced RGC death, the PKG inhibitor CN238 and the Kv-channel blocker Margatoxin (MrgX) showed the most pronounced rescue effects. Our results confirm an involvement of NO/cGMP/PKG signalling in RGC degeneration, highlight the potential of PKG and Kv1-channel targeting drugs for treatment development, and further suggest organotypic retinal explant cultures as a useful model for investigations into optic nerve damage.

{"title":"Inhibition of cGMP-Signalling Rescues Retinal Ganglion Cells From Axotomy-Induced Degeneration","authors":"Katia Ihadadene,&nbsp;Azdah Hamed A Fallatah,&nbsp;Yu Zhu,&nbsp;Arianna Tolone,&nbsp;François Paquet-Durand","doi":"10.1111/jnc.70072","DOIUrl":"https://doi.org/10.1111/jnc.70072","url":null,"abstract":"<p>The axons of retinal ganglion cells (RGCs) form the optic nerve, which relays visual information to the brain. RGC degeneration is the root cause of a variety of blinding diseases linked to optic nerve damage, including glaucoma, the second leading cause of blindness worldwide. The underlying cellular mechanisms of RGC degeneration are largely unclear; yet, they have been connected to excessive production of the signalling molecule nitric oxide (NO) by nitric oxide synthase (NOS). NO activates soluble guanylate cyclase (sGC), which subsequently produces the second messenger cyclic guanosine monophosphate (cGMP). This, in turn, activates protein kinase G (PKG), which can phosphorylate downstream protein targets. To study the role of NO/cGMP/PKG signalling in RGC degeneration, we used organotypic retinal explant cultures in which the optic nerve had been severed. We assessed the activity of NOS, RGC death and survival at different times after optic nerve transection. While NOS activity was high right after optic nerve transection, significant RGC loss occurred with a 24–48-h delay. We then treated retinal explants with inhibitors selectively targeting either NOS, sGC, PKG, or Kv1.3 and Kv1.6 voltage-gated potassium channels. While all four treatments reduced RGC death, the PKG inhibitor CN238 and the Kv-channel blocker Margatoxin (MrgX) showed the most pronounced rescue effects. Our results confirm an involvement of NO/cGMP/PKG signalling in RGC degeneration, highlight the potential of PKG and Kv1-channel targeting drugs for treatment development, and further suggest organotypic retinal explant cultures as a useful model for investigations into optic nerve damage.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70072","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143866050","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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 Double-Edged Sword: The Complex Function of Enteric Glial Cells in Neurodegenerative Diseases
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-23 DOI: 10.1111/jnc.70069
Ingrid Prata Mendonça, Christina Alves Peixoto

Over the past two decades, a growing number of studies have been conducted on the role of bidirectional communication through the gut–brain axis in the development of neurodegenerative diseases. These studies were driven by the curious fact that all of these diseases present varying degrees of intestinal involvement included in their wide range of symptoms. A population of cells belonging to the ENS, called enteric glial cells (EGCs), appears to actively participate in this communication between the intestine and the brain, but acting in a dualistic manner, sometimes in reactive gliosis releasing inflammatory mediators, sometimes promoting homeostasis and resilience in the face of inflammatory injuries. To date, the intracellular mechanisms that define the transcriptional profile expressed in EGCs in each situation have not yet been elucidated. This review proposes a discussion on: (1) the complex role of distinct phenotypes of enteric glial cells involved in neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) and multiple sclerosis (MS); and (2) innovative strategies such as IDO/TDO inhibitors, Brazil nuts, caffeic acid, polyphenols, among others, that act on EGCs and have the potential to treat neurodegenerative diseases.

{"title":"The Double-Edged Sword: The Complex Function of Enteric Glial Cells in Neurodegenerative Diseases","authors":"Ingrid Prata Mendonça,&nbsp;Christina Alves Peixoto","doi":"10.1111/jnc.70069","DOIUrl":"https://doi.org/10.1111/jnc.70069","url":null,"abstract":"<p>Over the past two decades, a growing number of studies have been conducted on the role of bidirectional communication through the gut–brain axis in the development of neurodegenerative diseases. These studies were driven by the curious fact that all of these diseases present varying degrees of intestinal involvement included in their wide range of symptoms. A population of cells belonging to the ENS, called enteric glial cells (EGCs), appears to actively participate in this communication between the intestine and the brain, but acting in a dualistic manner, sometimes in reactive gliosis releasing inflammatory mediators, sometimes promoting homeostasis and resilience in the face of inflammatory injuries. To date, the intracellular mechanisms that define the transcriptional profile expressed in EGCs in each situation have not yet been elucidated. This review proposes a discussion on: (1) the complex role of distinct phenotypes of enteric glial cells involved in neurodegenerative diseases, such as Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) and multiple sclerosis (MS); and (2) innovative strategies such as IDO/TDO inhibitors, Brazil nuts, caffeic acid, polyphenols, among others, that act on EGCs and have the potential to treat neurodegenerative diseases.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70069","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143861881","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Molecular Mechanisms of Propofol-Induced Cognitive Impairment: Suppression of Critical Hippocampal Pathways
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-23 DOI: 10.1111/jnc.70070
Xueyue Zhou, Shasha Dong, Yuhai Xu

Propofol, a commonly used anesthetic, is known to cause postoperative cognitive dysfunction (POCD), particularly after prolonged or high-dose administration. Its effects on neural remodeling in the hippocampal region, which is vital for cognitive function, remain poorly understood. This study employs single-cell RNA sequencing (scRNA-seq) and high-throughput transcriptomic analysis to elucidate the molecular mechanisms by which propofol impairs hippocampal neural remodeling. Our findings indicate that propofol suppresses the (5-Hydroxytryptamine Receptor 1A/Glutamate Receptor 2/Phosphoinositide 3-Kinase Regulatory Subunit 1) HTR1A/GRIA2/PIK3R1 signaling pathway, contributing to cognitive dysfunction in mice. In vitro experiments reveal that propofol treatment reduces the expression of HTR1A/GRIA2/PIK3R1-related factors, decreases neuronal activity and synaptic plasticity, and increases apoptosis and inflammation. In vivo experiments demonstrate significant impairments in spatial memory and learning abilities in mice treated with propofol. These results provide new insights into the long-term effects of anesthetic drugs and offer a scientific basis for their judicious use in clinical practice. The study highlights potential strategies and targets for preventing and treating POCD, emphasizing the importance of understanding the molecular mechanisms underlying anesthetic-induced cognitive dysfunction.

{"title":"Molecular Mechanisms of Propofol-Induced Cognitive Impairment: Suppression of Critical Hippocampal Pathways","authors":"Xueyue Zhou,&nbsp;Shasha Dong,&nbsp;Yuhai Xu","doi":"10.1111/jnc.70070","DOIUrl":"https://doi.org/10.1111/jnc.70070","url":null,"abstract":"<div>\u0000 \u0000 <p>Propofol, a commonly used anesthetic, is known to cause postoperative cognitive dysfunction (POCD), particularly after prolonged or high-dose administration. Its effects on neural remodeling in the hippocampal region, which is vital for cognitive function, remain poorly understood. This study employs single-cell RNA sequencing (scRNA-seq) and high-throughput transcriptomic analysis to elucidate the molecular mechanisms by which propofol impairs hippocampal neural remodeling. Our findings indicate that propofol suppresses the (5-Hydroxytryptamine Receptor 1A/Glutamate Receptor 2/Phosphoinositide 3-Kinase Regulatory Subunit 1) HTR1A/GRIA2/PIK3R1 signaling pathway, contributing to cognitive dysfunction in mice. In vitro experiments reveal that propofol treatment reduces the expression of HTR1A/GRIA2/PIK3R1-related factors, decreases neuronal activity and synaptic plasticity, and increases apoptosis and inflammation. In vivo experiments demonstrate significant impairments in spatial memory and learning abilities in mice treated with propofol. These results provide new insights into the long-term effects of anesthetic drugs and offer a scientific basis for their judicious use in clinical practice. The study highlights potential strategies and targets for preventing and treating POCD, emphasizing the importance of understanding the molecular mechanisms underlying anesthetic-induced cognitive dysfunction.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>\u0000 </div>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143861882","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Preface to the Special Issue “Touchscreen Testing to Investigate the Neurochemistry of Cognition”
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-17 DOI: 10.1111/jnc.70071
Miguel Skirzewski, Lisa M. Saksida, Tim J. Bussey

Touchscreen-based methodologies have enabled significant advancements in cognitive neuroscience by providing standardized, translationally relevant assessments of advanced cognitive functions in rodent models. This special issue highlights the potential of these systems to bridge animal and human research, providing insights into the neurochemical and biological mechanisms underlying cognition. The included studies explore diverse applications, from understanding the cognitive impacts of chronic stress and maternal immune activation to evaluating the effectiveness of novel therapeutics and assessing cross-species cognitive testing approaches that enhance translational relevance. By combining touchscreen technologies with cutting-edge approaches like electrophysiology and open science databases, these contributions underscore the critical role of automated systems in advancing translational research. Together, they lay the foundation for novel therapeutic strategies to address cognitive deficits in brain disorders.

{"title":"Preface to the Special Issue “Touchscreen Testing to Investigate the Neurochemistry of Cognition”","authors":"Miguel Skirzewski,&nbsp;Lisa M. Saksida,&nbsp;Tim J. Bussey","doi":"10.1111/jnc.70071","DOIUrl":"https://doi.org/10.1111/jnc.70071","url":null,"abstract":"<p>Touchscreen-based methodologies have enabled significant advancements in cognitive neuroscience by providing standardized, translationally relevant assessments of advanced cognitive functions in rodent models. This special issue highlights the potential of these systems to bridge animal and human research, providing insights into the neurochemical and biological mechanisms underlying cognition. The included studies explore diverse applications, from understanding the cognitive impacts of chronic stress and maternal immune activation to evaluating the effectiveness of novel therapeutics and assessing cross-species cognitive testing approaches that enhance translational relevance. By combining touchscreen technologies with cutting-edge approaches like electrophysiology and open science databases, these contributions underscore the critical role of automated systems in advancing translational research. Together, they lay the foundation for novel therapeutic strategies to address cognitive deficits in brain disorders.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70071","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143840666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Liver Diseases and Brain Disorders: Genetic Mechanisms and Biomarker Pathways in a Prospective Cohort Study From the UK Biobank
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-14 DOI: 10.1111/jnc.70066
Hai-Hua Guo, Pei-Yang Gao, Wei Zhang, Yan Fu, Hao-Chen Chi, Zi-Hao Zhang, Shuang-Ling Han, Bao-Lin Han, Yu-Ying Zhang, Wei Xu, Lan Tan, Hui-Fu Wang

Population-based evidence directly linking liver diseases to brain disorders is limited, and its genetic and biochemical associations remain unclear. Our aim is to examine the links between liver diseases and brain disorders. This prospective cohort study utilized data from 492 059 participants in the UK Biobank. We identified 508 cases of alcoholic liver disease (ALD), 583 cases of non-alcoholic fatty liver disease (NAFLD), and 557 cases of viral hepatitis (VH) based on International Classification of Diseases (ICD) codes. Initially, we employed multiple linear and logistic regression to assess associations between liver diseases, polygenic risk score (PRS), inflammatory and metabolic biomarkers, and brain function. Cox proportional hazard models were then applied to determine the impact of liver diseases on the incidence of brain disorders. Ultimately, structural equation models were used to explore potential genetic and biomarker pathways. During a median follow-up of 14.46 years, participants with ALD, NAFLD, and VH demonstrated poorer cognition, mental health, and motor function compared to the healthy group, with p < 0.05 for false discovery rate (FDR-Q < 0.05). They exhibited increased risks for dementia (hazard ratios [HRs]: 2.28–4.10; FDR-Q < 0.001), major depressive disorder (HRs: 2.25–3.23; FDR-Q < 0.001), and generalized anxiety disorder (HRs: 1.70–2.66; FDR-Q < 0.01). Additionally, C-reactive protein, neutrophil-to-lymphocyte ratio, platelets, and low-density lipoprotein lipid components mediated the associations between PRS, liver diseases, and brain disorders. Our findings demonstrated that liver diseases were risk factors for brain disorders, with genetic and biochemical associations contributing to these risks.

{"title":"Liver Diseases and Brain Disorders: Genetic Mechanisms and Biomarker Pathways in a Prospective Cohort Study From the UK Biobank","authors":"Hai-Hua Guo,&nbsp;Pei-Yang Gao,&nbsp;Wei Zhang,&nbsp;Yan Fu,&nbsp;Hao-Chen Chi,&nbsp;Zi-Hao Zhang,&nbsp;Shuang-Ling Han,&nbsp;Bao-Lin Han,&nbsp;Yu-Ying Zhang,&nbsp;Wei Xu,&nbsp;Lan Tan,&nbsp;Hui-Fu Wang","doi":"10.1111/jnc.70066","DOIUrl":"https://doi.org/10.1111/jnc.70066","url":null,"abstract":"<div>\u0000 \u0000 <p>Population-based evidence directly linking liver diseases to brain disorders is limited, and its genetic and biochemical associations remain unclear. Our aim is to examine the links between liver diseases and brain disorders. This prospective cohort study utilized data from 492 059 participants in the UK Biobank. We identified 508 cases of alcoholic liver disease (ALD), 583 cases of non-alcoholic fatty liver disease (NAFLD), and 557 cases of viral hepatitis (VH) based on International Classification of Diseases (ICD) codes. Initially, we employed multiple linear and logistic regression to assess associations between liver diseases, polygenic risk score (PRS), inflammatory and metabolic biomarkers, and brain function. Cox proportional hazard models were then applied to determine the impact of liver diseases on the incidence of brain disorders. Ultimately, structural equation models were used to explore potential genetic and biomarker pathways. During a median follow-up of 14.46 years, participants with ALD, NAFLD, and VH demonstrated poorer cognition, mental health, and motor function compared to the healthy group, with <i>p</i> &lt; 0.05 for false discovery rate (FDR-Q &lt; 0.05). They exhibited increased risks for dementia (hazard ratios [HRs]: 2.28–4.10; FDR-Q &lt; 0.001), major depressive disorder (HRs: 2.25–3.23; FDR-Q &lt; 0.001), and generalized anxiety disorder (HRs: 1.70–2.66; FDR-Q &lt; 0.01). Additionally, C-reactive protein, neutrophil-to-lymphocyte ratio, platelets, and low-density lipoprotein lipid components mediated the associations between PRS, liver diseases, and brain disorders. Our findings demonstrated that liver diseases were risk factors for brain disorders, with genetic and biochemical associations contributing to these risks.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>\u0000 </div>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143831367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Donepezil Improves PSD95 Expression, Mitigates Neuroinflammation via PI3K/Akt/NF-κB and Mitochondrial Dysfunction in a Rodent Model of Subarachnoid Haemorrhage 在蛛网膜下腔出血的啮齿动物模型中,多奈哌齐通过 PI3K/Akt/NF-κB 和线粒体功能障碍改善 PSD95 表达,减轻神经炎症
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-14 DOI: 10.1111/jnc.70063
Ahmed Shaney Rehman, Ammar Tasleem, Nemat Ali, Rehan Khan, Mohd. Salman, Pravir Kumar, Suhel Parvez

Mitochondrial dysfunction is a known contributor to subarachnoid haemorrhage (SAH) induced early brain damage (EBI), leading to poor neurological outcomes. An experimental SAH model was induced in adult male Wistar rats using endovascular perforation. Donepezil, an acetylcholinesterase (AChE) inhibitor (1 or 2 mg/kg body weight), was administered intraperitoneally 4 h after SAH. The severity of cerebral cortex injury was assessed using blood clot grading, behavioral tests and H and E staining. We carried out an assessment of neuroinflammatory markers using western blotting and immunofluorescence. Additionally, we examined neuronal architecture using H and E staining, measured mitochondrial redox imbalance or ROS and membrane potential (Δѱm) and analyzed mitochondrial morphology using transmission electron microscopy (TEM). Apoptotic markers and mitochondrial respiratory complexes were assessed by western blotting. Our results indicated that donepezil treatment significantly upregulated PSD95, α7-AChR, CaMKII, BDNF, CREB, and PI3K expression in cerebral cortical neurons in response to SAH. This was accompanied by improved neurological function, reduced brain edema, decreased neuronal degeneration, and increased levels of OXPHOS and ATP. In the cerebral cortex, donepezil inhibited mitochondria-associated neuronal apoptosis after SAH as revealed by increased membrane potential integrity of mitochondria, reducing the ratio of Bax to Bcl-2 and inhibiting caspase-3 activity. Additionally, donepezil upregulated synaptic proteins (PSD95), strengthening synaptic connections and supporting spatial working memory circuits via the neurotrophic factor BDNF in post-SAH rats. Our research concludes that donepezil has neuroprotective benefits by inhibiting SAH-induced mitochondrial-mediated cell death through the regulation of Drp1-mediated mitochondrial morphology changes.

{"title":"Donepezil Improves PSD95 Expression, Mitigates Neuroinflammation via PI3K/Akt/NF-κB and Mitochondrial Dysfunction in a Rodent Model of Subarachnoid Haemorrhage","authors":"Ahmed Shaney Rehman,&nbsp;Ammar Tasleem,&nbsp;Nemat Ali,&nbsp;Rehan Khan,&nbsp;Mohd. Salman,&nbsp;Pravir Kumar,&nbsp;Suhel Parvez","doi":"10.1111/jnc.70063","DOIUrl":"https://doi.org/10.1111/jnc.70063","url":null,"abstract":"<div>\u0000 \u0000 <p>Mitochondrial dysfunction is a known contributor to subarachnoid haemorrhage (SAH) induced early brain damage (EBI), leading to poor neurological outcomes. An experimental SAH model was induced in adult male Wistar rats using endovascular perforation. Donepezil, an acetylcholinesterase (AChE) inhibitor (1 or 2 mg/kg body weight), was administered intraperitoneally 4 h after SAH. The severity of cerebral cortex injury was assessed using blood clot grading, behavioral tests and H and E staining. We carried out an assessment of neuroinflammatory markers using western blotting and immunofluorescence. Additionally, we examined neuronal architecture using H and E staining, measured mitochondrial redox imbalance or ROS and membrane potential (Δѱm) and analyzed mitochondrial morphology using transmission electron microscopy (TEM). Apoptotic markers and mitochondrial respiratory complexes were assessed by western blotting. Our results indicated that donepezil treatment significantly upregulated PSD95, α7-AChR, CaMKII, BDNF, CREB, and PI3K expression in cerebral cortical neurons in response to SAH. This was accompanied by improved neurological function, reduced brain edema, decreased neuronal degeneration, and increased levels of OXPHOS and ATP. In the cerebral cortex, donepezil inhibited mitochondria-associated neuronal apoptosis after SAH as revealed by increased membrane potential integrity of mitochondria, reducing the ratio of Bax to Bcl-2 and inhibiting caspase-3 activity. Additionally, donepezil upregulated synaptic proteins (PSD95), strengthening synaptic connections and supporting spatial working memory circuits via the neurotrophic factor BDNF in post-SAH rats. Our research concludes that donepezil has neuroprotective benefits by inhibiting SAH-induced mitochondrial-mediated cell death through the regulation of Drp1-mediated mitochondrial morphology changes.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>\u0000 </div>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143831366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
RETRACTION: Lithium Inhibits Aluminum-Induced Apoptosis in Rabbit Hippocampus, by Preventing Cytochrome c Translocation, Bcl-2 Decrease, Bax Elevation and Caspase-3 Activation
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-09 DOI: 10.1111/jnc.70065

RETRACTION: O. Ghribi, M. M. Herman, N. K. Spaulding, and J. Savory, “Lithium Inhibits Aluminum-Induced Apoptosis in Rabbit Hippocampus, by Preventing Cytochrome c Translocation, Bcl-2 Decrease, Bax Elevation and Caspase-3 Activation,” Journal of Neurochemistry 82, no. 1 (2002): 137-145, https://doi.org/10.1046/j.1471-4159.2002.00957.x.

The above article, published online on 25 June 2002 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the journal Editor-in-Chief, Andrew Lawrence; the International Society for Neurochemistry; and John Wiley & Sons Ltd. A third party notified the journal about concerns of image duplication in this article. Further investigation by the publisher and the journal confirmed that images in Figure 2b had been duplicated and rotated, bands in Figure 3 had been duplicated and rotated, an image had been duplicated between Figure 4b(i) and Figure 5a, an actin band had been duplicated between Figure 2a and Figure 3, and band in Figure 1 had been potentially duplicated and resized. Some authors could not be reached, and the remaining authors otherwise did not respond to inquiries by the journal and publisher. The retraction has been agreed to because the evidence of image duplication, rotation, and resizing fundamentally compromises the conclusions presented in the article and, due to the lack of original data, the conclusions cannot be verified. The authors did not respond to our notice regarding the retraction.

{"title":"RETRACTION: Lithium Inhibits Aluminum-Induced Apoptosis in Rabbit Hippocampus, by Preventing Cytochrome c Translocation, Bcl-2 Decrease, Bax Elevation and Caspase-3 Activation","authors":"","doi":"10.1111/jnc.70065","DOIUrl":"https://doi.org/10.1111/jnc.70065","url":null,"abstract":"<p><b>RETRACTION</b>: O. Ghribi, M. M. Herman, N. K. Spaulding, and J. Savory, “Lithium Inhibits Aluminum-Induced Apoptosis in Rabbit Hippocampus, by Preventing Cytochrome c Translocation, Bcl-2 Decrease, Bax Elevation and Caspase-3 Activation,” <i>Journal of Neurochemistry</i> 82, no. 1 (2002): 137-145, https://doi.org/10.1046/j.1471-4159.2002.00957.x.</p><p>The above article, published online on 25 June 2002 in Wiley Online Library (wileyonlinelibrary.com), has been retracted by agreement between the journal Editor-in-Chief, Andrew Lawrence; the International Society for Neurochemistry; and John Wiley &amp; Sons Ltd. A third party notified the journal about concerns of image duplication in this article. Further investigation by the publisher and the journal confirmed that images in Figure 2b had been duplicated and rotated, bands in Figure 3 had been duplicated and rotated, an image had been duplicated between Figure 4b(i) and Figure 5a, an actin band had been duplicated between Figure 2a and Figure 3, and band in Figure 1 had been potentially duplicated and resized. Some authors could not be reached, and the remaining authors otherwise did not respond to inquiries by the journal and publisher. The retraction has been agreed to because the evidence of image duplication, rotation, and resizing fundamentally compromises the conclusions presented in the article and, due to the lack of original data, the conclusions cannot be verified. The authors did not respond to our notice regarding the retraction.</p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70065","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143801824","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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 Duality of Astrocyte Neuromodulation: Astrocytes Sense Neuromodulators and Are Neuromodulators
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-07 DOI: 10.1111/jnc.70054
Justin Lines, Michelle Corkrum, Juan Aguilar, Alfonso Araque

Neuromodulation encompasses different processes that regulate neuronal and network function. Classical neuromodulators originating from long-range nuclei, such as acetylcholine, norepinephrine, or dopamine, act with a slower time course and wider spatial range than fast synaptic transmission and action potential firing. Accumulating evidence in vivo indicates that astrocytes, which are known to actively participate in synaptic function at tripartite synapses, are also involved in neuromodulatory processes. The present article reviews recent findings obtained in vivo indicating that astrocytes express receptors for neuromodulators that elevate their internal calcium and stimulate the release of gliotransmitters, which regulate synaptic and network function, and hence mediate, at least partially, the effects of neuromodulators. In addition, we propose that astrocytes act in local support of neuromodulators by spatially and temporally integrating neuronal and neuromodulatory signals to regulate neural network function. The presence of astrocyte-neuron hysteresis loops suggests astrocyte–neuron interaction at tripartite synapses scales up to astrocyte–neuronal networks that modulate neural network function. We finally propose that astrocytes sense the environmental conditions, including neuromodulators and network function states, and provide homeostatic control that maximizes the dynamic range of neural network activity. In summary, we propose that astrocytes are critical in mediating the effects of neuromodulators, and they also act as neuromodulators to provide neural network homeostasis thus optimizing information processing in the brain. Hence, astrocytes sense ongoing neuronal activity along with neuromodulators and, acting as neuromodulators, inform the neurons about the state of the internal system and the external world.

{"title":"The Duality of Astrocyte Neuromodulation: Astrocytes Sense Neuromodulators and Are Neuromodulators","authors":"Justin Lines,&nbsp;Michelle Corkrum,&nbsp;Juan Aguilar,&nbsp;Alfonso Araque","doi":"10.1111/jnc.70054","DOIUrl":"https://doi.org/10.1111/jnc.70054","url":null,"abstract":"<p>Neuromodulation encompasses different processes that regulate neuronal and network function. Classical neuromodulators originating from long-range nuclei, such as acetylcholine, norepinephrine, or dopamine, act with a slower time course and wider spatial range than fast synaptic transmission and action potential firing. Accumulating evidence in vivo indicates that astrocytes, which are known to actively participate in synaptic function at tripartite synapses, are also involved in neuromodulatory processes. The present article reviews recent findings obtained in vivo indicating that astrocytes express receptors for neuromodulators that elevate their internal calcium and stimulate the release of gliotransmitters, which regulate synaptic and network function, and hence mediate, at least partially, the effects of neuromodulators. In addition, we propose that astrocytes act in local support of neuromodulators by spatially and temporally integrating neuronal and neuromodulatory signals to regulate neural network function. The presence of astrocyte-neuron hysteresis loops suggests astrocyte–neuron interaction at tripartite synapses scales up to astrocyte–neuronal networks that modulate neural network function. We finally propose that astrocytes sense the environmental conditions, including neuromodulators and network function states, and provide homeostatic control that maximizes the dynamic range of neural network activity. In summary, we propose that astrocytes are critical in mediating the effects of neuromodulators, and they also act as neuromodulators to provide neural network homeostasis thus optimizing information processing in the brain. Hence, astrocytes sense ongoing neuronal activity along with neuromodulators and, acting as neuromodulators, inform the neurons about the state of the internal system and the external world.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70054","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143786778","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
α2δ-1-Linked NMDA and AMPA Receptors in Neuropathic Pain and Gabapentinoid Action
IF 4.2 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-07 DOI: 10.1111/jnc.70064
Yuying Huang, Shao-Rui Chen, Hui-Lin Pan

Chronic neuropathic pain is a debilitating condition that presents a significant therapeutic challenge. Unlike nociceptive pain, neuropathic pain is predominantly driven by glutamate NMDA receptors (NMDARs) and/or Ca2+-permeable AMPA receptors (CP-AMPARs) at synapses between primary afferent nerves and excitatory neurons in the spinal dorsal horn. The α2δ-1 protein, encoded by Cacna2d1 and historically recognized as a subunit of voltage-activated Ca2+ channels, is the primary target of gabapentinoids, such as gabapentin and pregabalin, which are widely prescribed for neuropathic pain and epilepsy. However, gabapentinoids have minimal effects on Ca2+ channel activity. Recent studies reveal that α2δ-1 plays a pivotal role in amplifying nociceptive input to the spinal cord in neuropathic pain. This action is mediated through its dynamic physical interactions with phosphorylated NMDARs and GluA1/GluA2 subunits via its intrinsically disordered C-terminal region. α2δ-1 not only promotes synaptic trafficking of NMDARs but also disrupts heteromeric assembly of GluA1/GluA2 subunits in the spinal dorsal horn. The central function of α2δ-1 is to elevate intracellular Ca2+ concentrations at both presynaptic and postsynaptic sites, augmenting nociceptive transmission. Consequently, α2δ-1 serves as a dual regulator coordinating synaptic expression of NMDARs and GluA1 homomeric CP-AMPARs, a function that underlies the therapeutic actions of gabapentinoids. By inhibiting α2δ-1, gabapentinoids reduce the hyperactivity of synaptic α2δ-1-bound NMDARs and CP-AMPARs, thereby dampening the excessive excitatory synaptic transmission characteristic of neuropathic pain. These newly identified roles of α2δ-1 in orchestrating glutamatergic synaptic plasticity suggest that gabapentinoids could be repurposed for treating other neurological disorders involving dysregulated synaptic NMDARs and CP-AMPARs.

慢性神经病理性疼痛使人衰弱,给治疗带来了巨大挑战。与痛觉痛不同,神经病理性疼痛主要由脊髓背角初级传入神经与兴奋性神经元之间突触上的谷氨酸 NMDA 受体(NMDARs)和/或钙离子渗透性 AMPA 受体(CP-AMPARs)驱动。α2δ-1蛋白由Cacna2d1编码,历来被认为是电压激活的Ca2+通道的亚基,是加巴喷丁和普瑞巴林等加巴喷丁类药物的主要靶点,这些药物被广泛用于治疗神经性疼痛和癫痫。然而,加巴喷丁类药物对 Ca2+ 通道活性的影响微乎其微。最近的研究表明,α2δ-1 在神经病理性疼痛中对脊髓痛觉输入的放大起着关键作用。α2δ-1通过其内在无序的C端区域与磷酸化的NMDARs和GluA1/GluA2亚基进行动态物理相互作用,从而发挥这种作用。α2δ-1不仅能促进NMDARs的突触贩运,还能破坏脊髓背角中GluA1/GluA2亚基的异构组装。α2δ-1的核心功能是提高突触前和突触后部位的细胞内Ca2+浓度,从而增强痛觉传导。因此,α2δ-1 是协调 NMDARs 和 GluA1 同源 CP-AMPARs 突触表达的双重调节器,这也是加巴喷丁类药物治疗作用的基础。通过抑制α2δ-1,加巴喷丁类药物可降低与突触α2δ-1结合的NMDARs和CP-AMPARs的过度活性,从而抑制神经病理性疼痛特有的过度兴奋性突触传递。这些新发现的α2δ-1在协调谷氨酸能突触可塑性方面的作用表明,加巴喷丁类药物可重新用于治疗涉及突触NMDARs和CP-AMPARs失调的其他神经系统疾病。
{"title":"α2δ-1-Linked NMDA and AMPA Receptors in Neuropathic Pain and Gabapentinoid Action","authors":"Yuying Huang,&nbsp;Shao-Rui Chen,&nbsp;Hui-Lin Pan","doi":"10.1111/jnc.70064","DOIUrl":"https://doi.org/10.1111/jnc.70064","url":null,"abstract":"<p>Chronic neuropathic pain is a debilitating condition that presents a significant therapeutic challenge. Unlike nociceptive pain, neuropathic pain is predominantly driven by glutamate NMDA receptors (NMDARs) and/or Ca<sup>2+</sup>-permeable AMPA receptors (CP-AMPARs) at synapses between primary afferent nerves and excitatory neurons in the spinal dorsal horn. The α2δ-1 protein, encoded by <i>Cacna2d1</i> and historically recognized as a subunit of voltage-activated Ca<sup>2+</sup> channels, is the primary target of gabapentinoids, such as gabapentin and pregabalin, which are widely prescribed for neuropathic pain and epilepsy. However, gabapentinoids have minimal effects on Ca<sup>2+</sup> channel activity. Recent studies reveal that α2δ-1 plays a pivotal role in amplifying nociceptive input to the spinal cord in neuropathic pain. This action is mediated through its dynamic physical interactions with phosphorylated NMDARs and GluA1/GluA2 subunits via its intrinsically disordered C-terminal region. α2δ-1 not only promotes synaptic trafficking of NMDARs but also disrupts heteromeric assembly of GluA1/GluA2 subunits in the spinal dorsal horn. The central function of α2δ-1 is to elevate intracellular Ca<sup>2+</sup> concentrations at both presynaptic and postsynaptic sites, augmenting nociceptive transmission. Consequently, α2δ-1 serves as a dual regulator coordinating synaptic expression of NMDARs and GluA1 homomeric CP-AMPARs, a function that underlies the therapeutic actions of gabapentinoids. By inhibiting α2δ-1, gabapentinoids reduce the hyperactivity of synaptic α2δ-1-bound NMDARs and CP-AMPARs, thereby dampening the excessive excitatory synaptic transmission characteristic of neuropathic pain. These newly identified roles of α2δ-1 in orchestrating glutamatergic synaptic plasticity suggest that gabapentinoids could be repurposed for treating other neurological disorders involving dysregulated synaptic NMDARs and CP-AMPARs.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":16527,"journal":{"name":"Journal of Neurochemistry","volume":"169 4","pages":""},"PeriodicalIF":4.2,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jnc.70064","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143787263","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Journal of Neurochemistry
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