首页 > 最新文献

ACS Chemical Neuroscience最新文献

英文 中文
O-GlcNAc Modification of α-Synuclein Can Alter Monomer Dynamics to Control Aggregation Kinetics. α-突触核蛋白的 O-GlcNAc 修饰可改变单体动力学,从而控制聚集动力学。
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 Epub Date: 2024-07-31 DOI: 10.1021/acschemneuro.4c00301
Kasun Gamage, Binyou Wang, Eldon R Hard, Thong Van, Ana Galesic, George R Phillips, Matthew Pratt, Lisa J Lapidus

The intrinsically disordered protein α-Synuclein is identified as a major toxic aggregate in Parkinson's as well as several other neurodegenerative diseases. Recent work on this protein has focused on the effects of posttranslational modifications on aggregation kinetics. Among them, O-GlcNAcylation of α-Synuclein has been observed to inhibit the aggregation propensity of the protein. Here, we investigate the monomer dynamics of two O-GlcNAcylated α-Synucleins, α-Syn(gT72), and α-Syn(gS87) and correlate them with the aggregation kinetics. We find that, compared to the unmodified protein, glycosylation at T72 makes the protein less compact and more diffusive, while glycosylation at S87 makes the protein more compact and less diffusive. Based on a model of the earliest steps in aggregation, we predict that T72 should aggregate slower than unmodified protein, which is confirmed by ThT fluorescence measurements. In contrast, S87 should aggregate faster, which is not mirrored in ThT kinetics of later fibril formation but does not rule out a higher rate of formation of small oligomers. Together, these results show that posttranslational modifications do not uniformly affect aggregation propensity.

内在紊乱蛋白α-突触核蛋白被确定为帕金森病和其他几种神经退行性疾病的主要毒性聚集体。有关这种蛋白质的最新研究集中于翻译后修饰对聚集动力学的影响。其中,α-突触核蛋白的 O-GlcNAcylation 被观察到可抑制该蛋白的聚集倾向。在这里,我们研究了两种 O-GlcNA 化的α-Synuclein(α-Syn(gT72)和α-Syn(gS87))的单体动力学,并将其与聚集动力学相关联。我们发现,与未修饰的蛋白质相比,T72 处的糖基化使蛋白质更不紧密,扩散性更强,而 S87 处的糖基化使蛋白质更紧密,扩散性更弱。根据聚合最早步骤的模型,我们预测 T72 的聚合速度应该比未修饰的蛋白质慢,ThT 荧光测量证实了这一点。与此相反,S87 的聚集速度应该更快,这并不反映在后来纤维形成的 ThT 动力学中,但也不排除小寡聚体形成速度更快的可能。这些结果共同表明,翻译后修饰对聚集倾向的影响并不一致。
{"title":"O-GlcNAc Modification of α-Synuclein Can Alter Monomer Dynamics to Control Aggregation Kinetics.","authors":"Kasun Gamage, Binyou Wang, Eldon R Hard, Thong Van, Ana Galesic, George R Phillips, Matthew Pratt, Lisa J Lapidus","doi":"10.1021/acschemneuro.4c00301","DOIUrl":"10.1021/acschemneuro.4c00301","url":null,"abstract":"<p><p>The intrinsically disordered protein α-Synuclein is identified as a major toxic aggregate in Parkinson's as well as several other neurodegenerative diseases. Recent work on this protein has focused on the effects of posttranslational modifications on aggregation kinetics. Among them, O-GlcNAcylation of α-Synuclein has been observed to inhibit the aggregation propensity of the protein. Here, we investigate the monomer dynamics of two O-GlcNAcylated α-Synucleins, α-Syn(gT72), and α-Syn(gS87) and correlate them with the aggregation kinetics. We find that, compared to the unmodified protein, glycosylation at T72 makes the protein less compact and more diffusive, while glycosylation at S87 makes the protein more compact and less diffusive. Based on a model of the earliest steps in aggregation, we predict that T72 should aggregate slower than unmodified protein, which is confirmed by ThT fluorescence measurements. In contrast, S87 should aggregate faster, which is not mirrored in ThT kinetics of later fibril formation but does not rule out a higher rate of formation of small oligomers. Together, these results show that posttranslational modifications do not uniformly affect aggregation propensity.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11342298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854059","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
Discovery and Profiling of New Multimodal Phenylglycinamide Derivatives as Potent Antiseizure and Antinociceptive Drug Candidates 作为强效抗癫痫和抗癫痫药物候选物的新型多模式苯基甘氨酰胺衍生物的发现和剖析
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 DOI: 10.1021/acschemneuro.4c0043810.1021/acschemneuro.4c00438
Marcin Jakubiec, Michał Abram, Mirosław Zagaja, Katarzyna Socała, Vanja Panic, Gniewomir Latacz, Szczepan Mogilski, Małgorzata Szafarz, Joanna Szala-Rycaj, Jerry Saunders, Peter J. West, Dorota Nieoczym, Katarzyna Przejczowska-Pomierny, Bartłomiej Szulczyk, Anna Krupa, Elżbieta Wyska, Piotr Wlaź, Cameron S. Metcalf, Karen Wilcox, Marta Andres-Mach, Rafał M. Kamiński and Krzysztof Kamiński*, 

We developed a focused series of original phenyl-glycinamide derivatives which showed potent activity across in vivo mouse seizure models, namely, maximal electroshock (MES) and 6 Hz (using both 32 and 44 mA current intensities) seizure models. Following intraperitoneal (i.p.) administration, compound (R)-32, which was identified as a lead molecule, demonstrated potent protection against all seizure models with ED50 values of 73.9 mg/kg (MES test), 18.8 mg/kg (6 Hz, 32 mA test), and 26.5 mg/kg (6 Hz, 44 mA test). Furthermore, (R)-32 demonstrated efficacy in both the PTZ-induced kindling paradigm and the ivPTZ seizure threshold test. The expression of neurotrophic factors, such as mature brain-derived neurotrophic factor (mBDNF) and nerve growth factor (NGF), in the hippocampus and/or cortex of mice, and the levels of glutamate and GABA were normalized after PTZ-induced kindling by (R)-32. Importantly, besides antiseizure activity, (R)-32 demonstrated potent antinociceptive efficacy in formalin-induced pain, capsaicin-induced pain, as well as oxaliplatin- and streptozotocin-induced peripheral neuropathy in mice (i.p.). No influence on muscular strength and body temperature in mice was observed. Pharmacokinetic studies and in vitro ADME-Tox data (i.e., high metabolic stability in human liver microsomes, a weak influence on CYPs, no hepatotoxicity, satisfactory passive transport, etc.) proved favorable drug-like properties of (R)-32. Thermal stability of (R)-32 shown in thermogravimetry and differential scanning calorimetry gives the opportunity to develop innovative oral solid dosage forms loaded with this compound. The in vitro binding and functional assays indicated its multimodal mechanism of action. (R)-32, beyond TRPV1 antagonism, inhibited calcium and sodium currents at a concentration of 10 μM. Therefore, the data obtained in the current studies justify a more detailed preclinical development of (R)-32 for epilepsy and pain indications.

我们开发了一系列具有针对性的苯基甘氨酰胺原始衍生物,这些衍生物在体内小鼠癫痫发作模型,即最大电击(MES)和 6 Hz(使用 32 和 44 mA 电流强度)癫痫发作模型中显示出强大的活性。腹腔给药后,被确定为先导分子的化合物(R)-32对所有癫痫发作模型都表现出有效的保护作用,其ED50值分别为73.9毫克/千克(MES试验)、18.8毫克/千克(6赫兹、32毫安试验)和26.5毫克/千克(6赫兹、44毫安试验)。此外,(R)-32在PTZ诱导的点燃范式和ivPTZ癫痫阈值试验中均表现出疗效。(R)-32能使小鼠海马和/或皮层中成熟脑源性神经营养因子(mBDNF)和神经生长因子(NGF)等神经营养因子的表达以及谷氨酸和GABA的水平在PTZ诱导的点燃后恢复正常。重要的是,除了抗癫痫活性外,(R)-32 还对福尔马林诱导的疼痛、辣椒素诱导的疼痛以及奥沙利铂和链脲佐菌素诱导的小鼠周围神经病变(静脉注射)具有强效的抗痛觉功效。对小鼠的肌肉力量和体温没有影响。药代动力学研究和体外 ADME-Tox 数据(即在人体肝脏微粒体中的高代谢稳定性、对 CYPs 的微弱影响、无肝毒性、令人满意的被动转运等)证明 (R)-32 具有良好的类药物特性。热重分析和差示扫描量热分析表明,(R)-32 具有热稳定性,这为开发含该化合物的创新口服固体制剂提供了机会。体外结合和功能测试表明了该化合物的多模式作用机制。除了 TRPV1 拮抗作用外,(R)-32 还能在 10 μM 浓度下抑制钙电流和钠电流。因此,目前研究获得的数据证明,(R)-32 有理由用于癫痫和疼痛适应症的更详细的临床前开发。
{"title":"Discovery and Profiling of New Multimodal Phenylglycinamide Derivatives as Potent Antiseizure and Antinociceptive Drug Candidates","authors":"Marcin Jakubiec,&nbsp;Michał Abram,&nbsp;Mirosław Zagaja,&nbsp;Katarzyna Socała,&nbsp;Vanja Panic,&nbsp;Gniewomir Latacz,&nbsp;Szczepan Mogilski,&nbsp;Małgorzata Szafarz,&nbsp;Joanna Szala-Rycaj,&nbsp;Jerry Saunders,&nbsp;Peter J. West,&nbsp;Dorota Nieoczym,&nbsp;Katarzyna Przejczowska-Pomierny,&nbsp;Bartłomiej Szulczyk,&nbsp;Anna Krupa,&nbsp;Elżbieta Wyska,&nbsp;Piotr Wlaź,&nbsp;Cameron S. Metcalf,&nbsp;Karen Wilcox,&nbsp;Marta Andres-Mach,&nbsp;Rafał M. Kamiński and Krzysztof Kamiński*,&nbsp;","doi":"10.1021/acschemneuro.4c0043810.1021/acschemneuro.4c00438","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00438https://doi.org/10.1021/acschemneuro.4c00438","url":null,"abstract":"<p >We developed a focused series of original phenyl-glycinamide derivatives which showed potent activity across <i>in vivo</i> mouse seizure models, namely, maximal electroshock (MES) and 6 Hz (using both 32 and 44 mA current intensities) seizure models. Following intraperitoneal (<i>i.p</i>.) administration, compound <b>(</b><b><i>R</i></b><b>)-32</b>, which was identified as a lead molecule, demonstrated potent protection against all seizure models with ED<sub>50</sub> values of 73.9 mg/kg (MES test), 18.8 mg/kg (6 Hz, 32 mA test), and 26.5 mg/kg (6 Hz, 44 mA test). Furthermore, <b>(</b><b><i>R</i></b><b>)-32</b> demonstrated efficacy in both the PTZ-induced kindling paradigm and the <i>iv</i>PTZ seizure threshold test. The expression of neurotrophic factors, such as mature brain-derived neurotrophic factor (mBDNF) and nerve growth factor (NGF), in the hippocampus and/or cortex of mice, and the levels of glutamate and GABA were normalized after PTZ-induced kindling by <b>(<i>R</i>)-32</b>. Importantly, besides antiseizure activity, (<b><i>R</i></b>)<b>-32</b> demonstrated potent antinociceptive efficacy in formalin-induced pain, capsaicin-induced pain, as well as oxaliplatin- and streptozotocin-induced peripheral neuropathy in mice (<i>i.p</i>.). No influence on muscular strength and body temperature in mice was observed. Pharmacokinetic studies and <i>in vitro</i> ADME-Tox data (<i>i.e.</i>, high metabolic stability in human liver microsomes, a weak influence on CYPs, no hepatotoxicity, satisfactory passive transport, <i>etc.</i>) proved favorable drug-like properties of (<b><i>R</i></b>)<b>-32</b>. Thermal stability of (<b><i>R</i></b>)-<b>32</b> shown in thermogravimetry and differential scanning calorimetry gives the opportunity to develop innovative oral solid dosage forms loaded with this compound. The <i>in vitro</i> binding and functional assays indicated its multimodal mechanism of action. (<b><i>R</i></b>)<b>-32</b>, beyond TRPV1 antagonism, inhibited calcium and sodium currents at a concentration of 10 μM. Therefore, the data obtained in the current studies justify a more detailed preclinical development of (<b><i>R</i></b>)<b>-32</b> for epilepsy and pain indications.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acschemneuro.4c00438","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142135323","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
Pharmacological Activities and Molecular Mechanisms of Sinapic Acid in Neurological Disorders. 西那皮酸在神经系统疾病中的药理活性和分子机制。
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 Epub Date: 2024-07-31 DOI: 10.1021/acschemneuro.4c00349
Mahan Farzan, Behnaz Abedi, Iman Bhia, Atossa Madanipour, Mahour Farzan, Mohammad Bhia, Ava Aghaei, Iman Kheirollahi, Mahzad Motallebi, Hossein Amini-Khoei, Yavuz Nuri Ertas

Sinapic acid (SA) is a phenylpropanoid derivative found in various natural sources that exhibits remarkable versatile properties, including antioxidant, anti-inflammatory, and metal-chelating capabilities, establishing itself as a promising candidate for the prevention and treatment of conditions affecting the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), ischemic stroke, and other neurological disorders. These effects also include neuroprotection in epilepsy models, as evidenced by a reduction in seizure-like behavior, cell death in specific hippocampal regions, and lowered neuroinflammatory markers. In AD, SA treatment enhances memory, reverses cognitive deficits, and attenuates astrocyte activation. SA also has positive effects on cognition by improving memory and lowering oxidative stress. This is shown by lower levels of oxidative stress markers, higher levels of antioxidant enzyme activity, and better memory retention. Additionally, in ischemic stroke and PD models, SA provides microglial protection and exerts anti-inflammatory effects. This review emphasizes SA's multifaceted neuroprotective properties and its potential role in the prevention and treatment of various brain disorders. Despite the need for further research to fully understand its mechanisms of action and clinical applicability, SA stands out as a valuable bioactive compound in the ongoing quest to combat neurodegenerative diseases and enhance the quality of life for affected individuals.

西那平酸(SA)是一种存在于各种天然资源中的苯丙类衍生物,具有显著的多功能特性,包括抗氧化、抗炎和金属螯合能力,是预防和治疗影响中枢神经系统疾病(如阿尔茨海默病(AD)、帕金森病(PD)、缺血性中风和其他神经系统疾病)的理想候选物质。这些作用还包括在癫痫模型中的神经保护作用,具体表现为减少癫痫发作样行为、特定海马区的细胞死亡以及降低神经炎症标志物。在注意力缺失症中,SA 治疗可增强记忆、逆转认知障碍并减轻星形胶质细胞的激活。通过改善记忆和降低氧化应激,SA 还能对认知产生积极影响。具体表现为氧化应激标志物水平降低、抗氧化酶活性提高以及记忆保持能力增强。此外,在缺血性中风和帕金森病模型中,SA 还能提供微神经胶质细胞保护并发挥抗炎作用。本综述强调了 SA 的多方面神经保护特性及其在预防和治疗各种脑部疾病中的潜在作用。尽管还需要进一步的研究来充分了解其作用机制和临床适用性,但在对抗神经退行性疾病和提高患者生活质量的不断探索中,SA 已成为一种有价值的生物活性化合物。
{"title":"Pharmacological Activities and Molecular Mechanisms of Sinapic Acid in Neurological Disorders.","authors":"Mahan Farzan, Behnaz Abedi, Iman Bhia, Atossa Madanipour, Mahour Farzan, Mohammad Bhia, Ava Aghaei, Iman Kheirollahi, Mahzad Motallebi, Hossein Amini-Khoei, Yavuz Nuri Ertas","doi":"10.1021/acschemneuro.4c00349","DOIUrl":"10.1021/acschemneuro.4c00349","url":null,"abstract":"<p><p>Sinapic acid (SA) is a phenylpropanoid derivative found in various natural sources that exhibits remarkable versatile properties, including antioxidant, anti-inflammatory, and metal-chelating capabilities, establishing itself as a promising candidate for the prevention and treatment of conditions affecting the central nervous system, such as Alzheimer's disease (AD), Parkinson's disease (PD), ischemic stroke, and other neurological disorders. These effects also include neuroprotection in epilepsy models, as evidenced by a reduction in seizure-like behavior, cell death in specific hippocampal regions, and lowered neuroinflammatory markers. In AD, SA treatment enhances memory, reverses cognitive deficits, and attenuates astrocyte activation. SA also has positive effects on cognition by improving memory and lowering oxidative stress. This is shown by lower levels of oxidative stress markers, higher levels of antioxidant enzyme activity, and better memory retention. Additionally, in ischemic stroke and PD models, SA provides microglial protection and exerts anti-inflammatory effects. This review emphasizes SA's multifaceted neuroprotective properties and its potential role in the prevention and treatment of various brain disorders. Despite the need for further research to fully understand its mechanisms of action and clinical applicability, SA stands out as a valuable bioactive compound in the ongoing quest to combat neurodegenerative diseases and enhance the quality of life for affected individuals.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854060","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
Healthy Plasma Exosomes Exert Potential Neuroprotective Effects against Methylmalonic Acid-Induced Hippocampal Neuron Injury. 健康血浆外泌体对甲基丙二酸诱导的海马神经元损伤具有潜在的神经保护作用
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 Epub Date: 2024-07-18 DOI: 10.1021/acschemneuro.4c00224
Wei Zhou, Huizhong Li, Jinxiu Song, Feng Suo, Maosheng Gu, Suhua Qi

Exosomes have shown good potential for alleviating neurological deficits and delaying memory deterioration, but the neuroprotective effects of exosomes remain unknown. Methylmalonic acidemia is a metabolic disorder characterized by the accumulation of methylmalonic acid (MMA) in various tissues that inhibits neuronal survival and function, leading to accelerated neurological deterioration. Effective therapies to mitigate these symptoms are lacking. The purpose of this study was to explore the neuroprotective effects of plasma exosomes on cells and a mouse model of MMA-induced injury. We evaluated the ability of plasma exosomes to reduce the neuronal apoptosis, cross the blood-brain barrier, and affect various parameters related to neuronal function. MMA promoted cell apoptosis, disrupted the metabolic balance, and altered the expression of B-cell lymphoma-2 (Bcl-2), Bcl2-associated X (Bax), and synaptophysin-1 (Syp-1), and these changes may be involved in MMA-induced neuronal apoptosis. Additionally, plasma exosomes normalized learning and memory and protected against MMA-induced neuronal apoptosis. Our findings indicate that neurological deficits are linked to the pathogenesis of methylmalonic acidemia, and healthy plasma exosomes may exert neuroprotective and therapeutic effects by altering the expression of exosomal microRNAs, facilitating neuronal functional recovery in the context of this inherited metabolic disease. Intravenous plasma-derived exosome treatment may be a novel clinical therapeutic strategy for methylmalonic acidemia.

外泌体在缓解神经功能缺损和延缓记忆衰退方面显示出良好的潜力,但外泌体的神经保护作用仍然未知。甲基丙二酸血症是一种代谢性疾病,其特点是甲基丙二酸(MMA)在各种组织中积累,抑制神经元的存活和功能,导致神经功能加速衰退。目前尚缺乏缓解这些症状的有效疗法。本研究的目的是探索血浆外泌体对细胞和 MMA 诱导损伤的小鼠模型的神经保护作用。我们评估了血浆外泌体减少神经元凋亡、穿过血脑屏障以及影响与神经元功能相关的各种参数的能力。MMA促进了细胞凋亡,破坏了代谢平衡,改变了B细胞淋巴瘤-2(Bcl-2)、Bcl2相关X(Bax)和突触素-1(Syp-1)的表达,这些变化可能与MMA诱导的神经元凋亡有关。此外,血浆外泌体还能使学习和记忆正常化,并防止 MMA 诱导的神经元凋亡。我们的研究结果表明,神经功能缺陷与甲基丙二酸血症的发病机制有关,而健康的血浆外泌体可能会通过改变外泌体microRNA的表达来发挥神经保护和治疗作用,从而促进这种遗传性代谢疾病的神经元功能恢复。静脉注射血浆外泌体治疗可能是治疗甲基丙二酸血症的一种新型临床治疗策略。
{"title":"Healthy Plasma Exosomes Exert Potential Neuroprotective Effects against Methylmalonic Acid-Induced Hippocampal Neuron Injury.","authors":"Wei Zhou, Huizhong Li, Jinxiu Song, Feng Suo, Maosheng Gu, Suhua Qi","doi":"10.1021/acschemneuro.4c00224","DOIUrl":"10.1021/acschemneuro.4c00224","url":null,"abstract":"<p><p>Exosomes have shown good potential for alleviating neurological deficits and delaying memory deterioration, but the neuroprotective effects of exosomes remain unknown. Methylmalonic acidemia is a metabolic disorder characterized by the accumulation of methylmalonic acid (MMA) in various tissues that inhibits neuronal survival and function, leading to accelerated neurological deterioration. Effective therapies to mitigate these symptoms are lacking. The purpose of this study was to explore the neuroprotective effects of plasma exosomes on cells and a mouse model of MMA-induced injury. We evaluated the ability of plasma exosomes to reduce the neuronal apoptosis, cross the blood-brain barrier, and affect various parameters related to neuronal function. MMA promoted cell apoptosis, disrupted the metabolic balance, and altered the expression of B-cell lymphoma-2 (Bcl-2), Bcl2-associated X (Bax), and synaptophysin-1 (Syp-1), and these changes may be involved in MMA-induced neuronal apoptosis. Additionally, plasma exosomes normalized learning and memory and protected against MMA-induced neuronal apoptosis. Our findings indicate that neurological deficits are linked to the pathogenesis of methylmalonic acidemia, and healthy plasma exosomes may exert neuroprotective and therapeutic effects by altering the expression of exosomal microRNAs, facilitating neuronal functional recovery in the context of this inherited metabolic disease. Intravenous plasma-derived exosome treatment may be a novel clinical therapeutic strategy for methylmalonic acidemia.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141722649","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
Exploring the Therapeutic Potential of Benfotiamine in a Sporadic Alzheimer's-Like Disease Rat Model: Insights into Insulin Signaling and Cognitive function. 探索苯磷硫胺在散发性阿尔茨海默病大鼠模型中的治疗潜力:洞察胰岛素信号转导和认知功能
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 Epub Date: 2024-07-15 DOI: 10.1021/acschemneuro.4c00113
Camila A E F Cardinali, Yandara A Martins, Ruan C M Moraes, Andressa P Costa, Mayke B Alencar, Ariel M Silber, Andrea S Torrão

Alzheimer's disease (AD) is a complex neurodegenerative process, also considered a metabolic condition due to alterations in glucose metabolism and insulin signaling pathways in the brain, which share similarities with diabetes. This study aimed to investigate the therapeutic effects of benfotiamine (BFT), a vitamin B1 analog, in the early stages of the neurodegenerative process in a sporadic model of Alzheimer's-like disease induced by intracerebroventricular injection of streptozotocin (STZ). Supplementation with 150 mg/kg of BFT for 7 days reversed the cognitive impairment in short- and long-term memories caused by STZ in rodents. We attribute these effects to BFT's ability to modulate glucose transporters type 1 and 3 (GLUT1 and GLUT3) in the hippocampus, inhibit GSK3 activity in the hippocampus, and modulate the insulin signaling in the hippocampus and entorhinal cortex, as well as reduce the activation of apoptotic pathways (BAX) in the hippocampus. Therefore, BFT emerges as a promising and accessible intervention in the initial treatment of conditions similar to AD.

阿尔茨海默病(AD)是一种复杂的神经退行性病变过程,由于大脑中葡萄糖代谢和胰岛素信号通路的改变,它也被认为是一种代谢性疾病,与糖尿病有相似之处。本研究旨在探讨维生素 B1 类似物苯磷硫胺(BFT)在通过脑室内注射链脲佐菌素(STZ)诱导的零星阿尔茨海默氏症样疾病模型的神经退行性过程早期阶段的治疗效果。连续 7 天补充 150 毫克/千克的 BFT 可逆转 STZ 对啮齿动物短期和长期记忆造成的认知障碍。我们将这些作用归因于 BFT 能够调节海马中的葡萄糖转运体 1 型和 3 型(GLUT1 和 GLUT3)、抑制海马中 GSK3 的活性、调节海马和内视网膜皮层中的胰岛素信号转导,以及减少海马中细胞凋亡通路(BAX)的激活。因此,BFT有望成为治疗与AD相似病症的初步干预措施。
{"title":"Exploring the Therapeutic Potential of Benfotiamine in a Sporadic Alzheimer's-Like Disease Rat Model: Insights into Insulin Signaling and Cognitive function.","authors":"Camila A E F Cardinali, Yandara A Martins, Ruan C M Moraes, Andressa P Costa, Mayke B Alencar, Ariel M Silber, Andrea S Torrão","doi":"10.1021/acschemneuro.4c00113","DOIUrl":"10.1021/acschemneuro.4c00113","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a complex neurodegenerative process, also considered a metabolic condition due to alterations in glucose metabolism and insulin signaling pathways in the brain, which share similarities with diabetes. This study aimed to investigate the therapeutic effects of benfotiamine (BFT), a vitamin B1 analog, in the early stages of the neurodegenerative process in a sporadic model of Alzheimer's-like disease induced by intracerebroventricular injection of streptozotocin (STZ). Supplementation with 150 mg/kg of BFT for 7 days reversed the cognitive impairment in short- and long-term memories caused by STZ in rodents. We attribute these effects to BFT's ability to modulate glucose transporters type 1 and 3 (GLUT1 and GLUT3) in the hippocampus, inhibit GSK3 activity in the hippocampus, and modulate the insulin signaling in the hippocampus and entorhinal cortex, as well as reduce the activation of apoptotic pathways (BAX) in the hippocampus. Therefore, BFT emerges as a promising and accessible intervention in the initial treatment of conditions similar to AD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11342302/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141615219","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
Psilocybin Facilitates Fear Extinction: Importance of Dose, Context, and Serotonin Receptors. 迷幻药促进恐惧消退:剂量、情境和羟色胺受体的重要性。
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 Epub Date: 2024-08-01 DOI: 10.1021/acschemneuro.4c00279
Samuel C Woodburn, Caleb M Levitt, Allison M Koester, Alex C Kwan

A variety of classic psychedelics and MDMA have been shown to enhance fear extinction in rodent models. This has translational significance because a standard treatment for post-traumatic stress disorder (PTSD) is prolonged exposure therapy. However, few studies have investigated psilocybin's potential effect on fear learning paradigms. More specifically, the extents to which dose, timing of administration, and serotonin receptors may influence psilocybin's effect on fear extinction are not understood. In this study, we used a delay fear conditioning paradigm to determine the effects of psilocybin on fear extinction, extinction retention, and fear renewal in male and female mice. Psilocybin robustly enhances fear extinction when given acutely prior to testing for all doses tested. Psilocybin also exerts long-term effects to elevate extinction retention and suppress fear renewal in a novel context, although these changes were sensitive to dose. Analysis of sex differences showed that females may respond to a narrower range of doses than males. Administration of psilocybin prior to fear learning or immediately after extinction yielded no change in behavior, indicating that concurrent extinction experience is necessary for the drug's effects. Cotreatment with a 5-HT2A receptor antagonist blocked psilocybin's effects for extinction, extinction retention, and fear renewal, whereas 5-HT1A receptor antagonism attenuated only the effect on fear renewal. Collectively, these results highlight dose, context, and serotonin receptors as crucial factors in psilocybin's ability to facilitate fear extinction. The study provides preclinical evidence to support investigating psilocybin as a pharmacological adjunct for extinction-based therapy for PTSD.

研究表明,多种经典迷幻药和亚甲二氧基甲基安非他明(MDMA)可增强啮齿动物模型的恐惧消退能力。这具有转化意义,因为创伤后应激障碍(PTSD)的标准治疗方法是长时间暴露疗法。然而,很少有研究调查了迷幻药对恐惧学习范式的潜在影响。更具体地说,剂量、给药时间和血清素受体可能会在多大程度上影响迷幻药对恐惧消退的作用,目前尚不清楚。在这项研究中,我们使用延迟恐惧条件反射范例来确定银环蛇毒素对雌雄小鼠恐惧消退、消退保持和恐惧更新的影响。在所有测试剂量的小鼠中,如果在测试前急性给予迷幻药,都能显著增强其恐惧消退能力。在新的环境中,迷幻药还能产生长期效应,提高熄灭保持率和抑制恐惧更新,尽管这些变化对剂量很敏感。对性别差异的分析表明,与男性相比,女性可能对较窄的剂量范围做出反应。在恐惧学习之前或消退之后立即服用迷幻药不会导致行为改变,这表明同时消退经历是药物作用的必要条件。与 5-HT2A 受体拮抗剂共用可阻断西洛滨对消退、消退保持和恐惧更新的影响,而 5-HT1A 受体拮抗剂只减弱对恐惧更新的影响。总之,这些结果突出表明,剂量、情境和血清素受体是影响迷幻药促进恐惧消退能力的关键因素。这项研究提供了临床前证据,支持将迷幻药作为一种药理辅助疗法,用于创伤后应激障碍的消退治疗。
{"title":"Psilocybin Facilitates Fear Extinction: Importance of Dose, Context, and Serotonin Receptors.","authors":"Samuel C Woodburn, Caleb M Levitt, Allison M Koester, Alex C Kwan","doi":"10.1021/acschemneuro.4c00279","DOIUrl":"10.1021/acschemneuro.4c00279","url":null,"abstract":"<p><p>A variety of classic psychedelics and MDMA have been shown to enhance fear extinction in rodent models. This has translational significance because a standard treatment for post-traumatic stress disorder (PTSD) is prolonged exposure therapy. However, few studies have investigated psilocybin's potential effect on fear learning paradigms. More specifically, the extents to which dose, timing of administration, and serotonin receptors may influence psilocybin's effect on fear extinction are not understood. In this study, we used a delay fear conditioning paradigm to determine the effects of psilocybin on fear extinction, extinction retention, and fear renewal in male and female mice. Psilocybin robustly enhances fear extinction when given acutely prior to testing for all doses tested. Psilocybin also exerts long-term effects to elevate extinction retention and suppress fear renewal in a novel context, although these changes were sensitive to dose. Analysis of sex differences showed that females may respond to a narrower range of doses than males. Administration of psilocybin prior to fear learning or immediately after extinction yielded no change in behavior, indicating that concurrent extinction experience is necessary for the drug's effects. Cotreatment with a 5-HT<sub>2A</sub> receptor antagonist blocked psilocybin's effects for extinction, extinction retention, and fear renewal, whereas 5-HT<sub>1A</sub> receptor antagonism attenuated only the effect on fear renewal. Collectively, these results highlight dose, context, and serotonin receptors as crucial factors in psilocybin's ability to facilitate fear extinction. The study provides preclinical evidence to support investigating psilocybin as a pharmacological adjunct for extinction-based therapy for PTSD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141858192","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
Neuromicrobiology Comes of Age: The Multifaceted Interactions between the Microbiome and the Nervous System. 神经微生物学时代的到来:微生物组与神经系统之间的多方面相互作用》(Neuromicrobiology Comes of Age: The Multifaceted Interactions between the Microbiome and the Nervous System.
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-21 Epub Date: 2024-08-05 DOI: 10.1021/acschemneuro.4c00307
Ashwarya S Devason, Christoph A Thaiss, Cesar de la Fuente-Nunez

The past decade has seen an explosion in our knowledge about the interactions between gut microbiota, the central nervous system, and the immune system. The gut-brain axis has recently gained much attention due to its role in regulating host physiology. This review explores recent findings concerning potential pathways linking the gut-brain axis to the initiation, pathophysiology, and development of neurological disorders. Our objective of this work is to uncover causative factors and pinpoint particular pathways and therapeutic targets that may facilitate the translation of experimental animal research into practical applications for human patients. We highlight three distinct yet interrelated mechanisms: (1) disruptions of both the intestinal and blood-brain barriers, (2) persistent neuroinflammation, and (3) the role of the vagus nerve.

过去十年间,我们对肠道微生物群、中枢神经系统和免疫系统之间相互作用的了解激增。最近,肠道-大脑轴因其在调节宿主生理方面的作用而备受关注。本综述探讨了有关肠道-大脑轴与神经系统疾病的发生、病理生理学和发展之间潜在联系的最新发现。我们这项工作的目的是揭示致病因素,并指出特定的途径和治疗靶点,以促进将动物实验研究转化为对人类患者的实际应用。我们强调了三种不同但又相互关联的机制:(1) 肠屏障和血脑屏障的破坏,(2) 持续的神经炎症,以及 (3) 迷走神经的作用。
{"title":"Neuromicrobiology Comes of Age: The Multifaceted Interactions between the Microbiome and the Nervous System.","authors":"Ashwarya S Devason, Christoph A Thaiss, Cesar de la Fuente-Nunez","doi":"10.1021/acschemneuro.4c00307","DOIUrl":"10.1021/acschemneuro.4c00307","url":null,"abstract":"<p><p>The past decade has seen an explosion in our knowledge about the interactions between gut microbiota, the central nervous system, and the immune system. The gut-brain axis has recently gained much attention due to its role in regulating host physiology. This review explores recent findings concerning potential pathways linking the gut-brain axis to the initiation, pathophysiology, and development of neurological disorders. Our objective of this work is to uncover causative factors and pinpoint particular pathways and therapeutic targets that may facilitate the translation of experimental animal research into practical applications for human patients. We highlight three distinct yet interrelated mechanisms: (1) disruptions of both the intestinal and blood-brain barriers, (2) persistent neuroinflammation, and (3) the role of the vagus nerve.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892231","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
Neural Development and Repair Induced by Femtosecond Laser Stimulation 飞秒激光刺激诱导的神经发育与修复
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-20 DOI: 10.1021/acschemneuro.4c0031010.1021/acschemneuro.4c00310
Fan Qi*, Hao He and Yujie Zhu*, 

Dendritic spines function as postsynaptic sites, receiving excitatory signals from presynaptic axons. The synaptic plasticity of spines underlies the refinement of neuronal circuits. Neural cognitive disorders are commonly associated with the impairment and elimination of dendritic spines. In this study, we report an all-optical method to activate dendritic spine growth and regeneration by a single short flash of femtosecond laser stimulation. The inhibited development and loss of spines can be rescued by a transient illumination of the laser inside a micrometer region of the soma by activating the extracellular signal-regulated kinase (ERK) signaling pathway. The rescued neurons exhibit function. Hence we provide a potential noninvasive method for the regeneration of dendritic spines.

树突棘作为突触后部位,接收来自突触前轴突的兴奋信号。棘突的突触可塑性是完善神经元回路的基础。神经认知障碍通常与树突棘受损和消失有关。在这项研究中,我们报告了一种全光学方法,通过飞秒激光的单次短闪刺激来激活树突棘的生长和再生。通过激活细胞外信号调节激酶(ERK)信号通路,瞬时照射神经元基质微米区域内的激光可拯救被抑制的树突棘发育和丧失。获救的神经元会表现出功能。因此,我们为树突棘的再生提供了一种潜在的非侵入性方法。
{"title":"Neural Development and Repair Induced by Femtosecond Laser Stimulation","authors":"Fan Qi*,&nbsp;Hao He and Yujie Zhu*,&nbsp;","doi":"10.1021/acschemneuro.4c0031010.1021/acschemneuro.4c00310","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00310https://doi.org/10.1021/acschemneuro.4c00310","url":null,"abstract":"<p >Dendritic spines function as postsynaptic sites, receiving excitatory signals from presynaptic axons. The synaptic plasticity of spines underlies the refinement of neuronal circuits. Neural cognitive disorders are commonly associated with the impairment and elimination of dendritic spines. In this study, we report an all-optical method to activate dendritic spine growth and regeneration by a single short flash of femtosecond laser stimulation. The inhibited development and loss of spines can be rescued by a transient illumination of the laser inside a micrometer region of the soma by activating the extracellular signal-regulated kinase (ERK) signaling pathway. The rescued neurons exhibit function. Hence we provide a potential noninvasive method for the regeneration of dendritic spines.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142135533","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
Exploring Novel GSK-3β Inhibitors for Anti-Neuroinflammatory and Neuroprotective Effects: Synthesis, Crystallography, Computational Analysis, and Biological Evaluation 探索新型 GSK-3β 抑制剂的抗神经炎症和神经保护作用:合成、晶体学、计算分析和生物学评估
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-19 DOI: 10.1021/acschemneuro.4c0036510.1021/acschemneuro.4c00365
Izabella Góral, Tomasz Wichur, Emilia Sługocka, Przemysław Grygier, Monika Głuch-Lutwin, Barbara Mordyl, Ewelina Honkisz-Orzechowska, Natalia Szałaj, Justyna Godyń, Dawid Panek, Paula Zaręba, Anna Sarka, Paweł Żmudzki, Gniewomir Latacz, Katarzyna Pustelny, Adam Bucki, Anna Czarna, Filipe Menezes* and Anna Więckowska*, 

In the pathogenesis of Alzheimer’s disease, the overexpression of glycogen synthase kinase-3β (GSK-3β) stands out due to its multifaced nature, as it contributes to the promotion of amyloid β and tau protein accumulation, as well as neuroinflammatory processes. Therefore, in the present study, we have designed, synthesized, and evaluated a new series of GSK-3β inhibitors based on the N-(pyridin-2-yl)cyclopropanecarboxamide scaffold. We identified compound 36, demonstrating an IC50 of 70 nM against GSK-3β. Subsequently, through crystallography studies and quantum mechanical analysis, we elucidated its binding mode and identified the structural features crucial for interactions with the active site of GSK-3β, thereby understanding its inhibitory potency. Compound 36 was effective in the cellular model of hyperphosphorylated tau-induced neurodegeneration, where it restored cell viability after okadaic acid treatment and showed anti-inflammatory activity in the LPS model, significantly reducing NO, IL-6, and TNF-α release. In ADME-tox in vitro studies, we confirmed the beneficial profile of 36, including high permeability in PAMPA (Pe equals 9.4) and high metabolic stability in HLMs as well as lack of significant interactions with isoforms of the CYP enzymes and lack of considerable cytotoxicity on selected cell lines (IC50 > 100 μM on HT-22 cells and 89.3 μM on BV-2 cells). Based on promising pharmacological activities and favorable ADME-tox properties, compound 36 may be considered a promising candidate for in vivo research as well as constitute a reliable starting point for further studies.

在阿尔茨海默病的发病机制中,糖原合酶激酶-3β(GSK-3β)的过度表达因其多面性而引人注目,因为它有助于促进淀粉样β和tau蛋白的积累以及神经炎症过程。因此,在本研究中,我们基于 N-(吡啶-2-基)环丙烷甲酰胺支架设计、合成并评估了一系列新的 GSK-3β 抑制剂。我们发现了化合物 36,其对 GSK-3β 的 IC50 值为 70 nM。随后,通过晶体学研究和量子力学分析,我们阐明了其结合模式,确定了与 GSK-3β 活性位点相互作用的关键结构特征,从而了解了其抑制效力。化合物 36 在高磷酸化 tau 诱导的神经退行性病变的细胞模型中很有效,它能恢复 okadaic 酸处理后的细胞活力,并在 LPS 模型中显示出抗炎活性,能显著减少 NO、IL-6 和 TNF-α 的释放。在 ADME-tox 体外研究中,我们证实了 36 的有益特性,包括在 PAMPA 中的高渗透性(Pe 等于 9.4)和在 HLMs 中的高代谢稳定性,以及与 CYP 酶同工酶无明显相互作用和对选定细胞株无明显细胞毒性(IC50 > 对 HT-22 细胞为 100 μM,对 BV-2 细胞为 89.3 μM)。由于化合物 36 具有良好的药理活性和良好的 ADME 毒性,因此可将其作为体内研究的候选化合物,并作为进一步研究的可靠起点。
{"title":"Exploring Novel GSK-3β Inhibitors for Anti-Neuroinflammatory and Neuroprotective Effects: Synthesis, Crystallography, Computational Analysis, and Biological Evaluation","authors":"Izabella Góral,&nbsp;Tomasz Wichur,&nbsp;Emilia Sługocka,&nbsp;Przemysław Grygier,&nbsp;Monika Głuch-Lutwin,&nbsp;Barbara Mordyl,&nbsp;Ewelina Honkisz-Orzechowska,&nbsp;Natalia Szałaj,&nbsp;Justyna Godyń,&nbsp;Dawid Panek,&nbsp;Paula Zaręba,&nbsp;Anna Sarka,&nbsp;Paweł Żmudzki,&nbsp;Gniewomir Latacz,&nbsp;Katarzyna Pustelny,&nbsp;Adam Bucki,&nbsp;Anna Czarna,&nbsp;Filipe Menezes* and Anna Więckowska*,&nbsp;","doi":"10.1021/acschemneuro.4c0036510.1021/acschemneuro.4c00365","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00365https://doi.org/10.1021/acschemneuro.4c00365","url":null,"abstract":"<p >In the pathogenesis of Alzheimer’s disease, the overexpression of glycogen synthase kinase-3β (GSK-3β) stands out due to its multifaced nature, as it contributes to the promotion of amyloid β and tau protein accumulation, as well as neuroinflammatory processes. Therefore, in the present study, we have designed, synthesized, and evaluated a new series of GSK-3β inhibitors based on the <i>N</i>-(pyridin-2-yl)cyclopropanecarboxamide scaffold. We identified compound <b>36</b>, demonstrating an IC<sub>50</sub> of 70 nM against GSK-3β. Subsequently, through crystallography studies and quantum mechanical analysis, we elucidated its binding mode and identified the structural features crucial for interactions with the active site of GSK-3β, thereby understanding its inhibitory potency. Compound <b>36</b> was effective in the cellular model of hyperphosphorylated tau-induced neurodegeneration, where it restored cell viability after okadaic acid treatment and showed anti-inflammatory activity in the LPS model, significantly reducing NO, IL-6, and TNF-α release. In ADME-tox in vitro studies, we confirmed the beneficial profile of <b>36,</b> including high permeability in PAMPA (<i>Pe</i> equals 9.4) and high metabolic stability in HLMs as well as lack of significant interactions with isoforms of the CYP enzymes and lack of considerable cytotoxicity on selected cell lines (IC<sub>50</sub> &gt; 100 μM on HT-22 cells and 89.3 μM on BV-2 cells). Based on promising pharmacological activities and favorable ADME-tox properties, compound <b>36</b> may be considered a promising candidate for in vivo research as well as constitute a reliable starting point for further studies.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/epdf/10.1021/acschemneuro.4c00365","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142135221","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
Structural Reorganization Mechanism of the Aβ42 Fibril Mediated by N-Substituted Oligopyrrolamide ADH-353 N-取代低聚吡咯酰胺 ADH-353 介导的 Aβ42 纤维的结构重组机制
IF 4.1 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-19 DOI: 10.1021/acschemneuro.4c0025310.1021/acschemneuro.4c00253
Arushi Dabas,  and , Bhupesh Goyal*, 

The inhibition of amyloid-β (Aβ) fibrillation and clearance of Aβ aggregates have emerged as a potential pharmacological strategy to alleviate Aβ aggregate-induced neurotoxicity in Alzheimer’s disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged N-substituted oligopyrrolamides for its notable ability to inhibit Aβ fibrillation, disintegrate intracellular cytotoxic Aβ oligomers, and alleviate Aβ-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aβ42 fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aβ42 fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of β-strands and the emergence of a helix conformation, indicating a loss of the well-organized β-sheet-rich structure of the disease-relevant Aβ42 fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aβ42 fibril (ΔGbinding= −142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged N-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aβ42 fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aβ peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aβ42 fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aβ42 fibril. The illumination of key interactions responsible for the destabilization of the Aβ42 fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aβ aggregates in AD.

抑制淀粉样蛋白-β(Aβ)纤颤和清除 Aβ 聚集体已成为减轻 Aβ 聚集体诱发的阿尔茨海默病(AD)神经毒性的一种潜在药理策略。Maity等人从一个小型的带正电荷的N-取代寡吡咯酰胺类药物库中筛选出了ADH-353,因为它具有抑制Aβ纤维化、分解细胞内细胞毒性Aβ寡聚体以及减轻Aβ诱导的SH-SY5Y和N2a细胞细胞毒性的显著能力。然而,ADH-353 与 Aβ42 纤维相互作用并导致其破坏和随后清除的分子机制仍不清楚。因此,本研究利用分子动力学模拟揭示了 ADH-353 破坏神经毒性 Aβ42 纤维(PDB ID 2NAO)的详细分子机制。有趣的是,模拟过程中的构象快照描绘了 β 链的缩短和消失以及螺旋构象的出现,这表明与疾病相关的 Aβ42 纤维在加入 ADH-353 后失去了组织良好的富含 β 片的结构。ADH-353与Aβ42纤维紧密结合(ΔGbinding= -142.91 ± 1.61 kcal/mol),ADH-353带正电荷的N-丙胺侧链与Aβ42纤维的谷氨酸(Glu3、Glu11和Glu22)和天冬氨酸(Asp7和Asp23)残基之间的静电相互作用起了显著作用。这与异核单量子相干核磁共振研究结果非常吻合,该研究表明 ADH-353 与 Aβ 肽的结合是由静电和疏水接触驱动的。此外,加入 ADH-353 后,Aβ42 纤维链的结合亲和力显著下降,这表明链间相互作用减弱,导致 Aβ42 纤维的双马蹄形构象被破坏。这项研究揭示了ADH-353破坏Aβ42纤维稳定性的关键相互作用,这将大大有助于设计新的化学支架,提高清除AD中Aβ聚集体的功效。
{"title":"Structural Reorganization Mechanism of the Aβ42 Fibril Mediated by N-Substituted Oligopyrrolamide ADH-353","authors":"Arushi Dabas,&nbsp; and ,&nbsp;Bhupesh Goyal*,&nbsp;","doi":"10.1021/acschemneuro.4c0025310.1021/acschemneuro.4c00253","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00253https://doi.org/10.1021/acschemneuro.4c00253","url":null,"abstract":"<p >The inhibition of amyloid-β (Aβ) fibrillation and clearance of Aβ aggregates have emerged as a potential pharmacological strategy to alleviate Aβ aggregate-induced neurotoxicity in Alzheimer’s disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged <i>N</i>-substituted oligopyrrolamides for its notable ability to inhibit Aβ fibrillation, disintegrate intracellular cytotoxic Aβ oligomers, and alleviate Aβ-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aβ<sub>42</sub> fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aβ<sub>42</sub> fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of β-strands and the emergence of a helix conformation, indicating a loss of the well-organized β-sheet-rich structure of the disease-relevant Aβ<sub>42</sub> fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aβ<sub>42</sub> fibril (Δ<i>G</i><sub>binding</sub>= −142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged <i>N</i>-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aβ<sub>42</sub> fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aβ peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aβ<sub>42</sub> fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aβ<sub>42</sub> fibril. The illumination of key interactions responsible for the destabilization of the Aβ<sub>42</sub> fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aβ aggregates in AD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142135140","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
期刊
ACS Chemical Neuroscience
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