Pub Date : 2024-11-06Epub Date: 2024-10-29DOI: 10.1021/acschemneuro.4c00501
Mikhail Matveyenka, Abid Ali, Charles L Mitchell, Harris C Brown, Dmitry Kurouski
A hallmark of Parkinson disease (PD) is a progressive degeneration of neurons in the substantia nigra pars compacta, hypothalamus, and thalamus. Although the exact etiology of irreversible neuronal degeneration is unclear, a growing body of experimental evidence indicates that PD could be triggered by the abrupt aggregation of α-synuclein (α-Syn), a small membrane protein that is responsible for cell vesicle trafficking. Phospholipids uniquely alter the rate of α-Syn aggregation and, consequently, change the cytotoxicity of α-Syn oligomers and fibrils. However, the role of cholesterol in the aggregation of α-Syn remains unclear. In this study, we used Caenorhabditis elegans that overexpressed α-Syn to investigate the effect of low (15%), normal (30%), and high (60%) concentrations of cholesterol on α-Syn aggregation. We found that an increase in the concentration of cholesterol in diets substantially shortened the lifespan of C. elegans. Using biophysical methods, we also investigated the extent to which large unilamellar vesicles (LUVs) with low, normal, and high concentrations of cholesterol altered the rate of α-Syn aggregation. We found that only lipid membranes with a 60% concentration of cholesterol substantially accelerated the rate of protein aggregation. Cell assays revealed that α-Syn fibrils formed in the presence of LUVs with different concentrations of cholesterol exerted very similar levels of cytotoxicity to rat dopaminergic neurons. These results suggest that changes in the concentration of cholesterol in the plasma membrane, which in turn could be caused by nutritional preferences, could accelerate the onset and progression of PD.
{"title":"Cholesterol Accelerates Aggregation of α-Synuclein Simultaneously Increasing the Toxicity of Amyloid Fibrils.","authors":"Mikhail Matveyenka, Abid Ali, Charles L Mitchell, Harris C Brown, Dmitry Kurouski","doi":"10.1021/acschemneuro.4c00501","DOIUrl":"10.1021/acschemneuro.4c00501","url":null,"abstract":"<p><p>A hallmark of Parkinson disease (PD) is a progressive degeneration of neurons in the substantia nigra pars compacta, hypothalamus, and thalamus. Although the exact etiology of irreversible neuronal degeneration is unclear, a growing body of experimental evidence indicates that PD could be triggered by the abrupt aggregation of α-synuclein (α-Syn), a small membrane protein that is responsible for cell vesicle trafficking. Phospholipids uniquely alter the rate of α-Syn aggregation and, consequently, change the cytotoxicity of α-Syn oligomers and fibrils. However, the role of cholesterol in the aggregation of α-Syn remains unclear. In this study, we used <i>Caenorhabditis elegans</i> that overexpressed α-Syn to investigate the effect of low (15%), normal (30%), and high (60%) concentrations of cholesterol on α-Syn aggregation. We found that an increase in the concentration of cholesterol in diets substantially shortened the lifespan of <i>C. elegans</i>. Using biophysical methods, we also investigated the extent to which large unilamellar vesicles (LUVs) with low, normal, and high concentrations of cholesterol altered the rate of α-Syn aggregation. We found that only lipid membranes with a 60% concentration of cholesterol substantially accelerated the rate of protein aggregation. Cell assays revealed that α-Syn fibrils formed in the presence of LUVs with different concentrations of cholesterol exerted very similar levels of cytotoxicity to rat dopaminergic neurons. These results suggest that changes in the concentration of cholesterol in the plasma membrane, which in turn could be caused by nutritional preferences, could accelerate the onset and progression of PD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"4075-4081"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142520296","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}
Pub Date : 2024-11-06Epub Date: 2024-10-09DOI: 10.1021/acschemneuro.4c00519
Yu Nishio, Craig W Lindsley, Aaron M Bender
Tianeptine (1) is an unusual antidepressant in that its mechanism of action appears to be independent from any activity at serotonin receptors or monoamine transporters. In fact, tianeptine has been shown to be a moderately potent agonist for the mu opioid receptor (MOR) and to a lesser extent the delta opioid receptor (DOR). Additionally, tianeptine's efficacy may be related to its action on glutamate-mediated pathways of neuroplasticity. Regardless of which neurotransmitter system is primarily responsible for the observed efficacy, the MOR agonist activity is problematic with respect to abuse liability. Increasing numbers of case reports have demonstrated that tianeptine is indeed being used recreationally at doses far beyond what are considered therapeutically relevant or safe, and scheduling reclassifications or outright bans on tianeptine products are ongoing around the world. It is the aim of this review to discuss the medicinal chemistry and pharmacology of tianeptine and to summarize this intriguing discrepancy between tianeptine's historical use as a safe and effective antidepressant and its emerging potential for abuse.
{"title":"Classics in Chemical Neuroscience: Tianeptine.","authors":"Yu Nishio, Craig W Lindsley, Aaron M Bender","doi":"10.1021/acschemneuro.4c00519","DOIUrl":"10.1021/acschemneuro.4c00519","url":null,"abstract":"<p><p>Tianeptine (<b>1</b>) is an unusual antidepressant in that its mechanism of action appears to be independent from any activity at serotonin receptors or monoamine transporters. In fact, tianeptine has been shown to be a moderately potent agonist for the mu opioid receptor (MOR) and to a lesser extent the delta opioid receptor (DOR). Additionally, tianeptine's efficacy may be related to its action on glutamate-mediated pathways of neuroplasticity. Regardless of which neurotransmitter system is primarily responsible for the observed efficacy, the MOR agonist activity is problematic with respect to abuse liability. Increasing numbers of case reports have demonstrated that tianeptine is indeed being used recreationally at doses far beyond what are considered therapeutically relevant or safe, and scheduling reclassifications or outright bans on tianeptine products are ongoing around the world. It is the aim of this review to discuss the medicinal chemistry and pharmacology of tianeptine and to summarize this intriguing discrepancy between tianeptine's historical use as a safe and effective antidepressant and its emerging potential for abuse.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"3863-3873"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386349","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}
Pub Date : 2024-11-06Epub Date: 2024-10-17DOI: 10.1021/acschemneuro.4c00561
Georgia K Williams, Jordy Akkermans, Matt Lawson, Patryk Syta, Steven Staelens, Mohit H Adhikari, A Jennifer Morton, Björn Nitzsche, Johannes Boltze, Chris Christou, Daniele Bertoglio, Muneer Ahamed
Huntington's disease (HD) is a neurodegenerative disease that causes cognitive, movement, behavioral, and sleep disturbances, which over time result in progressive disability and eventually death. Clinical translation of novel therapeutics and imaging probes could be accelerated by additional testing in well-characterized large animal models of HD. The major goal of our preliminary cross-sectional study is to demonstrate the feasibility and utility of the unique transgenic sheep model of HD (OVT73) in positron emission tomography (PET) imaging. PET imaging studies were performed in healthy merino sheep (6 year old, n = 3) and OVT73 HD sheep (5.5 year old, n = 3, and 11 year old, n = 3). Region-of-interest and brain atlas labels were defined for regional analyses by using a sheep brain template. [18F]fluorodeoxyglucose ([18F]FDG) was employed to compare the regional brain glucose metabolism and variations in FDG uptake between control and HD sheep. We also used [18F]fluoro-3,4-dihydroxyphenylalanine ([18F]FDOPA) to compare the extent of striatal dysfunction and evaluated the binding potential (BPND) in key brain regions between the groups. Compared with healthy controls and 11 year old HD sheep, the 5.5 year old HD sheep exhibited significantly increased [18F]FDG uptake in several cortical and subcortical brain regions (P < 0.05-0.01). No difference in [18F]FDG uptake was observed between healthy controls and 11 year old HD sheep. Analysis of the [18F]FDOPA BPND parametric maps revealed clusters of reduced binding potential in the 5.5 year old and 11 year old HD sheep compared to the 6 year old control sheep. In this first-of-its-kind study, we showed the usefulness and validity of HD sheep model in imaging cerebral glucose metabolism and dopamine uptake using PET imaging. The identification of discrete patterns of metabolic abnormality using [18F]FDG and decline of [18F]FDOPA uptake may provide a useful means of quantifying early HD-related changes in these models, particularly in the transition from presymptomatic to early symptomatic phases of HD.
亨廷顿氏病(Huntington's disease,HD)是一种神经退行性疾病,会引起认知、运动、行为和睡眠障碍,随着时间的推移会导致进行性残疾并最终死亡。通过在特征明确的大型 HD 动物模型中进行更多测试,可以加速新型疗法和成像探针的临床转化。我们的初步横断面研究的主要目标是证明独特的 HD 转基因绵羊模型(OVT73)在正电子发射断层扫描(PET)成像中的可行性和实用性。正电子发射断层成像研究在健康美利奴羊(6 岁,n = 3)和 OVT73 HD 羊(5.5 岁,n = 3 和 11 岁,n = 3)中进行。使用绵羊大脑模板为区域分析定义了感兴趣区和大脑图谱标签。我们使用[18F]氟脱氧葡萄糖([18F]FDG)来比较对照组和 HD 羊的区域脑糖代谢和 FDG 摄取的变化。我们还使用[18F]氟-3,4-二羟基苯丙氨酸([18F]FDOPA)比较了两组绵羊纹状体功能障碍的程度,并评估了两组绵羊主要脑区的结合电位(BPND)。与健康对照组和11岁的HD绵羊相比,5.5岁的HD绵羊在几个皮层和皮层下脑区的[18F]FDG摄取量明显增加(P < 0.05-0.01)。健康对照组和 11 岁 HD 羊的[18F]FDG 摄取量没有差异。对[18F]FDOPA BPND参数图的分析表明,与6岁的对照组绵羊相比,5.5岁和11岁的HD绵羊的结合潜力降低。在这项首创性研究中,我们利用 PET 成像技术展示了 HD 羊模型在脑葡萄糖代谢和多巴胺摄取成像中的实用性和有效性。利用[18F]FDG和[18F]FDOPA摄取量的下降识别代谢异常的离散模式,可为量化这些模型中与HD相关的早期变化提供有用的方法,尤其是在HD从症状前阶段向症状早期阶段过渡的过程中。
{"title":"Imaging Glucose Metabolism and Dopaminergic Dysfunction in Sheep (<i>Ovis aries</i>) Brain Using Positron Emission Tomography Imaging Reveals Abnormalities in OVT73 Huntington's Disease Sheep.","authors":"Georgia K Williams, Jordy Akkermans, Matt Lawson, Patryk Syta, Steven Staelens, Mohit H Adhikari, A Jennifer Morton, Björn Nitzsche, Johannes Boltze, Chris Christou, Daniele Bertoglio, Muneer Ahamed","doi":"10.1021/acschemneuro.4c00561","DOIUrl":"10.1021/acschemneuro.4c00561","url":null,"abstract":"<p><p>Huntington's disease (HD) is a neurodegenerative disease that causes cognitive, movement, behavioral, and sleep disturbances, which over time result in progressive disability and eventually death. Clinical translation of novel therapeutics and imaging probes could be accelerated by additional testing in well-characterized large animal models of HD. The major goal of our preliminary cross-sectional study is to demonstrate the feasibility and utility of the unique transgenic sheep model of HD (OVT73) in positron emission tomography (PET) imaging. PET imaging studies were performed in healthy merino sheep (6 year old, <i>n</i> = 3) and OVT73 HD sheep (5.5 year old, <i>n</i> = 3, and 11 year old, <i>n</i> = 3). Region-of-interest and brain atlas labels were defined for regional analyses by using a sheep brain template. [<sup>18</sup>F]fluorodeoxyglucose ([<sup>18</sup>F]FDG) was employed to compare the regional brain glucose metabolism and variations in FDG uptake between control and HD sheep. We also used [<sup>18</sup>F]fluoro-3,4-dihydroxyphenylalanine ([<sup>18</sup>F]FDOPA) to compare the extent of striatal dysfunction and evaluated the binding potential (BP<sub>ND</sub>) in key brain regions between the groups. Compared with healthy controls and 11 year old HD sheep, the 5.5 year old HD sheep exhibited significantly increased [<sup>18</sup>F]FDG uptake in several cortical and subcortical brain regions (<i>P</i> < 0.05-0.01). No difference in [<sup>18</sup>F]FDG uptake was observed between healthy controls and 11 year old HD sheep. Analysis of the [<sup>18</sup>F]FDOPA BP<sub>ND</sub> parametric maps revealed clusters of reduced binding potential in the 5.5 year old and 11 year old HD sheep compared to the 6 year old control sheep. In this first-of-its-kind study, we showed the usefulness and validity of HD sheep model in imaging cerebral glucose metabolism and dopamine uptake using PET imaging. The identification of discrete patterns of metabolic abnormality using [<sup>18</sup>F]FDG and decline of [<sup>18</sup>F]FDOPA uptake may provide a useful means of quantifying early HD-related changes in these models, particularly in the transition from presymptomatic to early symptomatic phases of HD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"4082-4091"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453214","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}
A large number of clinical studies demonstrate that the ketogenic diet (KD) may be an effective approach to the reduction of epileptic seizures in children and adults. Such dietary therapy could also help pregnant women with epilepsy, especially since most antiseizure drugs have teratogenic action. However, there is a lack of medical data, considering the safety of using KD during gestation for the progeny. Therefore, we examined the influence of KD used prenatally in rats on the elemental composition of the selected brain regions in their offspring. For this purpose, synchrotron radiation-induced X-ray fluorescence (SR-XRF) microscopy was utilized, and elements such as P, S, K, Ca, Fe, and Zn were determined. Moreover, to verify whether the possible effects of KD are temporary or long-term, different stages of animal postnatal development were taken into account in our experiment. The obtained results confirmed the great applicability of SR-XRF microscopy to track the element changes occurring in the brain during postnatal development as well as those induced by prenatal exposure to the high-fat diet. The topographic analysis of the brains taken from offspring of mothers fed with KD during pregnancy and appropriate control individuals showed a potential influence of such dietary treatment on the brain levels of elements such as P and S. In the oldest progeny, a significant reduction of the surface of brain areas characterized by an increased P and S content, which histologically/morphologically correspond to white matter structures, was noticed. In turn, quantitative elemental analysis showed significantly decreased levels of Fe in the striatum and white matter of 30-day-old rats exposed prenatally to KD. This effect was temporary and was not noticed in adult animals. The observed abnormalities may be related to the changes in the accumulation of sphingomyelin and sulfatides and may testify about disturbances in the structure and integrity of the myelin, present in the white matter.
大量临床研究表明,生酮饮食(KD)可能是减少儿童和成人癫痫发作的有效方法。这种饮食疗法也可以帮助患有癫痫的孕妇,特别是因为大多数抗癫痫药物都有致畸作用。然而,考虑到在妊娠期间使用 KD 对胎儿的安全性,目前还缺乏医学数据。因此,我们研究了大鼠产前使用 KD 对其后代选定脑区元素组成的影响。为此,我们利用同步辐射诱导 X 射线荧光(SR-XRF)显微镜测定了 P、S、K、Ca、Fe 和 Zn 等元素。此外,为了验证 KD 可能产生的影响是暂时的还是长期的,我们在实验中考虑了动物出生后发育的不同阶段。所获得的结果证实,SR-XRF 显微镜非常适用于追踪产后发育过程中大脑中发生的元素变化,以及产前暴露于高脂肪饮食所诱发的元素变化。对母亲在怀孕期间喂食 KD 的后代和适当对照组的大脑进行的地形分析表明,这种饮食处理对大脑中 P 和 S 等元素的含量有潜在影响。反过来,定量元素分析表明,产前暴露于 KD 的 30 天大老鼠纹状体和白质中的铁含量明显下降。这种影响是暂时的,在成年动物中没有发现。观察到的异常情况可能与鞘磷脂和硫化物的积累变化有关,并可能证明白质中存在的髓鞘的结构和完整性受到了干扰。
{"title":"Element Changes Occurring in Brain Point at the White Matter Abnormalities in Rats Exposed to the Ketogenic Diet During Prenatal Life.","authors":"Marzena Rugieł, Zuzanna Setkowicz, Mateusz Czyzycki, Rolf Simon, Tilo Baumbach, Joanna Chwiej","doi":"10.1021/acschemneuro.4c00283","DOIUrl":"10.1021/acschemneuro.4c00283","url":null,"abstract":"<p><p>A large number of clinical studies demonstrate that the ketogenic diet (KD) may be an effective approach to the reduction of epileptic seizures in children and adults. Such dietary therapy could also help pregnant women with epilepsy, especially since most antiseizure drugs have teratogenic action. However, there is a lack of medical data, considering the safety of using KD during gestation for the progeny. Therefore, we examined the influence of KD used prenatally in rats on the elemental composition of the selected brain regions in their offspring. For this purpose, synchrotron radiation-induced X-ray fluorescence (SR-XRF) microscopy was utilized, and elements such as P, S, K, Ca, Fe, and Zn were determined. Moreover, to verify whether the possible effects of KD are temporary or long-term, different stages of animal postnatal development were taken into account in our experiment. The obtained results confirmed the great applicability of SR-XRF microscopy to track the element changes occurring in the brain during postnatal development as well as those induced by prenatal exposure to the high-fat diet. The topographic analysis of the brains taken from offspring of mothers fed with KD during pregnancy and appropriate control individuals showed a potential influence of such dietary treatment on the brain levels of elements such as P and S. In the oldest progeny, a significant reduction of the surface of brain areas characterized by an increased P and S content, which histologically/morphologically correspond to white matter structures, was noticed. In turn, quantitative elemental analysis showed significantly decreased levels of Fe in the striatum and white matter of 30-day-old rats exposed prenatally to KD. This effect was temporary and was not noticed in adult animals. The observed abnormalities may be related to the changes in the accumulation of sphingomyelin and sulfatides and may testify about disturbances in the structure and integrity of the myelin, present in the white matter.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"3932-3944"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142491025","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}
Pub Date : 2024-11-06Epub Date: 2024-10-23DOI: 10.1021/acschemneuro.4c00345
Lei Wu, Jianhuai Chen, Qiao Yu, Chao Lu, Yachun Shu
The occurrence and development of depression are closely related to disorders of the brain and peripheral substances. Abnormal metabolites in the blood affect the signal regulation function of the nerve center, which is one of the key factors for depression episodes. This study was focused on metabolites in serum and the mechanism of its antidepressant in the hippocampus. In the present study, serum metabolites in patients with depression were screened by metabolomic techniques. Various depressive mouse models and behavioral tests were used to assess its antidepressant effects. The expressions of inflammatory signaling were detected by using Western blot, ELISA, and immunofluorescence. We found that the metabolite hypoxanthine in the serum of patients with depression was significantly reduced, and the same result was also found in two mouse models of depression such as chronic unpredictable mild stress (CUMS) and social defeat stress (SD). By administering different doses of hypoxanthine (5, 10, 15 mg/kg), we found that only 15 mg/kg was able to significantly reduce the latency and increase food consumption in the novelty suppressed-feeding test (NSF), which was also able to reverse the depressive phenotypes of mice in the CUMS model after a single administration at 2 h later. Hypoxanthine obviously reduced the expressions of inflammation in serum and downregulated the expressions of MAPK and NLRP3-related pathways in the hippocampus in CUMS mice. Moreover, hypoxanthine also suppressed the activations of glial cells including GFAP and IBA-1 in hippocampal CA1, CA3, and dentate gyrus (DG). To sum up, hypoxanthine exerted antidepressant effect relying on the inhibition of peripheral and hippocampal inflammations by regulating MAPK, NLRP3-related pathways, and glial cells. This was the first time that we have found a disordered metabolite in patients with depression and further systematically demonstrated its efficacy and potential mechanism of antidepressants, providing new ideas for antidepressant drug development.
抑郁症的发生和发展与大脑和外周物质的紊乱密切相关。血液中代谢物异常会影响神经中枢的信号调节功能,是抑郁症发作的关键因素之一。本研究主要关注血清中的代谢物及其在海马中的抗抑郁机制。本研究采用代谢组学技术对抑郁症患者的血清代谢物进行了筛查。研究还使用了多种抑郁小鼠模型和行为测试来评估其抗抑郁作用。通过 Western 印迹、ELISA 和免疫荧光检测了炎症信号的表达。我们发现,抑郁症患者血清中的代谢物次黄嘌呤含量明显降低,在慢性不可预测轻度应激(CUMS)和社会挫败应激(SD)等两种抑郁症小鼠模型中也发现了同样的结果。通过给予不同剂量的次黄嘌呤(5、10、15毫克/千克),我们发现只有15毫克/千克的次黄嘌呤能够明显减少新奇抑制性进食试验(NSF)的潜伏期并增加食物消耗量,而且在2小时后单次给药还能逆转CUMS模型小鼠的抑郁表型。次黄嘌呤明显降低了CUMS小鼠血清中炎症的表达,并下调了海马中MAPK和NLRP3相关通路的表达。此外,次黄嘌呤还抑制了海马CA1、CA3和齿状回(DG)神经胶质细胞(包括GFAP和IBA-1)的活化。总之,次黄嘌呤通过调节MAPK、NLRP3相关通路和神经胶质细胞,抑制外周和海马炎症,从而发挥抗抑郁作用。这是我们首次在抑郁症患者体内发现一种紊乱的代谢物,并进一步系统地证明了其抗抑郁的功效和潜在机制,为抗抑郁药物的研发提供了新思路。
{"title":"Hypoxanthine Produces Rapid Antidepressant Effects by Suppressing Inflammation in Serum and Hippocampus.","authors":"Lei Wu, Jianhuai Chen, Qiao Yu, Chao Lu, Yachun Shu","doi":"10.1021/acschemneuro.4c00345","DOIUrl":"10.1021/acschemneuro.4c00345","url":null,"abstract":"<p><p>The occurrence and development of depression are closely related to disorders of the brain and peripheral substances. Abnormal metabolites in the blood affect the signal regulation function of the nerve center, which is one of the key factors for depression episodes. This study was focused on metabolites in serum and the mechanism of its antidepressant in the hippocampus. In the present study, serum metabolites in patients with depression were screened by metabolomic techniques. Various depressive mouse models and behavioral tests were used to assess its antidepressant effects. The expressions of inflammatory signaling were detected by using Western blot, ELISA, and immunofluorescence. We found that the metabolite hypoxanthine in the serum of patients with depression was significantly reduced, and the same result was also found in two mouse models of depression such as chronic unpredictable mild stress (CUMS) and social defeat stress (SD). By administering different doses of hypoxanthine (5, 10, 15 mg/kg), we found that only 15 mg/kg was able to significantly reduce the latency and increase food consumption in the novelty suppressed-feeding test (NSF), which was also able to reverse the depressive phenotypes of mice in the CUMS model after a single administration at 2 h later. Hypoxanthine obviously reduced the expressions of inflammation in serum and downregulated the expressions of MAPK and NLRP3-related pathways in the hippocampus in CUMS mice. Moreover, hypoxanthine also suppressed the activations of glial cells including GFAP and IBA-1 in hippocampal CA1, CA3, and dentate gyrus (DG). To sum up, hypoxanthine exerted antidepressant effect relying on the inhibition of peripheral and hippocampal inflammations by regulating MAPK, NLRP3-related pathways, and glial cells. This was the first time that we have found a disordered metabolite in patients with depression and further systematically demonstrated its efficacy and potential mechanism of antidepressants, providing new ideas for antidepressant drug development.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"3970-3980"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142491026","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}
Alzheimer's disease (AD) is a common neurodegenerative disease causing cognitive and memory decline. AD is characterized by the deposition of amyloid-β and hypophosphorylated forms of tau protein. AD brains are found to be associated with neurodegeneration, oxidative stress, and inflammation. Cannabidiol (CBD) shows neuroprotective, antioxidant, and anti-inflammatory properties and simultaneously reduces amyloid-β production and tau hyperphosphorylation. The brain-derived neurotrophic factor (BDNF) plays a vital role in the development and maintenance of the plasticity of the central nervous system. A decline of BDNF levels in AD patients results in reduced plasticity and neuronal cell death. Current therapeutics against AD are limited to only symptomatic relief, necessitating a therapeutic strategy that reverses cognitive decline. In this scenario, combination therapy of CBD and BDNF could be a fruitful strategy for the treatment of AD. We designed mannose-conjugated chitosan-coated poly(d,l-lactide-co-glycolide (PLGA) (CHTMAN-PLGA) nanoparticles for the codelivery of CBD and BDNF to the brain. Chitosan is modified with mannose to specifically target the glucose transporter-1 (GLUT-1) receptor abundantly present in the blood-brain barrier for selectively delivering therapeutics to the brain. The CBD-encapsulated nanoparticles showed an average hydrodynamic diameter of 306 ± 8.12 nm and a zeta potential of 31.7 ± 1.53 mV. The coated nanoparticles prolonged encapsulated CBD release from the PLGA matrix. The coated nanoparticles exhibited sustained release of CBD for up to 22 days with 91.68 ± 2.91% release of the encapsulated drug. The coated nanoparticles, which had a high positive zeta potential (31.7 ± 1.53 mV), encapsulated the plasmid DNA. The qualitative transfection efficiency was investigated using CHTMAN-PLGA-CBD/pGFP in bEND.3, primary astrocytes, and primary neurons, while the quantitative transfection efficiency of the delivery system was determined using CHTMAN-PLGA-CBD/pBDNF. In vitro, the pBDNF transfection study revealed that the BDNF expression was 4-fold higher for CHTMAN-PLGA-CBD/pBDNF than for naked pBDNF in all of the cell lines. The cytotoxicity and hemocompatibility of the designed nanoparticles were tested in bEND.3 cells and red blood cells, respectively, and the nanoparticles were found to be nontoxic and hemocompatible. Hence, mannose-conjugated chitosan-coated PLGA nanoparticles could be useful as brain-targeting delivery vehicles for the codelivery of CBD and BDNF for possible AD treatment.
{"title":"Mannose-Functionalized Chitosan-Coated PLGA Nanoparticles for Brain-Targeted Codelivery of CBD and BDNF for the Treatment of Alzheimer's Disease.","authors":"Arun Kumar Mahanta, Bivek Chaulagain, Riddhi Trivedi, Jagdish Singh","doi":"10.1021/acschemneuro.4c00392","DOIUrl":"10.1021/acschemneuro.4c00392","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a common neurodegenerative disease causing cognitive and memory decline. AD is characterized by the deposition of amyloid-β and hypophosphorylated forms of tau protein. AD brains are found to be associated with neurodegeneration, oxidative stress, and inflammation. Cannabidiol (CBD) shows neuroprotective, antioxidant, and anti-inflammatory properties and simultaneously reduces amyloid-β production and tau hyperphosphorylation. The brain-derived neurotrophic factor (BDNF) plays a vital role in the development and maintenance of the plasticity of the central nervous system. A decline of BDNF levels in AD patients results in reduced plasticity and neuronal cell death. Current therapeutics against AD are limited to only symptomatic relief, necessitating a therapeutic strategy that reverses cognitive decline. In this scenario, combination therapy of CBD and BDNF could be a fruitful strategy for the treatment of AD. We designed mannose-conjugated chitosan-coated poly(d,l-lactide-<i>co</i>-glycolide (PLGA) (CHTMAN-PLGA) nanoparticles for the codelivery of CBD and BDNF to the brain. Chitosan is modified with mannose to specifically target the glucose transporter-1 (GLUT-1) receptor abundantly present in the blood-brain barrier for selectively delivering therapeutics to the brain. The CBD-encapsulated nanoparticles showed an average hydrodynamic diameter of 306 ± 8.12 nm and a zeta potential of 31.7 ± 1.53 mV. The coated nanoparticles prolonged encapsulated CBD release from the PLGA matrix. The coated nanoparticles exhibited sustained release of CBD for up to 22 days with 91.68 ± 2.91% release of the encapsulated drug. The coated nanoparticles, which had a high positive zeta potential (31.7 ± 1.53 mV), encapsulated the plasmid DNA. The qualitative transfection efficiency was investigated using CHTMAN-PLGA-CBD/pGFP in bEND.3, primary astrocytes, and primary neurons, while the quantitative transfection efficiency of the delivery system was determined using CHTMAN-PLGA-CBD/pBDNF. In vitro, the pBDNF transfection study revealed that the BDNF expression was 4-fold higher for CHTMAN-PLGA-CBD/pBDNF than for naked pBDNF in all of the cell lines. The cytotoxicity and hemocompatibility of the designed nanoparticles were tested in bEND.3 cells and red blood cells, respectively, and the nanoparticles were found to be nontoxic and hemocompatible. Hence, mannose-conjugated chitosan-coated PLGA nanoparticles could be useful as brain-targeting delivery vehicles for the codelivery of CBD and BDNF for possible AD treatment.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"4021-4032"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386352","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}
Pub Date : 2024-11-06Epub Date: 2024-10-21DOI: 10.1021/acschemneuro.4c00484
Debsankar Saha Roy, Ankit Singh, Vidita A Vaidya, Daniel Huster, Kaustubh R Mote, Sudipta Maiti
Serotonergic psychedelics, known for their hallucinogenic effects, have attracted interest due to their ability to enhance neuronal plasticity and potential therapeutic benefits. Although psychedelic-enhanced neuroplasticity is believed to require activation of 5-hydroxytryptamine (serotonin) 2A receptors (5-HT2ARs), serotonin itself is less effective in promoting such plasticity. Also, the psychoplastogenic effects of these molecules correlate with their lipophilicity, leading to suggestions that they act by influencing the intracellular receptors. However, their lipophilicity also implies that a significant quantity of lipids is accumulated in the lipid bilayer, potentially altering the physical properties of the membrane. Here, we probe whether the serotonergic psychedelic 2,5-dimethoxy-4-iodoamphetamine (DOI) can affect the properties of artificial lipid bilayers and if that can potentially affect processes such as membrane fusion. Solid-state NMR spectroscopy shows that the DOI strongly induces disorder in the lipid acyl chains. Atomic force microscopy shows that it can shrink the ordered domains in a biphasic lipid bilayer and can reduce the force needed to form nanopores in the membrane. Fluorescence correlation spectroscopy shows that DOI can promote vesicle association, and total internal fluorescence microscopy shows that it enhances vesicle fusion to a supported lipid bilayer. While serotonin has also recently been shown to cause similar effects, DOI is more than two orders of magnitude more potent in evoking these. Our results suggest that the receptor-independent effects of serotonergic psychedelics on lipid membranes may contribute to their biological actions, especially those that require significant membrane remodeling, such as neuronal plasticity.
血清素能迷幻剂以其致幻作用而闻名,由于其能够增强神经元的可塑性和潜在的治疗效果而备受关注。虽然迷幻药增强的神经可塑性被认为需要激活 5-羟色胺(5-羟色胺)2A 受体(5-HT2ARs),但 5-羟色胺本身在促进这种可塑性方面的效果较差。此外,这些分子的精神塑性作用与它们的亲脂性相关,因此有人认为它们是通过影响细胞内受体而发挥作用的。然而,它们的亲脂性也意味着脂质双分子层中积累了大量的脂质,可能会改变膜的物理特性。在此,我们探究了5-羟色胺能迷幻剂2,5-二甲氧基-4-碘安非他明(DOI)是否会影响人工脂质双分子层的特性,以及是否会对膜融合等过程产生潜在影响。固态核磁共振光谱显示,DOI 会强烈诱发脂质酰基链的紊乱。原子力显微镜显示,它能缩小双相脂质双分子层中的有序结构域,并能降低在膜中形成纳米孔所需的力。荧光相关光谱学显示,DOI 能促进囊泡的结合,而全内部荧光显微镜则显示,它能增强囊泡与支撑脂质双分子层的融合。虽然血清素最近也被证明能产生类似的效应,但 DOI 在唤起这些效应方面的作用要强两个数量级以上。我们的研究结果表明,5-羟色胺能迷幻剂对脂质膜的受体无关效应可能有助于它们的生物作用,特别是那些需要显著膜重塑的作用,如神经元可塑性。
{"title":"Effects of a Serotonergic Psychedelic on the Lipid Bilayer.","authors":"Debsankar Saha Roy, Ankit Singh, Vidita A Vaidya, Daniel Huster, Kaustubh R Mote, Sudipta Maiti","doi":"10.1021/acschemneuro.4c00484","DOIUrl":"10.1021/acschemneuro.4c00484","url":null,"abstract":"<p><p>Serotonergic psychedelics, known for their hallucinogenic effects, have attracted interest due to their ability to enhance neuronal plasticity and potential therapeutic benefits. Although psychedelic-enhanced neuroplasticity is believed to require activation of 5-hydroxytryptamine (serotonin) 2A receptors (5-HT<sub>2A</sub>Rs), serotonin itself is less effective in promoting such plasticity. Also, the psychoplastogenic effects of these molecules correlate with their lipophilicity, leading to suggestions that they act by influencing the intracellular receptors. However, their lipophilicity also implies that a significant quantity of lipids is accumulated in the lipid bilayer, potentially altering the physical properties of the membrane. Here, we probe whether the serotonergic psychedelic 2,5-dimethoxy-4-iodoamphetamine (DOI) can affect the properties of artificial lipid bilayers and if that can potentially affect processes such as membrane fusion. Solid-state NMR spectroscopy shows that the DOI strongly induces disorder in the lipid acyl chains. Atomic force microscopy shows that it can shrink the ordered domains in a biphasic lipid bilayer and can reduce the force needed to form nanopores in the membrane. Fluorescence correlation spectroscopy shows that DOI can promote vesicle association, and total internal fluorescence microscopy shows that it enhances vesicle fusion to a supported lipid bilayer. While serotonin has also recently been shown to cause similar effects, DOI is more than two orders of magnitude more potent in evoking these. Our results suggest that the receptor-independent effects of serotonergic psychedelics on lipid membranes may contribute to their biological actions, especially those that require significant membrane remodeling, such as neuronal plasticity.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"4066-4074"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453211","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}
Pub Date : 2024-11-06Epub Date: 2024-10-14DOI: 10.1021/acschemneuro.4c00328
Carmen Pérez de la Lastra Aranda, Carlota Tosat-Bitrián, Gracia Porras, Ruxandra Dafinca, Diego Muñoz-Torrero, Kevin Talbot, Ángeles Martín-Requero, Ana Martínez, Valle Palomo
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that currently lacks effective therapy. Given the heterogeneity of clinical and molecular profiles of ALS patients, personalized diagnostics and pathological characterization represent a powerful strategy to optimize patient stratification, thereby enabling personalized treatment. Immortalized lymphocytes from sporadic and genetic ALS patients recapitulate some pathological hallmarks of the disease, facilitating the fundamental task of drug screening. However, the molecular aggregation of ALS has not been characterized in this patient-derived cellular model. Indeed, protein aggregation is one of the most prominent features of neurodegenerative diseases, and therefore, models to test drugs against personalized pathological aggregation could help discover improved therapies. With this work, we aimed to characterize the aggregation profile of ALS immortalized lymphocytes and test several drug candidates with different mechanisms of action. In addition, we have evaluated the molecular aggregation in motor neurons derived from two hiPSC cell lines corresponding to ALS patients with different mutations in TARDBP. The results provide valuable insight into the different characterization of sporadic and genetic ALS patients' immortalized lymphocytes, their differential response to drug treatment, and the usefulness of proteome homeostasis characterization in patients' cells.
肌萎缩性脊髓侧索硬化症(ALS)是一种进行性神经退行性疾病,目前缺乏有效的治疗方法。鉴于肌萎缩侧索硬化症患者临床和分子特征的异质性,个性化诊断和病理特征描述是优化患者分层的有力策略,从而实现个性化治疗。来自散发性和遗传性 ALS 患者的永久化淋巴细胞再现了该病的一些病理特征,为药物筛选的基本任务提供了便利。然而,在这种源于患者的细胞模型中,渐冻人症的分子聚集特征尚未得到描述。事实上,蛋白质聚集是神经退行性疾病最显著的特征之一,因此,针对个性化病理聚集测试药物的模型有助于发现更好的疗法。通过这项工作,我们旨在描述 ALS 永生淋巴细胞的聚集特征,并测试几种具有不同作用机制的候选药物。此外,我们还评估了从两个与 TARDBP 不同突变的 ALS 患者对应的 hiPSC 细胞系中提取的运动神经元的分子聚集情况。这些结果为我们深入了解散发性和遗传性 ALS 患者永生淋巴细胞的不同特征、它们对药物治疗的不同反应以及患者细胞中蛋白质组平衡特征的有用性提供了宝贵的见解。
{"title":"Proteome Aggregation in Cells Derived from Amyotrophic Lateral Sclerosis Patients for Personalized Drug Evaluation.","authors":"Carmen Pérez de la Lastra Aranda, Carlota Tosat-Bitrián, Gracia Porras, Ruxandra Dafinca, Diego Muñoz-Torrero, Kevin Talbot, Ángeles Martín-Requero, Ana Martínez, Valle Palomo","doi":"10.1021/acschemneuro.4c00328","DOIUrl":"10.1021/acschemneuro.4c00328","url":null,"abstract":"<p><p>Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder that currently lacks effective therapy. Given the heterogeneity of clinical and molecular profiles of ALS patients, personalized diagnostics and pathological characterization represent a powerful strategy to optimize patient stratification, thereby enabling personalized treatment. Immortalized lymphocytes from sporadic and genetic ALS patients recapitulate some pathological hallmarks of the disease, facilitating the fundamental task of drug screening. However, the molecular aggregation of ALS has not been characterized in this patient-derived cellular model. Indeed, protein aggregation is one of the most prominent features of neurodegenerative diseases, and therefore, models to test drugs against personalized pathological aggregation could help discover improved therapies. With this work, we aimed to characterize the aggregation profile of ALS immortalized lymphocytes and test several drug candidates with different mechanisms of action. In addition, we have evaluated the molecular aggregation in motor neurons derived from two hiPSC cell lines corresponding to ALS patients with different mutations in <i>TARDBP</i>. The results provide valuable insight into the different characterization of sporadic and genetic ALS patients' immortalized lymphocytes, their differential response to drug treatment, and the usefulness of proteome homeostasis characterization in patients' cells.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"3945-3953"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453215","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}
Pub Date : 2024-11-06Epub Date: 2024-10-24DOI: 10.1021/acschemneuro.4c00687
Hanming Zheng, Jiaru He, Greta S P Mok, Zhihai Qiu
Ultrasound neuromodulation is a promising noninvasive technique capable of penetrating the skull and precisely targeting deep brain regions with millimeter accuracy. Recent studies have demonstrated that transcranial ultrasound stimulation (TUS) of sleep-related brain areas can induce sleep in mice and even trigger a reversible, hibernation-like state without causing damage. Beyond its utility in preclinical models of central nervous system diseases, such as epilepsy, tremors, Alzheimer's disease, and depression, TUS holds significant potential for clinical translation. Given that many neurological disorders, including Alzheimer's and Parkinson's disease, are associated with sleep abnormalities, leveraging clinical TUS applications for these diseases also creates a pathway for translating this technology to sleep modulation in human use. These findings highlight the potential for ultrasound neuromodulation to advance neuroscience research and clinical applications in sleep control.
超声神经调控是一种前景广阔的非侵入性技术,能够穿透头骨,以毫米级的精度精确瞄准大脑深部区域。最近的研究表明,经颅超声刺激(TUS)睡眠相关脑区可以诱导小鼠睡眠,甚至引发可逆的类似冬眠的状态,而不会造成损伤。除了在癫痫、震颤、阿尔茨海默病和抑郁症等中枢神经系统疾病的临床前模型中发挥作用外,TUS 在临床转化方面也具有巨大潜力。鉴于包括阿尔茨海默病和帕金森病在内的许多神经系统疾病都与睡眠异常有关,利用临床应用 TUS 治疗这些疾病也为将这项技术转化为人类睡眠调节技术开辟了一条途径。这些发现凸显了超声神经调控在推进神经科学研究和睡眠控制临床应用方面的潜力。
{"title":"Ultrasound Neuromodulation for Sleep and Neurological Disorder Therapy: A Path to Clinical Translation.","authors":"Hanming Zheng, Jiaru He, Greta S P Mok, Zhihai Qiu","doi":"10.1021/acschemneuro.4c00687","DOIUrl":"10.1021/acschemneuro.4c00687","url":null,"abstract":"<p><p>Ultrasound neuromodulation is a promising noninvasive technique capable of penetrating the skull and precisely targeting deep brain regions with millimeter accuracy. Recent studies have demonstrated that transcranial ultrasound stimulation (TUS) of sleep-related brain areas can induce sleep in mice and even trigger a reversible, hibernation-like state without causing damage. Beyond its utility in preclinical models of central nervous system diseases, such as epilepsy, tremors, Alzheimer's disease, and depression, TUS holds significant potential for clinical translation. Given that many neurological disorders, including Alzheimer's and Parkinson's disease, are associated with sleep abnormalities, leveraging clinical TUS applications for these diseases also creates a pathway for translating this technology to sleep modulation in human use. These findings highlight the potential for ultrasound neuromodulation to advance neuroscience research and clinical applications in sleep control.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"3797-3799"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142491027","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}
Pub Date : 2024-11-06Epub Date: 2024-10-14DOI: 10.1021/acschemneuro.4c00399
Maxence Noel, Suttipong Suttapitugsakul, Mohui Wei, Catherine Tilton, Akul Y Mehta, Yasuyuki Matsumoto, Jamie Heimburg-Molinaro, Robert G Mealer, Richard D Cummings
The synapse is an essential connection between neuronal cells in which the membrane and secreted glycoproteins regulate neurotransmission. The post-translational modifications of glycoproteins with carbohydrates, although essential for their functions as well as their specific localization, are not well understood. Oddly, whereas galactose addition to glycoproteins is required for neuronal functions, galactosylation is severely restricted for Asn-linked on N-glycans in the brain, and genetic evidence highlights the important roles of galactose in brain functions and development. To explore this novel glycosylation, we exploited an orthogonal technology in which a biotinylated sialic acid derivative (CMP-biotin-Sia) is transferred to terminally galactosylated proteins by a recombinant sialyltransferase (rST6Gal1). This approach allowed us to identify the carrier proteins as well as their localization on brain sections. Immunohistochemical analysis of the biotinylated glycoproteins in brain sections demonstrates that they are largely positioned in the pre- and postsynaptic membranes. Consistent with this positioning, glycoproteomic analyses of the labeled glycoproteins identified a number of them that are involved in synaptic function, cell adhesion, and extracellular matrix interactions. The discovery of these galactosylated N-glycoproteins and their relative confinement to synapses provide novel insights into the unusual and specific nature of protein glycosylation in the brain.
{"title":"Unique Glycans in Synaptic Glycoproteins in Mouse Brain.","authors":"Maxence Noel, Suttipong Suttapitugsakul, Mohui Wei, Catherine Tilton, Akul Y Mehta, Yasuyuki Matsumoto, Jamie Heimburg-Molinaro, Robert G Mealer, Richard D Cummings","doi":"10.1021/acschemneuro.4c00399","DOIUrl":"10.1021/acschemneuro.4c00399","url":null,"abstract":"<p><p>The synapse is an essential connection between neuronal cells in which the membrane and secreted glycoproteins regulate neurotransmission. The post-translational modifications of glycoproteins with carbohydrates, although essential for their functions as well as their specific localization, are not well understood. Oddly, whereas galactose addition to glycoproteins is required for neuronal functions, galactosylation is severely restricted for Asn-linked on N-glycans in the brain, and genetic evidence highlights the important roles of galactose in brain functions and development. To explore this novel glycosylation, we exploited an orthogonal technology in which a biotinylated sialic acid derivative (CMP-biotin-Sia) is transferred to terminally galactosylated proteins by a recombinant sialyltransferase (rST6Gal1). This approach allowed us to identify the carrier proteins as well as their localization on brain sections. Immunohistochemical analysis of the biotinylated glycoproteins in brain sections demonstrates that they are largely positioned in the pre- and postsynaptic membranes. Consistent with this positioning, glycoproteomic analyses of the labeled glycoproteins identified a number of them that are involved in synaptic function, cell adhesion, and extracellular matrix interactions. The discovery of these galactosylated N-glycoproteins and their relative confinement to synapses provide novel insights into the unusual and specific nature of protein glycosylation in the brain.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"4033-4045"},"PeriodicalIF":4.1,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142453218","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}