首页 > 最新文献

Molecular Brain最新文献

英文 中文
Attenuation of estrogen and its receptors in the post-menopausal stage exacerbates dyslipidemia and leads to cognitive impairment. 绝经后雌激素及其受体的衰减加剧了血脂异常并导致认知障碍。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-11-20 DOI: 10.1186/s13041-023-01068-0
Qinghai Meng, Ying Chao, Shurui Zhang, Xue Ding, Han Feng, Chenyan Zhang, Bowen Liu, Weijie Zhu, Yu Li, Qichun Zhang, Huangjin Tong, Lixing Wu, Huimin Bian

Cognitive dysfunction increases as menopause progresses. We previously found that estrogen receptors (ERs) contribute to dyslipidemia, but the specific relationship between ERs, dyslipidemia and cognitive dysfunction remains poorly understood. In the present study, we analyzed sequencing data from female hippocampus and normal breast aspirate samples from normal and Alzheimer's disease (AD) women, and the results suggest that abnormal ERs signaling is associated with dyslipidemia and cognitive dysfunction. We replicated a mouse model of dyslipidemia and postmenopausal status in LDLR-/- mice and treated them with β-estradiol or simvastatin, and found that ovariectomy in LDLR-/- mice led to an exacerbation of dyslipidemia and increased hippocampal apoptosis and cognitive impairment, which were associated with reduced estradiol levels and ERα, ERβ and GPER expression. In vitro, a lipid overload model of SH-SY-5Y cells was established and treated with inhibitors of ERs. β-estradiol or simvastatin effectively attenuated dyslipidemia-induced neuronal apoptosis via upregulation of ERs, whereas ERα, ERβ and GPER inhibitors together abolished the protective effect of simvastatin on lipid-induced neuronal apoptosis. We conclude that decreased estrogen and its receptor function in the postmenopausal stage promote neuronal damage and cognitive impairment by exacerbating dyslipidemia, and that estrogen supplementation or lipid lowering is an effective way to ameliorate hippocampal damage and cognitive dysfunction via upregulation of ERs.

认知功能障碍随着更年期的进展而增加。我们之前发现雌激素受体(er)有助于血脂异常,但雌激素受体、血脂异常和认知功能障碍之间的具体关系仍然知之甚少。在本研究中,我们分析了来自女性海马和正常和阿尔茨海默病(AD)女性的正常乳腺抽吸样本的测序数据,结果表明异常的er信号与血脂异常和认知功能障碍有关。我们在LDLR-/-小鼠中复制了血脂异常和绝经后状态的小鼠模型,并用β-雌二醇或辛伐他汀治疗它们,发现LDLR-/-小鼠卵巢切除术导致血脂异常加剧,海马细胞凋亡增加和认知功能障碍,这与雌二醇水平和ERα, ERβ和GPER表达降低有关。体外建立SH-SY-5Y细胞脂质过载模型,并用er抑制剂处理。β-雌二醇或辛伐他汀可通过上调内质网有效减轻血脂异常诱导的神经元凋亡,而ERα、ERβ和GPER抑制剂共同消除辛伐他汀对脂质诱导的神经元凋亡的保护作用。我们认为,绝经后雌激素及其受体功能的下降通过加剧血脂异常而促进神经元损伤和认知功能障碍,补充雌激素或降脂是通过上调雌激素受体来改善海马损伤和认知功能障碍的有效途径。
{"title":"Attenuation of estrogen and its receptors in the post-menopausal stage exacerbates dyslipidemia and leads to cognitive impairment.","authors":"Qinghai Meng, Ying Chao, Shurui Zhang, Xue Ding, Han Feng, Chenyan Zhang, Bowen Liu, Weijie Zhu, Yu Li, Qichun Zhang, Huangjin Tong, Lixing Wu, Huimin Bian","doi":"10.1186/s13041-023-01068-0","DOIUrl":"10.1186/s13041-023-01068-0","url":null,"abstract":"<p><p>Cognitive dysfunction increases as menopause progresses. We previously found that estrogen receptors (ERs) contribute to dyslipidemia, but the specific relationship between ERs, dyslipidemia and cognitive dysfunction remains poorly understood. In the present study, we analyzed sequencing data from female hippocampus and normal breast aspirate samples from normal and Alzheimer's disease (AD) women, and the results suggest that abnormal ERs signaling is associated with dyslipidemia and cognitive dysfunction. We replicated a mouse model of dyslipidemia and postmenopausal status in LDLR<sup>-/-</sup> mice and treated them with β-estradiol or simvastatin, and found that ovariectomy in LDLR<sup>-/-</sup> mice led to an exacerbation of dyslipidemia and increased hippocampal apoptosis and cognitive impairment, which were associated with reduced estradiol levels and ERα, ERβ and GPER expression. In vitro, a lipid overload model of SH-SY-5Y cells was established and treated with inhibitors of ERs. β-estradiol or simvastatin effectively attenuated dyslipidemia-induced neuronal apoptosis via upregulation of ERs, whereas ERα, ERβ and GPER inhibitors together abolished the protective effect of simvastatin on lipid-induced neuronal apoptosis. We conclude that decreased estrogen and its receptor function in the postmenopausal stage promote neuronal damage and cognitive impairment by exacerbating dyslipidemia, and that estrogen supplementation or lipid lowering is an effective way to ameliorate hippocampal damage and cognitive dysfunction via upregulation of ERs.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10662842/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138176799","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
Effects of MAP4K inhibition on neurite outgrowth. MAP4K抑制对神经突生长的影响。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-11-18 DOI: 10.1186/s13041-023-01066-2
Di Ja Lasham, Reza K Arta, Abdul Fuad Hadi, Jun Egawa, Vance P Lemmon, Toshiyuki Takasugi, Michihiro Igarashi, Toshiyuki Someya

Protein kinases are responsible for protein phosphorylation and are involved in important intracellular signal transduction pathways in various cells, including neurons; however, a considerable number of poorly characterized kinases may be involved in neuronal development. Here, we considered mitogen-activated protein kinase kinase kinase kinases (MAP4Ks), related to as candidate regulators of neurite outgrowth and synaptogenesis, by examining the effects of a selective MAP4K inhibitor PF06260933. PF06260933 treatments of the cultured neurons reduced neurite lengths, not the number of synapses, and phosphorylation of GAP43 and JNK, relative to the control. These results suggest that MAP4Ks are physiologically involved in normal neuronal development and that the resultant impaired neurite outgrowth by diminished MAP4Ks' activity, is related to psychiatric disorders.

蛋白激酶负责蛋白质磷酸化,并参与包括神经元在内的各种细胞的重要细胞内信号转导途径;然而,相当数量的特征不明确的激酶可能参与神经元发育。在这里,我们通过检测一种选择性MAP4K抑制剂PF06260933的作用,考虑了丝裂原激活的蛋白激酶激酶激酶激酶激酶激酶激酶激酶激酶(MAP4Ks)作为神经突生长和突触发生的候选调节因子。与对照组相比,PF06260933处理减少了培养神经元的神经突长度,而不是突触数量,并减少了GAP43和JNK的磷酸化。这些结果表明,MAP4Ks在生理上参与了正常的神经元发育,并且由于MAP4Ks活性降低而导致的神经突生长受损与精神疾病有关。
{"title":"Effects of MAP4K inhibition on neurite outgrowth.","authors":"Di Ja Lasham, Reza K Arta, Abdul Fuad Hadi, Jun Egawa, Vance P Lemmon, Toshiyuki Takasugi, Michihiro Igarashi, Toshiyuki Someya","doi":"10.1186/s13041-023-01066-2","DOIUrl":"10.1186/s13041-023-01066-2","url":null,"abstract":"<p><p>Protein kinases are responsible for protein phosphorylation and are involved in important intracellular signal transduction pathways in various cells, including neurons; however, a considerable number of poorly characterized kinases may be involved in neuronal development. Here, we considered mitogen-activated protein kinase kinase kinase kinases (MAP4Ks), related to as candidate regulators of neurite outgrowth and synaptogenesis, by examining the effects of a selective MAP4K inhibitor PF06260933. PF06260933 treatments of the cultured neurons reduced neurite lengths, not the number of synapses, and phosphorylation of GAP43 and JNK, relative to the control. These results suggest that MAP4Ks are physiologically involved in normal neuronal development and that the resultant impaired neurite outgrowth by diminished MAP4Ks' activity, is related to psychiatric disorders.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10656890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138047376","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
Optogenetic activation of dopamine D1 receptors in island cells of medial entorhinal cortex inhibits temporal association learning. 内嗅皮层岛细胞多巴胺D1受体的光遗传学激活抑制了时间关联学习。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-11-14 DOI: 10.1186/s13041-023-01065-3
Jun Yokose, Naoki Yamamoto, Sachie K Ogawa, Takashi Kitamura

A critical feature of episodic memory formation is to associate temporally segregated events as an episode, called temporal association learning. The medial entorhinal cortical-hippocampal (EC-HPC) networks is essential for temporal association learning. We have previously demonstrated that pyramidal cells in the medial EC (MEC) layer III project to the hippocampal CA1 pyramidal cells and are necessary for trace fear conditioning (TFC), which is an associative learning between tone and aversive shock with the temporal gap. On the other hand, Island cells in MECII, project to GABAergic neurons in hippocampal CA1, suppress the MECIII input into the CA1 pyramidal cells through the feed-forward inhibition, and inhibit TFC. However, it remains unknown about how Island cells activity is regulated during TFC. In this study, we report that dopamine D1 receptor is preferentially expressed in Island cells in the MEC. Optogenetic activation of dopamine D1 receptors in Island cells facilitate the Island cell activity and inhibited hippocampal CA1 pyramidal cell activity during TFC. The optogenetic activation caused the impairment of TFC memory recall without affecting contextual fear memory recall. These results suggest that dopamine D1 receptor in Island cells have a crucial role for the regulation of temporal association learning.

情景记忆形成的一个关键特征是将时间上分离的事件作为一个情节联系起来,称为时间关联学习。内侧内嗅皮层-海马(EC-HPC)网络对时间关联学习至关重要。我们之前已经证明,内侧EC (MEC)第三层的锥体细胞与海马CA1锥体细胞相连,并且是微量恐惧条件反射(TFC)所必需的,这是一种具有时间间隙的声调和厌恶性休克之间的关联学习。另一方面,MECII中的岛细胞,投射到海马CA1的gaba能神经元,通过前馈抑制抑制MECIII输入到CA1锥体细胞,抑制TFC。然而,在TFC过程中岛细胞的活性是如何被调节的尚不清楚。在本研究中,我们报道了多巴胺D1受体在MEC的岛细胞中优先表达。TFC时,光遗传学激活岛细胞多巴胺D1受体促进岛细胞活性,抑制海马CA1锥体细胞活性。光基因激活导致TFC记忆回忆受损,但不影响情境恐惧记忆的回忆。这些结果表明,岛细胞多巴胺D1受体在时间关联学习的调节中起着至关重要的作用。
{"title":"Optogenetic activation of dopamine D1 receptors in island cells of medial entorhinal cortex inhibits temporal association learning.","authors":"Jun Yokose, Naoki Yamamoto, Sachie K Ogawa, Takashi Kitamura","doi":"10.1186/s13041-023-01065-3","DOIUrl":"10.1186/s13041-023-01065-3","url":null,"abstract":"<p><p>A critical feature of episodic memory formation is to associate temporally segregated events as an episode, called temporal association learning. The medial entorhinal cortical-hippocampal (EC-HPC) networks is essential for temporal association learning. We have previously demonstrated that pyramidal cells in the medial EC (MEC) layer III project to the hippocampal CA1 pyramidal cells and are necessary for trace fear conditioning (TFC), which is an associative learning between tone and aversive shock with the temporal gap. On the other hand, Island cells in MECII, project to GABAergic neurons in hippocampal CA1, suppress the MECIII input into the CA1 pyramidal cells through the feed-forward inhibition, and inhibit TFC. However, it remains unknown about how Island cells activity is regulated during TFC. In this study, we report that dopamine D1 receptor is preferentially expressed in Island cells in the MEC. Optogenetic activation of dopamine D1 receptors in Island cells facilitate the Island cell activity and inhibited hippocampal CA1 pyramidal cell activity during TFC. The optogenetic activation caused the impairment of TFC memory recall without affecting contextual fear memory recall. These results suggest that dopamine D1 receptor in Island cells have a crucial role for the regulation of temporal association learning.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10647136/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"107591768","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
Impact of volume and expression time in an AAV-delivered channelrhodopsin. AAV递送的通道视紫红质中体积和表达时间的影响。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-11-10 DOI: 10.1186/s13041-023-01067-1
Sanaz Ansarifar, Gabija Andreikė, Milad Nazari, Rodrigo Labouriau, Sadegh Nabavi, Andrea Moreno

Optogenetics has revolutionised neuroscience research, but at the same time has brought a plethora of new variables to consider when designing an experiment with AAV-based targeted gene delivery. Some concerns have been raised regarding the impact of AAV injection volume and expression time in relation to longitudinal experimental designs. In this study, we investigated the efficiency of optically evoked post-synaptic responses in connection to two variables: the volume of the injected virus and the expression time of the virus. For this purpose, we expressed the blue-shifted ChR2, oChIEF, employing a widely used AAV vector delivery strategy. We found that the volume of the injected virus has a minimal impact on the efficiency of optically-evoked postsynaptic population responses. The expression time, on the other hand, has a pronounced effect, with a gradual reduction in the population responses beyond 4 weeks of expression. We strongly advise to monitor time-dependent expression profiles when planning or conducting long-term experiments that depend on successful and stable channelrhodopsin expression.

光遗传学彻底改变了神经科学研究,但同时也带来了大量新的变量,在设计基于AAV的靶向基因递送实验时需要考虑。关于AAV注射量和表达时间对纵向实验设计的影响,已经提出了一些担忧。在这项研究中,我们研究了与两个变量有关的光学诱发突触后反应的效率:注射病毒的体积和病毒的表达时间。为此,我们使用广泛使用的AAV载体递送策略表达了蓝移ChR2,oChIEF。我们发现,注射病毒的体积对光学诱发的突触后群体反应的效率影响最小。另一方面,表达时间具有显著的影响,在表达4周后,群体反应逐渐减少。我们强烈建议在计划或进行依赖于成功和稳定的通道视紫红质表达的长期实验时,监测与时间相关的表达谱。
{"title":"Impact of volume and expression time in an AAV-delivered channelrhodopsin.","authors":"Sanaz Ansarifar, Gabija Andreikė, Milad Nazari, Rodrigo Labouriau, Sadegh Nabavi, Andrea Moreno","doi":"10.1186/s13041-023-01067-1","DOIUrl":"10.1186/s13041-023-01067-1","url":null,"abstract":"<p><p>Optogenetics has revolutionised neuroscience research, but at the same time has brought a plethora of new variables to consider when designing an experiment with AAV-based targeted gene delivery. Some concerns have been raised regarding the impact of AAV injection volume and expression time in relation to longitudinal experimental designs. In this study, we investigated the efficiency of optically evoked post-synaptic responses in connection to two variables: the volume of the injected virus and the expression time of the virus. For this purpose, we expressed the blue-shifted ChR2, oChIEF, employing a widely used AAV vector delivery strategy. We found that the volume of the injected virus has a minimal impact on the efficiency of optically-evoked postsynaptic population responses. The expression time, on the other hand, has a pronounced effect, with a gradual reduction in the population responses beyond 4 weeks of expression. We strongly advise to monitor time-dependent expression profiles when planning or conducting long-term experiments that depend on successful and stable channelrhodopsin expression.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10638758/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72210083","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
Chronic pregabalin treatment protects against spreading depolarization and alters hippocampal synaptic characteristics in a model of familial hemiplegic migraine-type 1. 在家族性1型偏瘫偏头痛模型中,普瑞巴林的慢性治疗可防止扩散性去极化并改变海马突触特征。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-11-03 DOI: 10.1186/s13041-023-01062-6
Stuart M Cain, Sascha R A Alles, Ray Gopaul, Louis-Philippe Bernier, Andrew C Yung, Andrew Bauman, Yi Yang, Glen B Baker, Piotr Kozlowski, Brian A MacVicar, Terrance P Snutch

Familial hemiplegic migraine type-1 (FHM-1) is a form of migraine with aura caused by mutations in the P/Q-type (Cav2.1) voltage-gated calcium channel. Pregabalin, used clinically in the treatment of chronic pain and epilepsy, inhibits P/Q-type calcium channel activity and recent studies suggest that it may have potential for the treatment of migraine. Spreading Depolarization (SD) is a neurophysiological phenomenon that can occur during migraine with aura by propagating a wave of silenced neuronal function through cortex and sometimes subcortical brain structures. Here, utilizing an optogenetic stimulation technique optimized to allow for non-invasive initiation of cortical SD, we demonstrate that chronic pregabalin administration [12 mg/kg/day (s.c.)] in vivo increased the threshold for cortical spreading depolarization in transgenic mice harboring the clinically-relevant Cav2.1S218L mutation (S218L). In addition, chronic pregabalin treatment limited subcortical propagation of recurrent spreading depolarization events to the striatum and hippocampus in both wild-type and S218L mice. To examine contributing underlying mechanisms of action of chronic pregabalin, we performed whole-cell patch-clamp electrophysiology in CA1 neurons in ex vivo brain slices from mice treated with chronic pregabalin vs vehicle. In WT mice, chronic pregabalin produced a decrease in spontaneous excitatory postsynaptic current (sEPSC) amplitude with no effect on frequency. In contrast, in S218L mice chronic pregabalin produced an increase in sEPSC amplitude and decreased frequency. These electrophysiological findings suggest that in FHM-1 mice chronic pregabalin acts through both pre- and post-synaptic mechanisms in CA1 hippocampal neurons to elicit FHM-1 genotype-specific inhibitory action. The results highlight the potential of chronic pregabalin to limit recurrent SD to subcortical brain structures during pathophysiological events in both the genetically-normal and FHM-1 brain. The work further provides insights into FHM-1 pathophysiology and the potential for chronic pregabalin treatment to prevent SD in migraineurs.

家族性1型偏瘫性偏头痛(FHM-1)是一种由P/Q型(Cav2.1)电压门控钙通道突变引起的先兆偏头痛。临床上用于治疗慢性疼痛和癫痫的普瑞巴林抑制P/Q型钙通道活性,最近的研究表明,它可能具有治疗偏头痛的潜力。扩散性去极化(SD)是一种神经生理学现象,在先兆偏头痛期间,通过皮层,有时是皮层下的大脑结构传播沉默的神经元功能。在这里,利用一种优化的光遗传学刺激技术,允许无创启动皮层SD,我们证明了体内长期给予普瑞巴林[12mg/kg/天(皮下注射)]增加了携带临床相关Cav2.1S218L突变(S218L)的转基因小鼠皮层扩散去极化的阈值。此外,在野生型和S218L小鼠中,慢性普瑞巴林治疗限制了复发性扩散去极化事件向纹状体和海马体的皮层下传播。为了研究慢性普瑞巴林的潜在作用机制,我们在用慢性普瑞巴林与载体治疗的小鼠离体脑切片中对CA1神经元进行了全细胞膜片钳电生理学研究。在WT小鼠中,慢性普瑞巴林导致自发兴奋性突触后电流(sEPSC)振幅降低,而对频率没有影响。相反,在S218L小鼠中,慢性普瑞巴林使sEPSC振幅增加,频率降低。这些电生理学发现表明,在FHM-1小鼠中,慢性普瑞巴林通过CA1海马神经元的突触前和突触后机制引发FHM-1基因型特异性抑制作用。该结果强调了慢性普瑞巴林在遗传正常和FHM-1脑的病理生理事件期间将复发性SD限制在皮质下脑结构的潜力。这项工作进一步深入了解了FHM-1的病理生理学以及普瑞巴林慢性治疗预防偏头痛SD的潜力。
{"title":"Chronic pregabalin treatment protects against spreading depolarization and alters hippocampal synaptic characteristics in a model of familial hemiplegic migraine-type 1.","authors":"Stuart M Cain, Sascha R A Alles, Ray Gopaul, Louis-Philippe Bernier, Andrew C Yung, Andrew Bauman, Yi Yang, Glen B Baker, Piotr Kozlowski, Brian A MacVicar, Terrance P Snutch","doi":"10.1186/s13041-023-01062-6","DOIUrl":"10.1186/s13041-023-01062-6","url":null,"abstract":"<p><p>Familial hemiplegic migraine type-1 (FHM-1) is a form of migraine with aura caused by mutations in the P/Q-type (Cav2.1) voltage-gated calcium channel. Pregabalin, used clinically in the treatment of chronic pain and epilepsy, inhibits P/Q-type calcium channel activity and recent studies suggest that it may have potential for the treatment of migraine. Spreading Depolarization (SD) is a neurophysiological phenomenon that can occur during migraine with aura by propagating a wave of silenced neuronal function through cortex and sometimes subcortical brain structures. Here, utilizing an optogenetic stimulation technique optimized to allow for non-invasive initiation of cortical SD, we demonstrate that chronic pregabalin administration [12 mg/kg/day (s.c.)] in vivo increased the threshold for cortical spreading depolarization in transgenic mice harboring the clinically-relevant Ca<sub>v</sub>2.1<sup>S218L</sup> mutation (S218L). In addition, chronic pregabalin treatment limited subcortical propagation of recurrent spreading depolarization events to the striatum and hippocampus in both wild-type and S218L mice. To examine contributing underlying mechanisms of action of chronic pregabalin, we performed whole-cell patch-clamp electrophysiology in CA1 neurons in ex vivo brain slices from mice treated with chronic pregabalin vs vehicle. In WT mice, chronic pregabalin produced a decrease in spontaneous excitatory postsynaptic current (sEPSC) amplitude with no effect on frequency. In contrast, in S218L mice chronic pregabalin produced an increase in sEPSC amplitude and decreased frequency. These electrophysiological findings suggest that in FHM-1 mice chronic pregabalin acts through both pre- and post-synaptic mechanisms in CA1 hippocampal neurons to elicit FHM-1 genotype-specific inhibitory action. The results highlight the potential of chronic pregabalin to limit recurrent SD to subcortical brain structures during pathophysiological events in both the genetically-normal and FHM-1 brain. The work further provides insights into FHM-1 pathophysiology and the potential for chronic pregabalin treatment to prevent SD in migraineurs.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10623724/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71483830","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
Limitations of the human iPSC-derived neuron model for early-onset Alzheimer's disease. 人类iPSC衍生的神经元模型对早发性阿尔茨海默病的局限性。
IF 3.3 3区 医学 Q2 NEUROSCIENCES Pub Date : 2023-11-03 DOI: 10.1186/s13041-023-01063-5
Phoebe Valdes, Kenneth W Henry, Michael Q Fitzgerald, Koushik Muralidharan, Andrew B Caldwell, Srinivasan Ramachandran, Lawrence S B Goldstein, William C Mobley, Douglas R Galasko, Shankar Subramaniam

Non-familial Alzheimer's disease (AD) occurring before 65 years of age is commonly referred to as early-onset Alzheimer's disease (EOAD) and constitutes ~ 5-6% of all AD cases (Mendez et al. in Continuum 25:34-51, 2019). While EOAD exhibits the same clinicopathological changes such as amyloid plaques, neurofibrillary tangles (NFTs), brain atrophy, and cognitive decline (Sirkis et al. in Mol Psychiatry 27:2674-88, 2022; Caldwell et al. in Mol Brain 15:83, 2022) as observed in the more prevalent late-onset AD (LOAD), EOAD patients tend to have more severe cognitive deficits, including visuospatial, language, and executive dysfunction (Sirkis et al. in Mol Psychiatry 27:2674-88, 2022). Patient-derived induced pluripotent stem cells (iPSCs) have been used to model and study penetrative, familial AD (FAD) mutations in APP, PSEN1, and PSEN2 (Valdes et al. in Research Square 1-30, 2022; Caldwell et al. in Sci Adv 6:1-16, 2020) but have been seldom used for sporadic forms of AD that display more heterogeneous disease mechanisms. In this study, we sought to characterize iPSC-derived neurons from EOAD patients via RNA sequencing. A modest difference in expression profiles between EOAD patients and non-demented control (NDC) subjects resulted in a limited number of differentially expressed genes (DEGs). Based on this analysis, we provide evidence that iPSC-derived neuron model systems, likely due to the loss of EOAD-associated epigenetic signatures arising from iPSC reprogramming, may not be ideal models for studying sporadic AD.

发生在65岁之前的非家族性阿尔茨海默病(AD)通常被称为早发性阿尔茨海默病,构成 ~ 占所有AD病例的5-6%(Mendez等人在Continuum 25:34-511919中)。虽然EOAD表现出与在更普遍的晚发性AD(LOAD)中观察到的相同的临床病理变化,如淀粉样蛋白斑块、神经原纤维缠结(NFT)、脑萎缩和认知能力下降(Sirkis等人在Mol Psychiatry 27:2674-882022;Caldwell等人在MolBrain 15:832022),和执行功能障碍(Sirkis等人,Mol Psychiatry 27:2674-882022)。患者来源的诱导多能干细胞(iPSC)已被用于模拟和研究APP、PSEN1和PSEN2中的穿透性家族性AD(FAD)突变(Valdes等人,Research Square 1-3022;Caldwell等人,Sci-Adv 6:1-162020),但很少用于表现出更多异质性疾病机制的散发形式的AD。在这项研究中,我们试图通过RNA测序来表征EOAD患者的iPSC衍生神经元。EOAD患者和非痴呆对照(NDC)受试者之间表达谱的适度差异导致差异表达基因(DEG)的数量有限。基于这一分析,我们提供了证据,证明iPSC衍生的神经元模型系统可能不是研究散发性AD的理想模型,这可能是由于iPSC重编程引起的EOAD相关表观遗传学特征的丧失。
{"title":"Limitations of the human iPSC-derived neuron model for early-onset Alzheimer's disease.","authors":"Phoebe Valdes, Kenneth W Henry, Michael Q Fitzgerald, Koushik Muralidharan, Andrew B Caldwell, Srinivasan Ramachandran, Lawrence S B Goldstein, William C Mobley, Douglas R Galasko, Shankar Subramaniam","doi":"10.1186/s13041-023-01063-5","DOIUrl":"10.1186/s13041-023-01063-5","url":null,"abstract":"<p><p>Non-familial Alzheimer's disease (AD) occurring before 65 years of age is commonly referred to as early-onset Alzheimer's disease (EOAD) and constitutes ~ 5-6% of all AD cases (Mendez et al. in Continuum 25:34-51, 2019). While EOAD exhibits the same clinicopathological changes such as amyloid plaques, neurofibrillary tangles (NFTs), brain atrophy, and cognitive decline (Sirkis et al. in Mol Psychiatry 27:2674-88, 2022; Caldwell et al. in Mol Brain 15:83, 2022) as observed in the more prevalent late-onset AD (LOAD), EOAD patients tend to have more severe cognitive deficits, including visuospatial, language, and executive dysfunction (Sirkis et al. in Mol Psychiatry 27:2674-88, 2022). Patient-derived induced pluripotent stem cells (iPSCs) have been used to model and study penetrative, familial AD (FAD) mutations in APP, PSEN1, and PSEN2 (Valdes et al. in Research Square 1-30, 2022; Caldwell et al. in Sci Adv 6:1-16, 2020) but have been seldom used for sporadic forms of AD that display more heterogeneous disease mechanisms. In this study, we sought to characterize iPSC-derived neurons from EOAD patients via RNA sequencing. A modest difference in expression profiles between EOAD patients and non-demented control (NDC) subjects resulted in a limited number of differentially expressed genes (DEGs). Based on this analysis, we provide evidence that iPSC-derived neuron model systems, likely due to the loss of EOAD-associated epigenetic signatures arising from iPSC reprogramming, may not be ideal models for studying sporadic AD.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.3,"publicationDate":"2023-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10623777/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71483831","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
MLKL regulates Cx43 ubiquitinational degradation and mediates neuronal necroptosis in ipsilateral thalamus after focal cortical infarction. MLKL调节Cx43的泛素降解并介导局灶性皮层梗死后同侧丘脑的神经元坏死。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-10-30 DOI: 10.1186/s13041-023-01064-4
Yanyan Tang, Quanhong Chu, Guanfeng Xie, Yafu Tan, Ziming Ye, Chao Qin

Necroptosis is known to play an important role in the pathophysiology of cerebral ischemia; however, its role in the occurrence of secondary thalamic injury after focal cerebral infarction and the mechanism about how mixed lineage kinase domain-like (MLKL) executes necroptosis in this pathophysiology are still unclear. In this study, Sprague-Dawley rats were subjected to distal branch of middle cerebral artery occlusion (dMCAO). The expression of MLKL, connexin 43 (Cx43) and Von Hippel-Lindau (VHL) in vitro and in vivo were assessed by Western blot. Bioinformatic methods were used to predict the potential binding sites where MLKL interacted with Cx43, and the ubiquitination degradation of Cx43 regulated by VHL. The interactions among MLKL, Cx43, VHL, and Ubiquitin were assessed by immunoprecipitation. Dye uptake assay were used to examine the Cx43 hemichannels. Intracellular Ca2+ concentration was measured using Fluo-4 AM. Overexpression and site-directed mutagenesis studies were used to study the mechanisms by which MLKL regulates Cx43 ubiquitinational degradation to mediate neuronal necroptosis. We found that MLKL and Cx43 were upregulated in the ventral posterolateral nucleus (VPN) of the ipsilateral thalamus after dMCAO. In the in vitro experiments MLKL and Cx43 were upregulated after TSZ-mediated necroptosis in SH-SY5Y cells. The interaction between MLKL and Cx43 inhibited the K48-linked ubiquitination of Cx43 in necroptotic SH-SY5Y cells. VHL is an E3 ubiquitin ligase for Cx43, and MLKL competes with VHL for binding to Cx43. Interaction of MLKL Ser454 with Cx43 can trigger the opening of Cx43 hemichannels, causing increased intracellular Ca2+, and cell necroptosis. This innovative study at animal models, cellular, and molecular levels is anticipated to clarify the roles of MLKL and Cx43 in thalamic damage after focal cortical infarction. Our findings may help identify novel targets for neurological recovery after cortical infarction.

众所周知,坏死在脑缺血的病理生理学中起着重要作用;然而,它在局灶性脑梗死后继发性丘脑损伤发生中的作用以及混合谱系激酶结构域样(MLKL)如何在这种病理生理学中执行坏死的机制仍不清楚。在本研究中,对Sprague-Dawley大鼠进行大脑中动脉远端支闭塞(dMCAO)。通过蛋白质印迹法评估MLKL、连接蛋白43(Cx43)和Von Hippel-Lindau(VHL)在体外和体内的表达。生物信息学方法用于预测MLKL与Cx43相互作用的潜在结合位点,以及VHL调节的Cx43的泛素化降解。通过免疫沉淀评估MLKL、Cx43、VHL和泛素之间的相互作用。用染料吸收测定法检测Cx43半通道。使用Fluo-4 AM测量细胞内Ca2+浓度。使用过表达和定点突变研究来研究MLKL调节Cx43泛素降解以介导神经元坏死的机制。我们发现,dMCAO后同侧丘脑腹侧后外侧核(VPN)的MLKL和Cx43上调。在体外实验中,TSZ介导的SH-SY5Y细胞坏死后,MLKL和Cx43上调。MLKL和Cx43之间的相互作用抑制了坏死性SH-SY5Y细胞中Cx43的K48连接的泛素化。VHL是Cx43的E3泛素连接酶,MLKL与VHL竞争与Cx43结合。MLKL Ser454与Cx43的相互作用可触发Cx43半通道的开放,导致细胞内Ca2+增加和细胞坏死。这项在动物模型、细胞和分子水平上的创新研究有望阐明MLKL和Cx43在局灶性皮质梗死后丘脑损伤中的作用。我们的发现可能有助于确定皮层梗死后神经恢复的新靶点。
{"title":"MLKL regulates Cx43 ubiquitinational degradation and mediates neuronal necroptosis in ipsilateral thalamus after focal cortical infarction.","authors":"Yanyan Tang,&nbsp;Quanhong Chu,&nbsp;Guanfeng Xie,&nbsp;Yafu Tan,&nbsp;Ziming Ye,&nbsp;Chao Qin","doi":"10.1186/s13041-023-01064-4","DOIUrl":"10.1186/s13041-023-01064-4","url":null,"abstract":"<p><p>Necroptosis is known to play an important role in the pathophysiology of cerebral ischemia; however, its role in the occurrence of secondary thalamic injury after focal cerebral infarction and the mechanism about how mixed lineage kinase domain-like (MLKL) executes necroptosis in this pathophysiology are still unclear. In this study, Sprague-Dawley rats were subjected to distal branch of middle cerebral artery occlusion (dMCAO). The expression of MLKL, connexin 43 (Cx43) and Von Hippel-Lindau (VHL) in vitro and in vivo were assessed by Western blot. Bioinformatic methods were used to predict the potential binding sites where MLKL interacted with Cx43, and the ubiquitination degradation of Cx43 regulated by VHL. The interactions among MLKL, Cx43, VHL, and Ubiquitin were assessed by immunoprecipitation. Dye uptake assay were used to examine the Cx43 hemichannels. Intracellular Ca<sup>2+</sup> concentration was measured using Fluo-4 AM. Overexpression and site-directed mutagenesis studies were used to study the mechanisms by which MLKL regulates Cx43 ubiquitinational degradation to mediate neuronal necroptosis. We found that MLKL and Cx43 were upregulated in the ventral posterolateral nucleus (VPN) of the ipsilateral thalamus after dMCAO. In the in vitro experiments MLKL and Cx43 were upregulated after TSZ-mediated necroptosis in SH-SY5Y cells. The interaction between MLKL and Cx43 inhibited the K48-linked ubiquitination of Cx43 in necroptotic SH-SY5Y cells. VHL is an E3 ubiquitin ligase for Cx43, and MLKL competes with VHL for binding to Cx43. Interaction of MLKL Ser454 with Cx43 can trigger the opening of Cx43 hemichannels, causing increased intracellular Ca<sup>2+</sup>, and cell necroptosis. This innovative study at animal models, cellular, and molecular levels is anticipated to clarify the roles of MLKL and Cx43 in thalamic damage after focal cortical infarction. Our findings may help identify novel targets for neurological recovery after cortical infarction.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10617209/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71413161","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
α-Synuclein propagation leads to synaptic abnormalities in the cortex through microglial synapse phagocytosis. α-突触核蛋白的传播通过小胶质细胞突触吞噬作用导致皮层突触异常。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-10-17 DOI: 10.1186/s13041-023-01059-1
Dayana Pérez-Acuña, Soo Jean Shin, Ka Hyun Rhee, Sang Jeong Kim, Seung-Jae Lee

The major neuropathologic feature of Parkinson's disease is the presence of widespread intracellular inclusions of α-synuclein known as Lewy bodies. Evidence suggests that these misfolded protein inclusions spread through the brain with disease progression. Changes in synaptic function precede neurodegeneration, and this extracellular α-synuclein can affect synaptic transmission. However, whether and how the spreading of α-synuclein aggregates modulates synaptic function before neuronal loss remains unknown. In the present study, we investigated the effect of intrastriatal injection of α-synuclein preformed fibrils (PFFs) on synaptic activity in the somatosensory cortex using a combination of whole-cell patch-clamp electrophysiology, histology, and Golgi-Cox staining. Intrastriatal PFF injection was followed by formation of phosphorylated α-synuclein inclusions in layer 5 of the somatosensory cortex, leading to a decrease in synapse density, dendritic spines, and spontaneous excitatory post-synaptic currents, without apparent neuronal loss. Additionally, three-dimensional reconstruction of microglia using confocal imaging showed an increase in the engulfment of synapses. Collectively, our data indicate that propagation of α-synuclein through neural networks causes abnormalities in synaptic structure and dynamics prior to neuronal loss.

帕金森病的主要神经病理学特征是存在广泛的细胞内α-突触核蛋白包涵体,称为路易体。有证据表明,这些错误折叠的蛋白质内含物随着疾病的进展而在大脑中传播。突触功能的变化先于神经退行性变,这种细胞外α-突触核蛋白可以影响突触传递。然而,在神经元丢失之前,α-突触核蛋白聚集体的传播是否以及如何调节突触功能仍然未知。在本研究中,我们采用全细胞膜片钳电生理学、组织学和高尔基-考克斯染色相结合的方法,研究了蛛网膜下腔注射α-突触核蛋白预制纤维(PFFs)对体感皮层突触活动的影响。纹状体内注射PFF后,在体感皮层第5层形成磷酸化的α-突触核蛋白内含物,导致突触密度、树突棘和自发兴奋性突触后电流降低,没有明显的神经元损失。此外,使用共焦成像对小胶质细胞进行三维重建显示突触的吞噬增加。总之,我们的数据表明,α-突触核蛋白通过神经网络的传播会在神经元丢失之前导致突触结构和动力学的异常。
{"title":"α-Synuclein propagation leads to synaptic abnormalities in the cortex through microglial synapse phagocytosis.","authors":"Dayana Pérez-Acuña, Soo Jean Shin, Ka Hyun Rhee, Sang Jeong Kim, Seung-Jae Lee","doi":"10.1186/s13041-023-01059-1","DOIUrl":"10.1186/s13041-023-01059-1","url":null,"abstract":"<p><p>The major neuropathologic feature of Parkinson's disease is the presence of widespread intracellular inclusions of α-synuclein known as Lewy bodies. Evidence suggests that these misfolded protein inclusions spread through the brain with disease progression. Changes in synaptic function precede neurodegeneration, and this extracellular α-synuclein can affect synaptic transmission. However, whether and how the spreading of α-synuclein aggregates modulates synaptic function before neuronal loss remains unknown. In the present study, we investigated the effect of intrastriatal injection of α-synuclein preformed fibrils (PFFs) on synaptic activity in the somatosensory cortex using a combination of whole-cell patch-clamp electrophysiology, histology, and Golgi-Cox staining. Intrastriatal PFF injection was followed by formation of phosphorylated α-synuclein inclusions in layer 5 of the somatosensory cortex, leading to a decrease in synapse density, dendritic spines, and spontaneous excitatory post-synaptic currents, without apparent neuronal loss. Additionally, three-dimensional reconstruction of microglia using confocal imaging showed an increase in the engulfment of synapses. Collectively, our data indicate that propagation of α-synuclein through neural networks causes abnormalities in synaptic structure and dynamics prior to neuronal loss.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10580656/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41236826","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
AAV-compatible optogenetic tools for activating endogenous calcium channels in vivo. 用于激活体内内源性钙通道的AAV兼容光遗传学工具。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-10-17 DOI: 10.1186/s13041-023-01061-7
Yeon Hee Kook, Hyoin Lee, Jinsu Lee, Yeonji Jeong, Jaerang Rho, Won Do Heo, Sangkyu Lee

Calcium ions (Ca2+) play pivotal roles in regulating diverse brain functions, including cognition, emotion, locomotion, and learning and memory. These functions are intricately regulated by a variety of Ca2+-dependent cellular processes, encompassing synaptic plasticity, neuro/gliotransmitter release, and gene expression. In our previous work, we developed 'monster OptoSTIM1' (monSTIM1), an improved OptoSTIM1 that selectively activates Ca2+-release-activated Ca2+ (CRAC) channels in the plasma membrane through blue light, allowing precise control over intracellular Ca2+ signaling and specific brain functions. However, the large size of the coding sequence of monSTIM1 poses a limitation for its widespread use, as it exceeds the packaging capacity of adeno-associated virus (AAV). To address this constraint, we have introduced monSTIM1 variants with reduced coding sequence sizes and established AAV-based systems for expressing them in neurons and glial cells in the mouse brain. Upon expression by AAVs, these monSTIM1 variants significantly increased the expression levels of cFos in neurons and astrocytes in the hippocampal CA1 region following non-invasive light illumination. The use of monSTIM1 variants offers a promising avenue for investigating the spatiotemporal roles of Ca2+-mediated cellular activities in various brain functions. Furthermore, this toolkit holds potential as a therapeutic strategy for addressing brain disorders associated with aberrant Ca2+ signaling.

钙离子(Ca2+)在调节不同的大脑功能中发挥着关键作用,包括认知、情绪、运动以及学习和记忆。这些功能受到各种Ca2+依赖性细胞过程的复杂调节,包括突触可塑性、神经/胶质递质释放和基因表达。在我们之前的工作中,我们开发了“monster OptoSTIM1”(monSTIM1),这是一种改进的OptosTM1,可以通过蓝光选择性激活质膜中的Ca2+释放激活的Ca2+(CRAC)通道,从而精确控制细胞内Ca2+信号和特定的脑功能。然而,monSTIM1编码序列的大尺寸限制了其广泛使用,因为它超过了腺相关病毒(AAV)的包装能力。为了解决这一限制,我们引入了编码序列大小减小的monSTIM1变体,并建立了基于AAV的系统,用于在小鼠大脑的神经元和神经胶质细胞中表达它们。在AAVs表达后,这些monSTIM1变体在无创光照后显著增加了海马CA1区神经元和星形胶质细胞中cFos的表达水平。monSTIM1变体的使用为研究Ca2+介导的细胞活动在各种脑功能中的时空作用提供了一条有前景的途径。此外,该工具包具有解决与异常Ca2+信号相关的大脑疾病的治疗策略的潜力。
{"title":"AAV-compatible optogenetic tools for activating endogenous calcium channels in vivo.","authors":"Yeon Hee Kook, Hyoin Lee, Jinsu Lee, Yeonji Jeong, Jaerang Rho, Won Do Heo, Sangkyu Lee","doi":"10.1186/s13041-023-01061-7","DOIUrl":"10.1186/s13041-023-01061-7","url":null,"abstract":"<p><p>Calcium ions (Ca<sup>2+</sup>) play pivotal roles in regulating diverse brain functions, including cognition, emotion, locomotion, and learning and memory. These functions are intricately regulated by a variety of Ca<sup>2+</sup>-dependent cellular processes, encompassing synaptic plasticity, neuro/gliotransmitter release, and gene expression. In our previous work, we developed 'monster OptoSTIM1' (monSTIM1), an improved OptoSTIM1 that selectively activates Ca<sup>2+</sup>-release-activated Ca<sup>2+</sup> (CRAC) channels in the plasma membrane through blue light, allowing precise control over intracellular Ca<sup>2+</sup> signaling and specific brain functions. However, the large size of the coding sequence of monSTIM1 poses a limitation for its widespread use, as it exceeds the packaging capacity of adeno-associated virus (AAV). To address this constraint, we have introduced monSTIM1 variants with reduced coding sequence sizes and established AAV-based systems for expressing them in neurons and glial cells in the mouse brain. Upon expression by AAVs, these monSTIM1 variants significantly increased the expression levels of cFos in neurons and astrocytes in the hippocampal CA1 region following non-invasive light illumination. The use of monSTIM1 variants offers a promising avenue for investigating the spatiotemporal roles of Ca<sup>2+</sup>-mediated cellular activities in various brain functions. Furthermore, this toolkit holds potential as a therapeutic strategy for addressing brain disorders associated with aberrant Ca<sup>2+</sup> signaling.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10583393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41236825","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
Anterior cingulate cortex regulates pain catastrophizing-like behaviors in rats. 大鼠前扣带皮层调节类似疼痛灾难的行为。
IF 3.6 3区 医学 Q2 Neuroscience Pub Date : 2023-10-13 DOI: 10.1186/s13041-023-01060-8
Hyun Jung Jee, Elaine Zhu, Mengqi Sun, Weizhuo Liu, Qiaosheng Zhang, Jing Wang

Negative pain expectation including pain catastrophizing is a well-known clinical phenomenon whereby patients amplify the aversive value of a painful or oftentimes even a similar, non-painful stimulus. Mechanisms of pain catastrophizing, however, remain elusive. Here, we modeled pain catastrophizing behavior in rats, and found that rats subjected to repeated noxious pin pricks on one paw demonstrated an aversive response to similar but non-noxious mechanical stimuli delivered to the contralateral paw. Optogenetic inhibition of pyramidal neuron activity in the anterior cingulate cortex (ACC) during the application of repetitive noxious pin pricks eliminated this catastrophizing behavior. Time-lapse calcium (Ca2+) imaging in the ACC further revealed an increase in spontaneous neural activity after the delivery of noxious stimuli. Together these results suggest that the experience of repeated noxious stimuli may drive hyperactivity in the ACC, causing increased avoidance of subthreshold stimuli, and that reducing this hyperactivity may play a role in treating pain catastrophizing.

包括疼痛灾难性在内的负面疼痛预期是一种众所周知的临床现象,患者会放大疼痛刺激的厌恶值,有时甚至是类似的非疼痛刺激。然而,疼痛造成灾难的机制仍然难以捉摸。在这里,我们模拟了大鼠的疼痛灾难性行为,并发现在一只爪子上反复受到有害针刺的大鼠对传递给对侧爪子的类似但无害的机械刺激表现出厌恶反应。在重复使用有毒针扎的过程中,对前扣带皮层(ACC)锥体神经元活动的光遗传学抑制消除了这种灾难性行为。ACC中的延时钙(Ca2+)成像进一步揭示了在递送有害刺激后自发神经活动的增加。总之,这些结果表明,重复的伤害性刺激可能会导致ACC过度活跃,导致对阈下刺激的回避增加,减少这种过度活跃可能在治疗疼痛灾难中发挥作用。
{"title":"Anterior cingulate cortex regulates pain catastrophizing-like behaviors in rats.","authors":"Hyun Jung Jee, Elaine Zhu, Mengqi Sun, Weizhuo Liu, Qiaosheng Zhang, Jing Wang","doi":"10.1186/s13041-023-01060-8","DOIUrl":"10.1186/s13041-023-01060-8","url":null,"abstract":"<p><p>Negative pain expectation including pain catastrophizing is a well-known clinical phenomenon whereby patients amplify the aversive value of a painful or oftentimes even a similar, non-painful stimulus. Mechanisms of pain catastrophizing, however, remain elusive. Here, we modeled pain catastrophizing behavior in rats, and found that rats subjected to repeated noxious pin pricks on one paw demonstrated an aversive response to similar but non-noxious mechanical stimuli delivered to the contralateral paw. Optogenetic inhibition of pyramidal neuron activity in the anterior cingulate cortex (ACC) during the application of repetitive noxious pin pricks eliminated this catastrophizing behavior. Time-lapse calcium (Ca<sup>2+</sup>) imaging in the ACC further revealed an increase in spontaneous neural activity after the delivery of noxious stimuli. Together these results suggest that the experience of repeated noxious stimuli may drive hyperactivity in the ACC, causing increased avoidance of subthreshold stimuli, and that reducing this hyperactivity may play a role in treating pain catastrophizing.</p>","PeriodicalId":18851,"journal":{"name":"Molecular Brain","volume":null,"pages":null},"PeriodicalIF":3.6,"publicationDate":"2023-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10576271/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41205591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Brain
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