Molecular Brain最新文献

筛选
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
Molecular Brain 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
{"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
Molecular Brain 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
{"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
Molecular Brain Pub Date : 2023-11-14 DOI: 10.1186/s13041-023-01065-3
Jun Yokose, Naoki Yamamoto, Sachie K Ogawa, Takashi Kitamura
{"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
Molecular Brain Pub Date : 2023-11-10 DOI: 10.1186/s13041-023-01067-1
Sanaz Ansarifar, Gabija Andreikė, Milad Nazari, Rodrigo Labouriau, Sadegh Nabavi, Andrea Moreno
{"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
Molecular Brain 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
{"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
Molecular Brain 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
{"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
Molecular Brain Pub Date : 2023-10-30 DOI: 10.1186/s13041-023-01064-4
Yanyan Tang, Quanhong Chu, Guanfeng Xie, Yafu Tan, Ziming Ye, Chao Qin
{"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
Molecular Brain 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
{"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
Molecular Brain 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
{"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
Molecular Brain 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
{"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信