首页 > 最新文献

Fluids and Barriers of the CNS最新文献

英文 中文
Endothelial EGLN3-PKM2 signaling induces the formation of acute astrocytic barrier to alleviate immune cell infiltration after subarachnoid hemorrhage. 内皮 EGLN3-PKM2 信号诱导急性星形胶质细胞屏障的形成,以减轻蛛网膜下腔出血后的免疫细胞浸润。
IF 5.9 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-05-16 DOI: 10.1186/s12987-024-00550-8
Mingxu Duan, Xufang Ru, Jiru Zhou, Yuanshu Li, Peiwen Guo, Wenbo Kang, Wenyan Li, Zhi Chen, Hua Feng, Yujie Chen

Background: Most subarachnoid hemorrhage (SAH) patients have no obvious hematoma lesions but exhibit blood-brain barrier dysfunction and vasogenic brain edema. However, there is a few days between blood‒brain barrier dysfunction and vasogenic brain edema. The present study sought to investigate whether this phenomenon is caused by endothelial injury induced by the acute astrocytic barrier, also known as the glial limitans.

Methods: Bioinformatics analyses of human endothelial cells and astrocytes under hypoxia were performed based on the GEO database. Wild-type, EGLN3 and PKM2 conditional knock-in mice were used to confirm glial limitan formation after SAH. Then, the effect of endothelial EGLN3-PKM2 signaling on temporal and spatial changes in glial limitans was evaluated in both in vivo and in vitro models of SAH.

Results: The data indicate that in the acute phase after SAH, astrocytes can form a temporary protective barrier, the glia limitans, around blood vessels that helps maintain barrier function and improve neurological prognosis. Molecular docking studies have shown that endothelial cells and astrocytes can promote glial limitans-based protection against early brain injury through EGLN3/PKM2 signaling and further activation of the PKC/ERK/MAPK signaling pathway in astrocytes after SAH.

Conclusion: Improving the ability to maintain glial limitans may be a new therapeutic strategy for improving the prognosis of SAH patients.

背景:大多数蛛网膜下腔出血(SAH)患者没有明显的血肿病变,但表现出血脑屏障功能障碍和血管源性脑水肿。然而,血脑屏障功能障碍与血管源性脑水肿之间相隔数天。本研究试图探究这一现象是否是由急性星形胶质屏障(又称胶质极限屏障)诱导的内皮损伤引起的:方法:基于 GEO 数据库对缺氧条件下的人类内皮细胞和星形胶质细胞进行生物信息学分析。用野生型、EGLN3和PKM2条件性基因敲入小鼠来证实SAH后神经胶质局限的形成。然后,在 SAH 体内和体外模型中评估了内皮 EGLN3-PKM2 信号传导对神经胶质局限的时间和空间变化的影响:结果:数据表明,在 SAH 后的急性期,星形胶质细胞可在血管周围形成一个临时的保护屏障--神经胶质局限,这有助于维持屏障功能和改善神经系统的预后。分子对接研究表明,内皮细胞和星形胶质细胞可通过 EGLN3/PKM2 信号传导和 SAH 后星形胶质细胞 PKC/ERK/MAPK 信号通路的进一步激活,促进基于神经胶质屏障的早期脑损伤保护:结论:提高维持神经胶质界限的能力可能是改善 SAH 患者预后的一种新的治疗策略。
{"title":"Endothelial EGLN3-PKM2 signaling induces the formation of acute astrocytic barrier to alleviate immune cell infiltration after subarachnoid hemorrhage.","authors":"Mingxu Duan, Xufang Ru, Jiru Zhou, Yuanshu Li, Peiwen Guo, Wenbo Kang, Wenyan Li, Zhi Chen, Hua Feng, Yujie Chen","doi":"10.1186/s12987-024-00550-8","DOIUrl":"10.1186/s12987-024-00550-8","url":null,"abstract":"<p><strong>Background: </strong>Most subarachnoid hemorrhage (SAH) patients have no obvious hematoma lesions but exhibit blood-brain barrier dysfunction and vasogenic brain edema. However, there is a few days between blood‒brain barrier dysfunction and vasogenic brain edema. The present study sought to investigate whether this phenomenon is caused by endothelial injury induced by the acute astrocytic barrier, also known as the glial limitans.</p><p><strong>Methods: </strong>Bioinformatics analyses of human endothelial cells and astrocytes under hypoxia were performed based on the GEO database. Wild-type, EGLN3 and PKM2 conditional knock-in mice were used to confirm glial limitan formation after SAH. Then, the effect of endothelial EGLN3-PKM2 signaling on temporal and spatial changes in glial limitans was evaluated in both in vivo and in vitro models of SAH.</p><p><strong>Results: </strong>The data indicate that in the acute phase after SAH, astrocytes can form a temporary protective barrier, the glia limitans, around blood vessels that helps maintain barrier function and improve neurological prognosis. Molecular docking studies have shown that endothelial cells and astrocytes can promote glial limitans-based protection against early brain injury through EGLN3/PKM2 signaling and further activation of the PKC/ERK/MAPK signaling pathway in astrocytes after SAH.</p><p><strong>Conclusion: </strong>Improving the ability to maintain glial limitans may be a new therapeutic strategy for improving the prognosis of SAH patients.</p>","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"21 1","pages":"42"},"PeriodicalIF":5.9,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11100217/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140957191","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Exposure to hyperbaric O2 levels leads to blood-brain barrier breakdown in rodents. 暴露于高压氧水平会导致啮齿动物的血脑屏障破裂。
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-05-16 DOI: 10.1186/s12987-024-00543-7
Yehuda M Danino, Ricarina Rabinovitz, Inbar Kirshenboim, Eilam Palzur, Chaim G Pick, Itamar Ish-Shalom, Yana Golovkin, Yehuda Arieli

Introduction: Hyperbaric oxygen has been used as a medical treatment tool in hyperbaric chambers and is an integral part of professional and combat divers' activity. In extreme cases, exposure to hyperbaric oxygen can develop central nervous system oxygen toxicity (CNS-OT), which leads to seizures and eventually death. CNS-OT is caused by neuronal hyperactivity due to high oxygen levels, potentially damaging brain cells including the blood-brain barrier (BBB). However, the effect of hyperbaric oxygen levels on the healthy BBB has not been characterized directly yet.

Methods: Six or three different groups of ~ eight rats or mice, respectively, were exposed to increasing levels of partial pressure of oxygen (0.21 to 5 ATA) in a hyperbaric chamber, followed by MRI scanning with gadolinium. Statistical significance (adjusted p-value ≤ 0.05) was assessed using linear regression and ordinary one-way (rats) or two-way (mice) ANOVA with correction of multiple comparison tests. In rats, the effect of 100% oxygen at 5 ATA was independently validated using FITC-Dextran (5 kDa). Statistical significance (p-value ≤ 0.05) was assessed using Welch's t-test and effect size was calculated by Cohen's D.

Results: In rats, analyzed MRI scans showed a significant trend of increase in the % gadolinium in brain tissues as a result of hyperbaric oxygen pressures (p-value = 0.0079). The most significant increase was measured at 4 ATA compared to air (adjusted p-value = 0.0461). Significant increased FITC-Dextran levels were measured in the rats' brains under 100% oxygen at 5 ATA versus air (p-value = 0.0327; Effect size = 2.0). In mice, a significant increase in gadolinium penetration into the hippocampus and frontal cortex was measured over time (adjusted p-value < 0.05) under 100% oxygen at 3 and 5 ATA versus air, and between the treatments (adjusted p-value < 0.0001).

Conclusions: The BBB is increasingly disrupted due to higher levels of hyperbaric oxygen in rodents, indicating a direct relation between hyperbaric oxygen and BBB dysregulation for the first time. We suggest considering this risk in different diving activities, and protocols using a hyperbaric chamber. On the other hand, this study highlights the potential therapeutic usage of hyperbaric oxygen for controlled drug delivery through the BBB into brain tissues in different brain-related diseases.

简介高压氧一直是高压氧舱中的医疗工具,也是专业潜水员和战斗潜水员活动中不可或缺的一部分。在极端情况下,接触高压氧会导致中枢神经系统氧中毒(CNS-OT),从而导致癫痫发作并最终死亡。中枢神经系统氧中毒是由于高浓度氧导致神经元过度活跃,从而可能损害脑细胞,包括血脑屏障(BBB)。然而,高压氧水平对健康 BBB 的影响尚未得到直接描述:方法:分别将六组或三组约八只大鼠或小鼠暴露于高压氧舱中不断升高的氧分压水平(0.21 至 5 ATA)中,然后用钆进行磁共振成像扫描。使用线性回归和普通单向(大鼠)或双向(小鼠)方差分析以及多重比较校正测试评估统计意义(调整后的 p 值≤ 0.05)。在大鼠中,使用 FITC-Dextran(5 kDa)独立验证了 5 ATA 时 100% 氧气的效果。统计显著性(p 值≤ 0.05)采用 Welch's t 检验,效应大小采用 Cohen's D 计算:对大鼠进行的磁共振成像扫描分析表明,高压氧导致脑组织中钆的百分比呈显著增加趋势(p 值 = 0.0079)。与空气相比,4 ATA 时的增加最为明显(调整后的 p 值 = 0.0461)。在 5 ATA 100%氧气条件下,与空气相比,大鼠大脑中的 FITC-二聚体水平显著增加(p 值 = 0.0327;效应大小 = 2.0)。在小鼠中,随着时间的推移,钆渗透到海马和额叶皮层的程度明显增加(调整后的 p 值结论):在啮齿类动物中,高压氧水平越高,生物BB越容易受到破坏,这首次表明高压氧与生物BB失调之间存在直接关系。我们建议在不同的潜水活动和使用高压氧舱的方案中考虑这一风险。另一方面,这项研究强调了高压氧在不同脑相关疾病中通过 BBB 向脑组织控制药物输送的潜在治疗用途。
{"title":"Exposure to hyperbaric O<sub>2</sub> levels leads to blood-brain barrier breakdown in rodents.","authors":"Yehuda M Danino, Ricarina Rabinovitz, Inbar Kirshenboim, Eilam Palzur, Chaim G Pick, Itamar Ish-Shalom, Yana Golovkin, Yehuda Arieli","doi":"10.1186/s12987-024-00543-7","DOIUrl":"https://doi.org/10.1186/s12987-024-00543-7","url":null,"abstract":"<p><strong>Introduction: </strong>Hyperbaric oxygen has been used as a medical treatment tool in hyperbaric chambers and is an integral part of professional and combat divers' activity. In extreme cases, exposure to hyperbaric oxygen can develop central nervous system oxygen toxicity (CNS-OT), which leads to seizures and eventually death. CNS-OT is caused by neuronal hyperactivity due to high oxygen levels, potentially damaging brain cells including the blood-brain barrier (BBB). However, the effect of hyperbaric oxygen levels on the healthy BBB has not been characterized directly yet.</p><p><strong>Methods: </strong>Six or three different groups of ~ eight rats or mice, respectively, were exposed to increasing levels of partial pressure of oxygen (0.21 to 5 ATA) in a hyperbaric chamber, followed by MRI scanning with gadolinium. Statistical significance (adjusted p-value ≤ 0.05) was assessed using linear regression and ordinary one-way (rats) or two-way (mice) ANOVA with correction of multiple comparison tests. In rats, the effect of 100% oxygen at 5 ATA was independently validated using FITC-Dextran (5 kDa). Statistical significance (p-value ≤ 0.05) was assessed using Welch's t-test and effect size was calculated by Cohen's D.</p><p><strong>Results: </strong>In rats, analyzed MRI scans showed a significant trend of increase in the % gadolinium in brain tissues as a result of hyperbaric oxygen pressures (p-value = 0.0079). The most significant increase was measured at 4 ATA compared to air (adjusted p-value = 0.0461). Significant increased FITC-Dextran levels were measured in the rats' brains under 100% oxygen at 5 ATA versus air (p-value = 0.0327; Effect size = 2.0). In mice, a significant increase in gadolinium penetration into the hippocampus and frontal cortex was measured over time (adjusted p-value < 0.05) under 100% oxygen at 3 and 5 ATA versus air, and between the treatments (adjusted p-value < 0.0001).</p><p><strong>Conclusions: </strong>The BBB is increasingly disrupted due to higher levels of hyperbaric oxygen in rodents, indicating a direct relation between hyperbaric oxygen and BBB dysregulation for the first time. We suggest considering this risk in different diving activities, and protocols using a hyperbaric chamber. On the other hand, this study highlights the potential therapeutic usage of hyperbaric oxygen for controlled drug delivery through the BBB into brain tissues in different brain-related diseases.</p>","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"21 1","pages":"41"},"PeriodicalIF":7.3,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11097412/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140957194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Reduced cerebrospinal fluid motion in patients with Parkinson's disease revealed by magnetic resonance imaging with low b-value diffusion weighted imaging. 低 b 值弥散加权成像磁共振成像显示帕金森病患者脑脊液运动减少。
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-05-09 DOI: 10.1186/s12987-024-00542-8
Gabriela Pierobon Mays, Kilian Hett, Jarrod Eisma, Colin D McKnight, Jason Elenberger, Alexander K Song, Ciaran Considine, Wesley T Richerson, Caleb Han, Adam Stark, Daniel O Claassen, Manus J Donahue

Background: Parkinson's disease is characterized by dopamine-responsive symptoms as well as aggregation of α-synuclein protofibrils. New diagnostic methods assess α-synuclein aggregation characteristics from cerebrospinal fluid (CSF) and recent pathophysiologic mechanisms suggest that CSF circulation disruptions may precipitate α-synuclein retention. Here, diffusion-weighted MRI with low-to-intermediate diffusion-weightings was applied to test the hypothesis that CSF motion is reduced in Parkinson's disease relative to healthy participants.

Methods: Multi-shell diffusion weighted MRI (spatial resolution = 1.8 × 1.8 × 4.0 mm) with low-to-intermediate diffusion weightings (b-values = 0, 50, 100, 200, 300, 700, and 1000 s/mm2) was applied over the approximate kinetic range of suprasellar cistern fluid motion at 3 Tesla in Parkinson's disease (n = 27; age = 66 ± 6.7 years) and non-Parkinson's control (n = 32; age = 68 ± 8.9 years) participants. Wilcoxon rank-sum tests were applied to test the primary hypothesis that the noise floor-corrected decay rate of CSF signal as a function of b-value, which reflects increasing fluid motion, is reduced within the suprasellar cistern of persons with versus without Parkinson's disease and inversely relates to choroid plexus activity assessed from perfusion-weighted MRI (significance-criteria: p < 0.05).

Results: Consistent with the primary hypothesis, CSF decay rates were higher in healthy (D = 0.00673 ± 0.00213 mm2/s) relative to Parkinson's disease (D = 0.00517 ± 0.00110 mm2/s) participants. This finding was preserved after controlling for age and sex and was observed in the posterior region of the suprasellar cistern (p < 0.001). An inverse correlation between choroid plexus perfusion and decay rate in the voxels within the suprasellar cistern (Spearman's-r=-0.312; p = 0.019) was observed.

Conclusions: Multi-shell diffusion MRI was applied to identify reduced CSF motion at the level of the suprasellar cistern in adults with versus without Parkinson's disease; the strengths and limitations of this methodology are discussed in the context of the growing literature on CSF flow.

背景:帕金森病的特征是多巴胺反应症状以及α-突触核蛋白原纤维的聚集。新的诊断方法可评估脑脊液(CSF)中α-突触核蛋白的聚集特征,而最新的病理生理机制表明,脑脊液循环障碍可能会导致α-突触核蛋白潴留。在此,我们采用中低度弥散加权核磁共振成像来检验帕金森病患者脑脊液运动相对于健康患者减少的假设:在帕金森病患者(n = 27;年龄 = 66 ± 6.7 岁)和非帕金森病对照组患者(n = 32;年龄 = 68 ± 8.9 岁)的近似鞘上贮液流运动范围内,在 3 特斯拉条件下应用多壳弥散加权 MRI(空间分辨率 = 1.8 × 1.8 × 4.0 mm)和中低弥散加权(b 值 = 0、50、100、200、300、700 和 1000 s/mm2)。应用 Wilcoxon 秩和检验来检验主要假设,即 CSF 信号的噪声底限校正衰减率与 b 值的函数关系(b 值反映了流体运动的增加)在帕金森病患者和非帕金森病患者的蝶鞍上贮水池内均有所降低,并且与灌注加权 MRI 评估的脉络丛活动成反比(显著性标准:p 结果:与主要假设一致,CSF 信号的噪声底限校正衰减率与 b 值的函数关系(b 值反映了流体运动的增加)在帕金森病患者和非帕金森病患者的蝶鞍上贮水池内均有所降低,并且与灌注加权 MRI 评估的脉络丛活动成反比:与主要假设一致,健康人(D = 0.00673 ± 0.00213 mm2/s)的 CSF 衰减率高于帕金森病患者(D = 0.00517 ± 0.00110 mm2/s)。在控制了年龄和性别后,这一结果仍保持不变,并在蝶窦上的后部区域观察到(P 结论):多壳弥散核磁共振成像可用于识别帕金森病成人患者与非帕金森病患者在蝶鞍上水平的脑脊液运动减弱情况;该方法的优势和局限性将在脑脊液流动文献不断增加的背景下进行讨论。
{"title":"Reduced cerebrospinal fluid motion in patients with Parkinson's disease revealed by magnetic resonance imaging with low b-value diffusion weighted imaging.","authors":"Gabriela Pierobon Mays, Kilian Hett, Jarrod Eisma, Colin D McKnight, Jason Elenberger, Alexander K Song, Ciaran Considine, Wesley T Richerson, Caleb Han, Adam Stark, Daniel O Claassen, Manus J Donahue","doi":"10.1186/s12987-024-00542-8","DOIUrl":"10.1186/s12987-024-00542-8","url":null,"abstract":"<p><strong>Background: </strong>Parkinson's disease is characterized by dopamine-responsive symptoms as well as aggregation of α-synuclein protofibrils. New diagnostic methods assess α-synuclein aggregation characteristics from cerebrospinal fluid (CSF) and recent pathophysiologic mechanisms suggest that CSF circulation disruptions may precipitate α-synuclein retention. Here, diffusion-weighted MRI with low-to-intermediate diffusion-weightings was applied to test the hypothesis that CSF motion is reduced in Parkinson's disease relative to healthy participants.</p><p><strong>Methods: </strong>Multi-shell diffusion weighted MRI (spatial resolution = 1.8 × 1.8 × 4.0 mm) with low-to-intermediate diffusion weightings (b-values = 0, 50, 100, 200, 300, 700, and 1000 s/mm<sup>2</sup>) was applied over the approximate kinetic range of suprasellar cistern fluid motion at 3 Tesla in Parkinson's disease (n = 27; age = 66 ± 6.7 years) and non-Parkinson's control (n = 32; age = 68 ± 8.9 years) participants. Wilcoxon rank-sum tests were applied to test the primary hypothesis that the noise floor-corrected decay rate of CSF signal as a function of b-value, which reflects increasing fluid motion, is reduced within the suprasellar cistern of persons with versus without Parkinson's disease and inversely relates to choroid plexus activity assessed from perfusion-weighted MRI (significance-criteria: p < 0.05).</p><p><strong>Results: </strong>Consistent with the primary hypothesis, CSF decay rates were higher in healthy (D = 0.00673 ± 0.00213 mm<sup>2</sup>/s) relative to Parkinson's disease (D = 0.00517 ± 0.00110 mm<sup>2</sup>/s) participants. This finding was preserved after controlling for age and sex and was observed in the posterior region of the suprasellar cistern (p < 0.001). An inverse correlation between choroid plexus perfusion and decay rate in the voxels within the suprasellar cistern (Spearman's-r=-0.312; p = 0.019) was observed.</p><p><strong>Conclusions: </strong>Multi-shell diffusion MRI was applied to identify reduced CSF motion at the level of the suprasellar cistern in adults with versus without Parkinson's disease; the strengths and limitations of this methodology are discussed in the context of the growing literature on CSF flow.</p>","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"21 1","pages":"40"},"PeriodicalIF":7.3,"publicationDate":"2024-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11080257/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140897898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The putative proton-coupled organic cation antiporter is involved in uptake of triptans into human brain capillary endothelial cells. 推定质子偶联有机阳离子反转运体参与了人脑毛细血管内皮细胞对三苯氧胺的吸收。
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-05-06 DOI: 10.1186/s12987-024-00544-6
Nana Svane, Alberte Bay Villekjær Pedersen, Anne Rodenberg, Burak Ozgür, Lasse Saaby, Christoffer Bundgaard, Mie Kristensen, Peer Tfelt-Hansen, Birger Brodin

Background: Triptans are anti-migraine drugs with a potential central site of action. However, it is not known to what extent triptans cross the blood-brain barrier (BBB). The aim of this study was therefore to determine if triptans pass the brain capillary endothelium and investigate the possible underlying mechanisms with focus on the involvement of the putative proton-coupled organic cation (H+/OC) antiporter. Additionally, we evaluated whether triptans interacted with the efflux transporter, P-glycoprotein (P-gp).

Methods: We investigated the cellular uptake characteristics of the prototypical H+/OC antiporter substrates, pyrilamine and oxycodone, and seven different triptans in the human brain microvascular endothelial cell line, hCMEC/D3. Triptan interactions with P-gp were studied using the IPEC-J2 MDR1 cell line. Lastly, in vivo neuropharmacokinetic assessment of the unbound brain-to-plasma disposition of eletriptan was conducted in wild type and mdr1a/1b knockout mice.

Results: We demonstrated that most triptans were able to inhibit uptake of the H+/OC antiporter substrate, pyrilamine, with eletriptan emerging as the strongest inhibitor. Eletriptan, almotriptan, and sumatriptan exhibited a pH-dependent uptake into hCMEC/D3 cells. Eletriptan demonstrated saturable uptake kinetics with an apparent Km of 89 ± 38 µM and a Jmax of 2.2 ± 0.7 nmol·min-1·mg protein-1 (n = 3). Bidirectional transport experiments across IPEC-J2 MDR1 monolayers showed that eletriptan is transported by P-gp, thus indicating that eletriptan is both a substrate of the H+/OC antiporter and P-gp. This was further confirmed in vivo, where the unbound brain-to-unbound plasma concentration ratio (Kp,uu) was 0.04 in wild type mice while the ratio rose to 1.32 in mdr1a/1b knockout mice.

Conclusions: We have demonstrated that the triptan family of compounds possesses affinity for the H+/OC antiporter proposing that the putative H+/OC antiporter plays a role in the BBB transport of triptans, particularly eletriptan. Our in vivo studies indicate that eletriptan is subjected to simultaneous brain uptake and efflux, possibly facilitated by the putative H+/OC antiporter and P-gp, respectively. Our findings offer novel insights into the potential central site of action involved in migraine treatment with triptans and highlight the significance of potential transporter related drug-drug interactions.

背景:曲坦类药物是一种抗偏头痛药物,具有潜在的中枢作用部位。然而,目前尚不清楚曲坦类药物能在多大程度上穿过血脑屏障(BBB)。因此,本研究旨在确定曲坦类药物是否能通过脑毛细血管内皮,并研究其可能的潜在机制,重点是质子偶联有机阳离子(H+/OC)反转运体的参与。此外,我们还评估了三苯氧胺是否与外排转运体 P 糖蛋白(P-gp)相互作用:方法:我们在人脑微血管内皮细胞系 hCMEC/D3 中研究了原型 H+/OC 反转运体底物吡咯烷酮和羟考酮以及七种不同的曲坦类药物的细胞摄取特性。使用 IPEC-J2 MDR1 细胞系研究了曲坦与 P-gp 的相互作用。最后,在野生型小鼠和 mdr1a/1b 基因敲除小鼠体内对依利普坦的非结合脑-血浆处置进行了神经药代动力学评估:结果:我们发现大多数曲坦类药物都能抑制H+/OC抗转运体底物吡咯胺的摄取,其中依来普坦是最强的抑制剂。依利普坦、阿莫曲坦和舒马曲坦在hCMEC/D3细胞中的摄取呈pH依赖性。依来普坦显示出饱和摄取动力学,表观 Km 为 89 ± 38 µM,Jmax 为 2.2 ± 0.7 nmol-min-1-mg 蛋白-1(n = 3)。在 IPEC-J2 MDR1 单层上的双向转运实验表明,依来普坦是通过 P-gp 转运的,因此表明依来普坦既是 H+/OC 反转运体的底物,也是 P-gp 的底物。这一点在体内得到了进一步证实,野生型小鼠的非结合脑浓度与非结合血浆浓度比值(Kp,uu)为 0.04,而 mdr1a/1b 基因敲除小鼠的这一比值则升至 1.32:结论:我们证明了三苯氧胺家族化合物对 H+/OC 反转运体具有亲和力,从而推定 H+/OC 反转运体在三苯氧胺(尤其是依利普坦)的 BBB 转运中发挥作用。我们的体内研究表明,依来普坦在大脑中会同时发生吸收和外流,可能分别由假定的 H+/OC 反转运体和 P-gp 促进。我们的研究结果为三苯氧胺类药物治疗偏头痛所涉及的潜在中枢作用部位提供了新的见解,并强调了与潜在转运体相关的药物间相互作用的重要性。
{"title":"The putative proton-coupled organic cation antiporter is involved in uptake of triptans into human brain capillary endothelial cells.","authors":"Nana Svane, Alberte Bay Villekjær Pedersen, Anne Rodenberg, Burak Ozgür, Lasse Saaby, Christoffer Bundgaard, Mie Kristensen, Peer Tfelt-Hansen, Birger Brodin","doi":"10.1186/s12987-024-00544-6","DOIUrl":"10.1186/s12987-024-00544-6","url":null,"abstract":"<p><strong>Background: </strong>Triptans are anti-migraine drugs with a potential central site of action. However, it is not known to what extent triptans cross the blood-brain barrier (BBB). The aim of this study was therefore to determine if triptans pass the brain capillary endothelium and investigate the possible underlying mechanisms with focus on the involvement of the putative proton-coupled organic cation (H<sup>+</sup>/OC) antiporter. Additionally, we evaluated whether triptans interacted with the efflux transporter, P-glycoprotein (P-gp).</p><p><strong>Methods: </strong>We investigated the cellular uptake characteristics of the prototypical H<sup>+</sup>/OC antiporter substrates, pyrilamine and oxycodone, and seven different triptans in the human brain microvascular endothelial cell line, hCMEC/D3. Triptan interactions with P-gp were studied using the IPEC-J2 MDR1 cell line. Lastly, in vivo neuropharmacokinetic assessment of the unbound brain-to-plasma disposition of eletriptan was conducted in wild type and mdr1a/1b knockout mice.</p><p><strong>Results: </strong>We demonstrated that most triptans were able to inhibit uptake of the H<sup>+</sup>/OC antiporter substrate, pyrilamine, with eletriptan emerging as the strongest inhibitor. Eletriptan, almotriptan, and sumatriptan exhibited a pH-dependent uptake into hCMEC/D3 cells. Eletriptan demonstrated saturable uptake kinetics with an apparent K<sub>m</sub> of 89 ± 38 µM and a J<sub>max</sub> of 2.2 ± 0.7 nmol·min<sup>-1</sup>·mg protein<sup>-1</sup> (n = 3). Bidirectional transport experiments across IPEC-J2 MDR1 monolayers showed that eletriptan is transported by P-gp, thus indicating that eletriptan is both a substrate of the H<sup>+</sup>/OC antiporter and P-gp. This was further confirmed in vivo, where the unbound brain-to-unbound plasma concentration ratio (K<sub>p,uu</sub>) was 0.04 in wild type mice while the ratio rose to 1.32 in mdr1a/1b knockout mice.</p><p><strong>Conclusions: </strong>We have demonstrated that the triptan family of compounds possesses affinity for the H<sup>+</sup>/OC antiporter proposing that the putative H<sup>+</sup>/OC antiporter plays a role in the BBB transport of triptans, particularly eletriptan. Our in vivo studies indicate that eletriptan is subjected to simultaneous brain uptake and efflux, possibly facilitated by the putative H<sup>+</sup>/OC antiporter and P-gp, respectively. Our findings offer novel insights into the potential central site of action involved in migraine treatment with triptans and highlight the significance of potential transporter related drug-drug interactions.</p>","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"21 1","pages":"39"},"PeriodicalIF":7.3,"publicationDate":"2024-05-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11071266/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140861845","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
A human pluripotent stem cell-derived in vitro model of the blood–brain barrier in cerebral malaria 源自人类多能干细胞的脑疟疾血脑屏障体外模型
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-05-01 DOI: 10.1186/s12987-024-00541-9
Adnan Gopinadhan, Jason M. Hughes, Andrea L. Conroy, Chandy C. John, Scott G. Canfield, Dibyadyuti Datta
Blood–brain barrier (BBB) disruption is a central feature of cerebral malaria (CM), a severe complication of Plasmodium falciparum (Pf) infections. In CM, sequestration of Pf-infected red blood cells (Pf-iRBCs) to brain endothelial cells combined with inflammation, hemolysis, microvasculature obstruction and endothelial dysfunction mediates BBB disruption, resulting in severe neurologic symptoms including coma and seizures, potentially leading to death or long-term sequelae. In vitro models have advanced our knowledge of CM-mediated BBB disruption, but their physiological relevance remains uncertain. Using human induced pluripotent stem cell-derived brain microvascular endothelial cells (hiPSC-BMECs), we aimed to develop a novel in vitro model of the BBB in CM, exhibiting enhanced barrier properties. hiPSC-BMECs were co-cultured with HB3var03 strain Pf-iRBCs up to 9 h. Barrier integrity was measured using transendothelial electrical resistance (TEER) and sodium fluorescein permeability assays. Localization and expression of tight junction (TJ) proteins (occludin, zonula occludens-1, claudin-5), cellular adhesion molecules (ICAM-1, VCAM-1), and endothelial surface markers (EPCR) were determined using immunofluorescence imaging (IF) and western blotting (WB). Expression of angiogenic and cell stress markers were measured using multiplex proteome profiler arrays. After 6-h of co-culture with Pf-iRBCs, hiPSC-BMECs showed reduced TEER and increased sodium fluorescein permeability compared to co-culture with uninfected RBCs, indicative of a leaky barrier. We observed disruptions in localization of occludin, zonula occludens-1, and claudin-5 by IF, but no change in protein expression by WB in Pf-iRBC co-cultures. Expression of ICAM-1 and VCAM-1 but not EPCR was elevated in hiPSC-BMECs with Pf-iRBC co-culture compared to uninfected RBC co-culture. In addition, there was an increase in expression of angiogenin, platelet factor-4, and phospho-heat shock protein-27 in the Pf-iRBCs co-culture compared to uninfected RBC co-culture. These findings demonstrate the validity of our hiPSC-BMECs based model of the BBB, that displays enhanced barrier integrity and appropriate TJ protein localization. In the hiPSC-BMEC co-culture with Pf-iRBCs, reduced TEER, increased paracellular permeability, changes in TJ protein localization, increase in expression of adhesion molecules, and markers of angiogenesis and cellular stress all point towards a novel model with enhanced barrier properties, suitable for investigating pathogenic mechanisms underlying BBB disruption in CM.
血脑屏障(BBB)破坏是恶性疟原虫(Pf)感染的严重并发症--脑疟疾(CM)的主要特征。在脑疟疾中,受 Pf 感染的红细胞(Pf-iRBCs)被固着在脑内皮细胞上,再加上炎症、溶血、微血管阻塞和内皮功能障碍,介导了 BBB 破坏,导致严重的神经症状,包括昏迷和癫痫发作,可能导致死亡或长期后遗症。体外模型增进了我们对 CM 介导的 BBB 破坏的了解,但其生理相关性仍不确定。我们利用人类诱导多能干细胞衍生的脑微血管内皮细胞(hiPSC-BMECs),旨在开发一种新型的 CM 中 BBB 体外模型,该模型具有增强的屏障特性。使用免疫荧光成像(IF)和免疫印迹(WB)测定了紧密连接(TJ)蛋白(occludin、zonula occludens-1、claudin-5)、细胞粘附分子(ICAM-1、VCAM-1)和内皮表面标志物(EPCR)的定位和表达。血管生成标记物和细胞应激标记物的表达采用多重蛋白质组分析仪阵列进行测定。与未感染的 RBCs 共同培养 6 小时后,hiPSC-BMECs 显示 TEER 降低,荧光素钠通透性增加,这表明屏障出现了渗漏。我们通过 IF 观察到,在 Pf-iRBC 共培养物中,闭锁素、闭锁带-1 和 Claudin-5 的定位受到破坏,但通过 WB 观察,蛋白表达没有变化。与未感染的 RBC 共培养相比,与 Pf-iRBC 共培养的 hiPSC-BMECs 中 ICAM-1 和 VCAM-1 的表达升高,但 EPCR 的表达没有升高。此外,与未感染的 RBC 共培养相比,Pf-iRBCs 共培养中血管生成素、血小板因子-4 和磷酸化热休克蛋白-27 的表达也有所增加。这些发现证明了我们基于 hiPSC-BMECs 的 BBB 模型的有效性,该模型显示出更强的屏障完整性和适当的 TJ 蛋白定位。在与 Pf-iRBCs 共同培养的 hiPSC-BMEC 中,TEER 减少、细胞旁通透性增加、TJ 蛋白定位发生变化、粘附分子表达增加以及血管生成和细胞应激标记物均表明这种新型模型具有增强的屏障特性,适合用于研究 CM 中 BBB 破坏的致病机制。
{"title":"A human pluripotent stem cell-derived in vitro model of the blood–brain barrier in cerebral malaria","authors":"Adnan Gopinadhan, Jason M. Hughes, Andrea L. Conroy, Chandy C. John, Scott G. Canfield, Dibyadyuti Datta","doi":"10.1186/s12987-024-00541-9","DOIUrl":"https://doi.org/10.1186/s12987-024-00541-9","url":null,"abstract":"Blood–brain barrier (BBB) disruption is a central feature of cerebral malaria (CM), a severe complication of Plasmodium falciparum (Pf) infections. In CM, sequestration of Pf-infected red blood cells (Pf-iRBCs) to brain endothelial cells combined with inflammation, hemolysis, microvasculature obstruction and endothelial dysfunction mediates BBB disruption, resulting in severe neurologic symptoms including coma and seizures, potentially leading to death or long-term sequelae. In vitro models have advanced our knowledge of CM-mediated BBB disruption, but their physiological relevance remains uncertain. Using human induced pluripotent stem cell-derived brain microvascular endothelial cells (hiPSC-BMECs), we aimed to develop a novel in vitro model of the BBB in CM, exhibiting enhanced barrier properties. hiPSC-BMECs were co-cultured with HB3var03 strain Pf-iRBCs up to 9 h. Barrier integrity was measured using transendothelial electrical resistance (TEER) and sodium fluorescein permeability assays. Localization and expression of tight junction (TJ) proteins (occludin, zonula occludens-1, claudin-5), cellular adhesion molecules (ICAM-1, VCAM-1), and endothelial surface markers (EPCR) were determined using immunofluorescence imaging (IF) and western blotting (WB). Expression of angiogenic and cell stress markers were measured using multiplex proteome profiler arrays. After 6-h of co-culture with Pf-iRBCs, hiPSC-BMECs showed reduced TEER and increased sodium fluorescein permeability compared to co-culture with uninfected RBCs, indicative of a leaky barrier. We observed disruptions in localization of occludin, zonula occludens-1, and claudin-5 by IF, but no change in protein expression by WB in Pf-iRBC co-cultures. Expression of ICAM-1 and VCAM-1 but not EPCR was elevated in hiPSC-BMECs with Pf-iRBC co-culture compared to uninfected RBC co-culture. In addition, there was an increase in expression of angiogenin, platelet factor-4, and phospho-heat shock protein-27 in the Pf-iRBCs co-culture compared to uninfected RBC co-culture. These findings demonstrate the validity of our hiPSC-BMECs based model of the BBB, that displays enhanced barrier integrity and appropriate TJ protein localization. In the hiPSC-BMEC co-culture with Pf-iRBCs, reduced TEER, increased paracellular permeability, changes in TJ protein localization, increase in expression of adhesion molecules, and markers of angiogenesis and cellular stress all point towards a novel model with enhanced barrier properties, suitable for investigating pathogenic mechanisms underlying BBB disruption in CM.","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"17 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140828673","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Choroid plexus enlargement in amyotrophic lateral sclerosis patients and its correlation with clinical disability and blood-CSF barrier permeability 肌萎缩侧索硬化症患者脉络丛扩大及其与临床残疾和血液-脑脊液屏障通透性的关系
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-04-17 DOI: 10.1186/s12987-024-00536-6
Tingjun Dai, Jianwei Lou, Deyuan Kong, Jinyu Li, Qingguo Ren, Yujing Chen, Sujuan Sun, Yan Yun, Xiaohan Sun, Yiru Yang, Kai Shao, Wei Li, Yuying Zhao, Xiangshui Meng, Chuanzhu Yan, Pengfei Lin, Shuangwu Liu
Using in vivo neuroimaging techniques, growing evidence has demonstrated that the choroid plexus (CP) volume is enlarged in patients with several neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. However, although animal and postmortem findings suggest that CP abnormalities are likely important pathological mechanisms underlying amyotrophic lateral sclerosis (ALS), the third most common neurodegenerative disease, no available study has been conducted to thoroughly assess CP abnormalities and their clinical relevance in vivo in ALS patients to date. Thus, we aimed to determine whether in vivo CP enlargement may occur in ALS patients. We also aimed to identify the relationships of CP volume with clinical disabilities and blood-CSF barrier (BCSFB) permeability in ALS patients. In this retrospective study, based on structural MRI data, CP volume was assessed using a Gaussian mixture model and underwent further manual correction in 155 ALS patients and 105 age- and sex-matched HCs from October 2021 to April 2023. The ALS Functional Rating Scale-Revised (ALSFRS-R) was used to assess clinical disability. The CSF/serum albumin quotient (Qalb) was used to assess BCSFB permeability. Moreover, all the ALS patients completed genetic testing, and according to genetic testing, the ALS patients were further divided into genetic ALS subgroup and sporadic ALS subgroup. We found that compared with HCs, ALS patients had a significantly higher CP volume (p < 0.001). Moreover, compared with HCs, CP volume was significantly increased in both ALS patients with and without known genetic mutations after family-wise error correction (p = 0.006 and p < 0.001, respectively), while there were no significant differences between the two ALS groups. Furthermore, the CP volume was significantly correlated with the ALSFRS-r score (r = -0.226; p = 0.005) and the Qalb (r = 0.479; p < 0.001) in ALS patients. Our study first demonstrates CP enlargement in vivo in ALS patients, and continues to suggest an important pathogenetic role for CP abnormalities in ALS. Moreover, assessing CP volume is likely a noninvasive and easy-to-implement approach for screening BCSFB dysfunction in ALS patients.
越来越多的证据表明,利用体内神经成像技术,脉络丛(CP)体积在包括阿尔茨海默病和帕金森病在内的多种神经退行性疾病患者中都会增大。然而,尽管动物和死后研究结果表明,脉络丛异常可能是肌萎缩性脊髓侧索硬化症(ALS)--第三大最常见的神经退行性疾病--的重要病理机制,但迄今为止还没有研究对 ALS 患者体内的脉络丛异常及其临床相关性进行彻底评估。因此,我们的目的是确定 ALS 患者体内是否会出现 CP 增大。我们还旨在确定 ALS 患者体内 CP 体积与临床残疾和血液-脑脊液屏障(BCSFB)通透性之间的关系。在这项回顾性研究中,基于结构性核磁共振成像数据,使用高斯混合模型评估了 CP 体积,并对 2021 年 10 月至 2023 年 4 月期间的 155 名 ALS 患者和 105 名年龄和性别匹配的 HC 进行了进一步的人工校正。ALS功能评定量表-修订版(ALSFRS-R)用于评估临床残疾情况。CSF/血清白蛋白商数(Qalb)用于评估BCSFB通透性。此外,所有 ALS 患者均完成了基因检测,并根据基因检测结果将 ALS 患者进一步分为遗传性 ALS 亚组和散发性 ALS 亚组。我们发现,与 HCs 相比,ALS 患者的 CP 容量明显更高(p < 0.001)。此外,与 HCs 相比,经家族性误差校正后,有已知基因突变和无已知基因突变的 ALS 患者的 CP 体积均明显增大(分别为 p = 0.006 和 p <0.001),而两个 ALS 组间无明显差异。此外,ALS 患者的 CP 体积与 ALSFRS-r 评分(r = -0.226;p = 0.005)和 Qalb(r = 0.479;p < 0.001)明显相关。我们的研究首次证明了 ALS 患者体内 CP 的增大,并继续表明 CP 异常在 ALS 中具有重要的致病作用。此外,评估 CP 体积可能是筛查 ALS 患者 BCSFB 功能障碍的一种非侵入性且易于实施的方法。
{"title":"Choroid plexus enlargement in amyotrophic lateral sclerosis patients and its correlation with clinical disability and blood-CSF barrier permeability","authors":"Tingjun Dai, Jianwei Lou, Deyuan Kong, Jinyu Li, Qingguo Ren, Yujing Chen, Sujuan Sun, Yan Yun, Xiaohan Sun, Yiru Yang, Kai Shao, Wei Li, Yuying Zhao, Xiangshui Meng, Chuanzhu Yan, Pengfei Lin, Shuangwu Liu","doi":"10.1186/s12987-024-00536-6","DOIUrl":"https://doi.org/10.1186/s12987-024-00536-6","url":null,"abstract":"Using in vivo neuroimaging techniques, growing evidence has demonstrated that the choroid plexus (CP) volume is enlarged in patients with several neurodegenerative diseases, including Alzheimer’s disease and Parkinson’s disease. However, although animal and postmortem findings suggest that CP abnormalities are likely important pathological mechanisms underlying amyotrophic lateral sclerosis (ALS), the third most common neurodegenerative disease, no available study has been conducted to thoroughly assess CP abnormalities and their clinical relevance in vivo in ALS patients to date. Thus, we aimed to determine whether in vivo CP enlargement may occur in ALS patients. We also aimed to identify the relationships of CP volume with clinical disabilities and blood-CSF barrier (BCSFB) permeability in ALS patients. In this retrospective study, based on structural MRI data, CP volume was assessed using a Gaussian mixture model and underwent further manual correction in 155 ALS patients and 105 age- and sex-matched HCs from October 2021 to April 2023. The ALS Functional Rating Scale-Revised (ALSFRS-R) was used to assess clinical disability. The CSF/serum albumin quotient (Qalb) was used to assess BCSFB permeability. Moreover, all the ALS patients completed genetic testing, and according to genetic testing, the ALS patients were further divided into genetic ALS subgroup and sporadic ALS subgroup. We found that compared with HCs, ALS patients had a significantly higher CP volume (p < 0.001). Moreover, compared with HCs, CP volume was significantly increased in both ALS patients with and without known genetic mutations after family-wise error correction (p = 0.006 and p < 0.001, respectively), while there were no significant differences between the two ALS groups. Furthermore, the CP volume was significantly correlated with the ALSFRS-r score (r = -0.226; p = 0.005) and the Qalb (r = 0.479; p < 0.001) in ALS patients. Our study first demonstrates CP enlargement in vivo in ALS patients, and continues to suggest an important pathogenetic role for CP abnormalities in ALS. Moreover, assessing CP volume is likely a noninvasive and easy-to-implement approach for screening BCSFB dysfunction in ALS patients.","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"100 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140612255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Analysis of ischemic stroke-mediated effects on blood–brain barrier properties along the arteriovenous axis assessed by intravital two-photon imaging 通过眼内双光子成像评估缺血性中风对动静脉轴血脑屏障特性的影响分析
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-04-15 DOI: 10.1186/s12987-024-00537-5
Jil Protzmann, Felix Jung, Lars Jakobsson, Linda Fredriksson
Early breach of the blood–brain barrier (BBB) and consequently extravasation of blood-borne substances into the brain parenchyma is a common hallmark of ischemic stroke. Although BBB breakdown is associated with an increased risk of cerebral hemorrhage and poor clinical prognosis, the cause and mechanism of this process are largely unknown. The aim of this study was to establish an imaging and analysis protocol which enables investigation of the dynamics of BBB breach in relation to hemodynamic properties along the arteriovenous axis. Using longitudinal intravital two-photon imaging following photothrombotic induction of ischemic stroke through a cranial window, we were able to study the response of the cerebral vasculature to ischemia, from the early critical hours to the days/weeks after the infarct. We demonstrate that disruption of the BBB and hemodynamic parameters, including perturbed blood flow, can be studied at single-vessel resolution in the three-dimensional space as early as 30 min after vessel occlusion. Further, we show that this protocol permits longitudinal studies on the response of individual blood vessels to ischemia over time, thus enabling detection of (maladaptive) vascular remodeling such as intussusception, angiogenic sprouting and entanglement of vessel networks. Taken together, this in vivo two-photon imaging and analysis protocol will be useful in future studies investigating the molecular and cellular mechanisms, and the spatial contribution, of BBB breach to disease progression which might ultimately aid the development of new and more precise treatment strategies for ischemic stroke.
血脑屏障(BBB)的早期破坏以及由此导致的血源性物质外渗进入脑实质是缺血性脑卒中的常见特征。虽然血脑屏障破裂与脑出血风险增加和临床预后不良有关,但这一过程的原因和机制在很大程度上仍不清楚。本研究的目的是建立一种成像和分析方案,以研究 BBB 破损的动态与沿动静脉轴的血流动力学特性的关系。在通过头颅开窗进行光栓诱导缺血性中风后,我们利用纵向体内双光子成像技术研究了脑血管对缺血的反应,包括从早期关键时刻到梗死后几天/几周的反应。我们证明,早在血管闭塞后 30 分钟,就可以在三维空间中以单血管分辨率研究 BBB 的破坏和血流动力学参数,包括紊乱的血流。此外,我们还展示了该方案允许对单个血管随时间变化对缺血的反应进行纵向研究,从而能够检测(不适应的)血管重塑,如血管内嵌、血管萌发和血管网络缠结。总之,这种活体双光子成像和分析方案将有助于今后研究 BBB 破损对疾病进展的分子和细胞机制以及空间贡献,最终有助于开发新的、更精确的缺血性中风治疗策略。
{"title":"Analysis of ischemic stroke-mediated effects on blood–brain barrier properties along the arteriovenous axis assessed by intravital two-photon imaging","authors":"Jil Protzmann, Felix Jung, Lars Jakobsson, Linda Fredriksson","doi":"10.1186/s12987-024-00537-5","DOIUrl":"https://doi.org/10.1186/s12987-024-00537-5","url":null,"abstract":"Early breach of the blood–brain barrier (BBB) and consequently extravasation of blood-borne substances into the brain parenchyma is a common hallmark of ischemic stroke. Although BBB breakdown is associated with an increased risk of cerebral hemorrhage and poor clinical prognosis, the cause and mechanism of this process are largely unknown. The aim of this study was to establish an imaging and analysis protocol which enables investigation of the dynamics of BBB breach in relation to hemodynamic properties along the arteriovenous axis. Using longitudinal intravital two-photon imaging following photothrombotic induction of ischemic stroke through a cranial window, we were able to study the response of the cerebral vasculature to ischemia, from the early critical hours to the days/weeks after the infarct. We demonstrate that disruption of the BBB and hemodynamic parameters, including perturbed blood flow, can be studied at single-vessel resolution in the three-dimensional space as early as 30 min after vessel occlusion. Further, we show that this protocol permits longitudinal studies on the response of individual blood vessels to ischemia over time, thus enabling detection of (maladaptive) vascular remodeling such as intussusception, angiogenic sprouting and entanglement of vessel networks. Taken together, this in vivo two-photon imaging and analysis protocol will be useful in future studies investigating the molecular and cellular mechanisms, and the spatial contribution, of BBB breach to disease progression which might ultimately aid the development of new and more precise treatment strategies for ischemic stroke.","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"52 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140573283","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
C9ORF72 patient-derived endothelial cells drive blood-brain barrier disruption and contribute to neurotoxicity C9ORF72 患者衍生内皮细胞导致血脑屏障破坏并引发神经毒性
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-04-11 DOI: 10.1186/s12987-024-00528-6
Ana Aragón-González, Allan C Shaw, Jannigje R Kok, Florence S Roussel, Cleide dos Santos Souza, Sarah M Granger, Tatyana Vetter, Yolanda de Diego, Kathrin C Meyer, Selina N Beal, Pamela J Shaw, Laura Ferraiuolo
The blood-brain barrier (BBB) serves as a highly intricate and dynamic interface connecting the brain and the bloodstream, playing a vital role in maintaining brain homeostasis. BBB dysfunction has been associated with multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS); however, the role of the BBB in neurodegeneration is understudied. We developed an ALS patient-derived model of the BBB by using cells derived from 5 patient donors carrying C9ORF72 mutations. Brain microvascular endothelial-like cells (BMEC-like cells) derived from C9ORF72-ALS patients showed altered gene expression, compromised barrier integrity, and increased P-glycoprotein transporter activity. In addition, mitochondrial metabolic tests demonstrated that C9ORF72-ALS BMECs display a significant decrease in basal glycolysis accompanied by increased basal and ATP-linked respiration. Moreover, our study reveals that C9-ALS derived astrocytes can further affect BMECs function and affect the expression of the glucose transporter Glut-1. Finally, C9ORF72 patient-derived BMECs form leaky barriers through a cell-autonomous mechanism and have neurotoxic properties towards motor neurons.
血脑屏障(BBB)是连接大脑和血液的一个高度复杂的动态界面,在维持大脑平衡方面发挥着至关重要的作用。血脑屏障功能障碍与包括肌萎缩性脊髓侧索硬化症(ALS)在内的多种神经退行性疾病有关;然而,人们对血脑屏障在神经退行性疾病中的作用研究不足。我们利用来自 5 位携带 C9ORF72 突变的患者供体的细胞,建立了 ALS 患者来源的 BBB 模型。来自 C9ORF72-ALS 患者的脑微血管内皮样细胞(BMEC-like cells)显示出基因表达改变、屏障完整性受损和 P 糖蛋白转运体活性增加。此外,线粒体代谢测试表明,C9ORF72-ALS BMECs 的基础糖酵解显著减少,同时基础呼吸和 ATP 链接呼吸增加。此外,我们的研究还发现,C9-ALS 衍生的星形胶质细胞会进一步影响 BMECs 的功能,并影响葡萄糖转运体 Glut-1 的表达。最后,C9ORF72 患者衍生的 BMECs 通过细胞自主机制形成漏屏障,并对运动神经元具有神经毒性。
{"title":"C9ORF72 patient-derived endothelial cells drive blood-brain barrier disruption and contribute to neurotoxicity","authors":"Ana Aragón-González, Allan C Shaw, Jannigje R Kok, Florence S Roussel, Cleide dos Santos Souza, Sarah M Granger, Tatyana Vetter, Yolanda de Diego, Kathrin C Meyer, Selina N Beal, Pamela J Shaw, Laura Ferraiuolo","doi":"10.1186/s12987-024-00528-6","DOIUrl":"https://doi.org/10.1186/s12987-024-00528-6","url":null,"abstract":"The blood-brain barrier (BBB) serves as a highly intricate and dynamic interface connecting the brain and the bloodstream, playing a vital role in maintaining brain homeostasis. BBB dysfunction has been associated with multiple neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS); however, the role of the BBB in neurodegeneration is understudied. We developed an ALS patient-derived model of the BBB by using cells derived from 5 patient donors carrying C9ORF72 mutations. Brain microvascular endothelial-like cells (BMEC-like cells) derived from C9ORF72-ALS patients showed altered gene expression, compromised barrier integrity, and increased P-glycoprotein transporter activity. In addition, mitochondrial metabolic tests demonstrated that C9ORF72-ALS BMECs display a significant decrease in basal glycolysis accompanied by increased basal and ATP-linked respiration. Moreover, our study reveals that C9-ALS derived astrocytes can further affect BMECs function and affect the expression of the glucose transporter Glut-1. Finally, C9ORF72 patient-derived BMECs form leaky barriers through a cell-autonomous mechanism and have neurotoxic properties towards motor neurons. ","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"44 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2024-04-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140573618","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation 空腹通过 PPAR δ 激活上调大鼠血脑屏障上的单羧酸盐转运体 MCT1
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-04-08 DOI: 10.1186/s12987-024-00526-8
Stéphanie Chasseigneaux, Véronique Cochois-Guégan, Lucas Lecorgne, Murielle Lochus, Sophie Nicolic, Corinne Blugeon, Laurent Jourdren, David Gomez-Zepeda, Stefan Tenzer, Sylvia Sanquer, Valérie Nivet-Antoine, Marie-Claude Menet, Jean-Louis Laplanche, Xavier Declèves, Salvatore Cisternino, Bruno Saubaméa
The blood-brain barrier (BBB) is pivotal for the maintenance of brain homeostasis and it strictly regulates the cerebral transport of a wide range of endogenous compounds and drugs. While fasting is increasingly recognized as a potential therapeutic intervention in neurology and psychiatry, its impact upon the BBB has not been studied. This study was designed to assess the global impact of fasting upon the repertoire of BBB transporters. We used a combination of in vivo and in vitro experiments to assess the response of the brain endothelium in male rats that were fed ad libitum or fasted for one to three days. Brain endothelial cells were acutely purified and transcriptionaly profiled using RNA-Seq. Isolated brain microvessels were used to assess the protein expression of selected BBB transporters through western blot. The molecular mechanisms involved in the adaptation to fasting were investigated in primary cultured rat brain endothelial cells. MCT1 activity was probed by in situ brain perfusion. Fasting did not change the expression of the main drug efflux ATP-binding cassette transporters or P-glycoprotein activity at the BBB but modulated a restrictive set of solute carrier transporters. These included the ketone bodies transporter MCT1, which is pivotal for the brain adaptation to fasting. Our findings in vivo suggested that PPAR δ, a major lipid sensor, was selectively activated in brain endothelial cells in response to fasting. This was confirmed in vitro where pharmacological agonists and free fatty acids selectively activated PPAR δ, resulting in the upregulation of MCT1 expression. Moreover, dosing rats with a specific PPAR δ antagonist blocked the upregulation of MCT1 expression and activity induced by fasting. Altogether, our study shows that fasting affects a selected set of BBB transporters which does not include the main drug efflux transporters. Moreover, we describe a previously unknown selective adaptive response of the brain vasculature to fasting which involves PPAR δ and is responsible for the up-regulation of MCT1 expression and activity. Our study opens new perspectives for the metabolic manipulation of the BBB in the healthy or diseased brain.
血脑屏障(BBB)是维持大脑平衡的关键,它严格控制着多种内源性化合物和药物的脑转运。虽然禁食越来越被认为是神经学和精神病学的一种潜在治疗干预措施,但其对 BBB 的影响尚未得到研究。本研究旨在评估禁食对 BBB 转运体的整体影响。我们采用体内和体外实验相结合的方法来评估自由进食或禁食一至三天的雄性大鼠脑内皮细胞的反应。急性纯化脑内皮细胞并使用 RNA-Seq 进行转录分析。利用分离的脑微血管通过Western印迹来评估选定的BBB转运体的蛋白表达。在原代培养的大鼠脑内皮细胞中研究了适应禁食的分子机制。通过原位脑灌注检测了MCT1的活性。禁食不会改变主要药物外流 ATP 结合盒转运体的表达或 P 糖蛋白在 BBB 的活性,但会调节一组限制性溶质载体转运体。其中包括酮体转运体 MCT1,它对大脑适应禁食起着关键作用。我们在体内的研究结果表明,PPAR δ(一种主要的脂质传感器)在大脑内皮细胞中被选择性地激活,以应对禁食。这一点在体外得到了证实,药理激动剂和游离脂肪酸可选择性地激活 PPAR δ,导致 MCT1 表达上调。此外,给大鼠服用特异性 PPAR δ 拮抗剂可阻断禁食诱导的 MCT1 表达和活性上调。总之,我们的研究表明,禁食会影响一组选定的 BBB 转运体,其中不包括主要的药物外流转运体。此外,我们还描述了一种以前未知的脑血管对禁食的选择性适应反应,这种反应涉及 PPAR δ,是 MCT1 表达和活性上调的原因。我们的研究为在健康或患病大脑中操纵 BBB 的新陈代谢开辟了新的前景。
{"title":"Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation","authors":"Stéphanie Chasseigneaux, Véronique Cochois-Guégan, Lucas Lecorgne, Murielle Lochus, Sophie Nicolic, Corinne Blugeon, Laurent Jourdren, David Gomez-Zepeda, Stefan Tenzer, Sylvia Sanquer, Valérie Nivet-Antoine, Marie-Claude Menet, Jean-Louis Laplanche, Xavier Declèves, Salvatore Cisternino, Bruno Saubaméa","doi":"10.1186/s12987-024-00526-8","DOIUrl":"https://doi.org/10.1186/s12987-024-00526-8","url":null,"abstract":"The blood-brain barrier (BBB) is pivotal for the maintenance of brain homeostasis and it strictly regulates the cerebral transport of a wide range of endogenous compounds and drugs. While fasting is increasingly recognized as a potential therapeutic intervention in neurology and psychiatry, its impact upon the BBB has not been studied. This study was designed to assess the global impact of fasting upon the repertoire of BBB transporters. We used a combination of in vivo and in vitro experiments to assess the response of the brain endothelium in male rats that were fed ad libitum or fasted for one to three days. Brain endothelial cells were acutely purified and transcriptionaly profiled using RNA-Seq. Isolated brain microvessels were used to assess the protein expression of selected BBB transporters through western blot. The molecular mechanisms involved in the adaptation to fasting were investigated in primary cultured rat brain endothelial cells. MCT1 activity was probed by in situ brain perfusion. Fasting did not change the expression of the main drug efflux ATP-binding cassette transporters or P-glycoprotein activity at the BBB but modulated a restrictive set of solute carrier transporters. These included the ketone bodies transporter MCT1, which is pivotal for the brain adaptation to fasting. Our findings in vivo suggested that PPAR δ, a major lipid sensor, was selectively activated in brain endothelial cells in response to fasting. This was confirmed in vitro where pharmacological agonists and free fatty acids selectively activated PPAR δ, resulting in the upregulation of MCT1 expression. Moreover, dosing rats with a specific PPAR δ antagonist blocked the upregulation of MCT1 expression and activity induced by fasting. Altogether, our study shows that fasting affects a selected set of BBB transporters which does not include the main drug efflux transporters. Moreover, we describe a previously unknown selective adaptive response of the brain vasculature to fasting which involves PPAR δ and is responsible for the up-regulation of MCT1 expression and activity. Our study opens new perspectives for the metabolic manipulation of the BBB in the healthy or diseased brain.","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"5 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2024-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140573287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SARS-CoV-2 causes dysfunction in human iPSC-derived brain microvascular endothelial cells potentially by modulating the Wnt signaling pathway SARS-CoV-2 可能通过调节 Wnt 信号通路导致人 iPSC 衍生脑微血管内皮细胞功能障碍
IF 7.3 1区 医学 Q1 NEUROSCIENCES Pub Date : 2024-04-08 DOI: 10.1186/s12987-024-00533-9
Shigeru Yamada, Tadahiro Hashita, Shota Yanagida, Hiroyuki Sato, Yukuto Yasuhiko, Kaori Okabe, Takamasa Noda, Motohiro Nishida, Tamihide Matsunaga, Yasunari Kanda
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which is associated with various neurological symptoms, including nausea, dizziness, headache, encephalitis, and epileptic seizures. SARS-CoV-2 is considered to affect the central nervous system (CNS) by interacting with the blood–brain barrier (BBB), which is defined by tight junctions that seal paracellular gaps between brain microvascular endothelial cells (BMECs). Although SARS-CoV-2 infection of BMECs has been reported, the detailed mechanism has not been fully elucidated. Using the original strain of SARS-CoV-2, the infection in BMECs was confirmed by a detection of intracellular RNA copy number and localization of viral particles. BMEC functions were evaluated by measuring transendothelial electrical resistance (TEER), which evaluates the integrity of tight junction dynamics, and expression levels of proinflammatory genes. BMEC signaling pathway was examined by comprehensive RNA-seq analysis. We observed that iPSC derived brain microvascular endothelial like cells (iPSC-BMELCs) were infected with SARS-CoV-2. SARS-CoV-2 infection resulted in decreased TEER. In addition, SARS-CoV-2 infection decreased expression levels of tight junction markers CLDN3 and CLDN11. SARS-CoV-2 infection also increased expression levels of proinflammatory genes, which are known to be elevated in patients with COVID-19. Furthermore, RNA-seq analysis revealed that SARS-CoV-2 dysregulated the canonical Wnt signaling pathway in iPSC-BMELCs. Modulation of the Wnt signaling by CHIR99021 partially inhibited the infection and the subsequent inflammatory responses. These findings suggest that SARS-CoV-2 infection causes BBB dysfunction via Wnt signaling. Thus, iPSC-BMELCs are a useful in vitro model for elucidating COVID-19 neuropathology and drug development.
严重急性呼吸系统综合征冠状病毒 2(SARS-CoV-2)会引起 2019 年冠状病毒病(COVID-19),并伴有各种神经系统症状,包括恶心、头晕、头痛、脑炎和癫痫发作。SARS-CoV-2 被认为是通过与血脑屏障(BBB)相互作用来影响中枢神经系统(CNS)的,而血脑屏障是由封闭脑微血管内皮细胞(BMECs)之间旁细胞间隙的紧密连接所定义的。虽然已有 SARS-CoV-2 感染 BMEC 的报道,但其详细机制尚未完全阐明。利用 SARS-CoV-2 原始菌株,通过检测细胞内 RNA 拷贝数和病毒颗粒定位,确认了 BMECs 的感染。通过测量跨内皮电阻(TEER)和促炎症基因的表达水平来评估 BMEC 的功能,TEER 可评估紧密连接动态的完整性。通过全面的 RNA-seq 分析研究了 BMEC 信号通路。我们观察到,iPSC 衍生的脑微血管内皮样细胞(iPSC-BMELCs)感染了 SARS-CoV-2。SARS-CoV-2 感染导致 TEER 下降。此外,SARS-CoV-2 感染还降低了紧密连接标记物 CLDN3 和 CLDN11 的表达水平。SARS-CoV-2 感染还增加了促炎症基因的表达水平,众所周知,这些基因在 COVID-19 患者中会升高。此外,RNA-seq分析显示,SARS-CoV-2对iPSC-BMELCs中的典型Wnt信号通路进行了失调。CHIR99021 对 Wnt 信号的调节部分抑制了感染和随后的炎症反应。这些发现表明,SARS-CoV-2感染会通过Wnt信号传导导致BBB功能障碍。因此,iPSC-BMELCs 是阐明 COVID-19 神经病理学和药物开发的有用体外模型。
{"title":"SARS-CoV-2 causes dysfunction in human iPSC-derived brain microvascular endothelial cells potentially by modulating the Wnt signaling pathway","authors":"Shigeru Yamada, Tadahiro Hashita, Shota Yanagida, Hiroyuki Sato, Yukuto Yasuhiko, Kaori Okabe, Takamasa Noda, Motohiro Nishida, Tamihide Matsunaga, Yasunari Kanda","doi":"10.1186/s12987-024-00533-9","DOIUrl":"https://doi.org/10.1186/s12987-024-00533-9","url":null,"abstract":"Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which is associated with various neurological symptoms, including nausea, dizziness, headache, encephalitis, and epileptic seizures. SARS-CoV-2 is considered to affect the central nervous system (CNS) by interacting with the blood–brain barrier (BBB), which is defined by tight junctions that seal paracellular gaps between brain microvascular endothelial cells (BMECs). Although SARS-CoV-2 infection of BMECs has been reported, the detailed mechanism has not been fully elucidated. Using the original strain of SARS-CoV-2, the infection in BMECs was confirmed by a detection of intracellular RNA copy number and localization of viral particles. BMEC functions were evaluated by measuring transendothelial electrical resistance (TEER), which evaluates the integrity of tight junction dynamics, and expression levels of proinflammatory genes. BMEC signaling pathway was examined by comprehensive RNA-seq analysis. We observed that iPSC derived brain microvascular endothelial like cells (iPSC-BMELCs) were infected with SARS-CoV-2. SARS-CoV-2 infection resulted in decreased TEER. In addition, SARS-CoV-2 infection decreased expression levels of tight junction markers CLDN3 and CLDN11. SARS-CoV-2 infection also increased expression levels of proinflammatory genes, which are known to be elevated in patients with COVID-19. Furthermore, RNA-seq analysis revealed that SARS-CoV-2 dysregulated the canonical Wnt signaling pathway in iPSC-BMELCs. Modulation of the Wnt signaling by CHIR99021 partially inhibited the infection and the subsequent inflammatory responses. These findings suggest that SARS-CoV-2 infection causes BBB dysfunction via Wnt signaling. Thus, iPSC-BMELCs are a useful in vitro model for elucidating COVID-19 neuropathology and drug development.","PeriodicalId":12321,"journal":{"name":"Fluids and Barriers of the CNS","volume":"80 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2024-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140573173","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Fluids and Barriers of the CNS
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