Pub Date : 2024-10-27DOI: 10.1016/j.neuroscience.2024.10.041
Boris Lamotte d'Incamps
Philippe Ascher spent his last two decades as an emeritus Professor, working in the heart of Paris. Together with his wife Jacsue they were hosted in Alain Marty's laboratory and enjoyed the happiest retirement. We started our collaboration a few years after they started their retirement research at the Saint Pères campus where I was working on spinal motoneurons' physiology. This period led us from NMDA receptors to the corelease of acetylcholine and glutamate by spinal motoneurons to Renshaw cells and then to the stoichiometric variants of nicotinic acetylcholine receptors. Here I present a brief history of our collaboration during this period.
{"title":"Renshaw cells, corelease and nicotinic receptors: The last journey in synaptic transmission.","authors":"Boris Lamotte d'Incamps","doi":"10.1016/j.neuroscience.2024.10.041","DOIUrl":"https://doi.org/10.1016/j.neuroscience.2024.10.041","url":null,"abstract":"<p><p>Philippe Ascher spent his last two decades as an emeritus Professor, working in the heart of Paris. Together with his wife Jacsue they were hosted in Alain Marty's laboratory and enjoyed the happiest retirement. We started our collaboration a few years after they started their retirement research at the Saint Pères campus where I was working on spinal motoneurons' physiology. This period led us from NMDA receptors to the corelease of acetylcholine and glutamate by spinal motoneurons to Renshaw cells and then to the stoichiometric variants of nicotinic acetylcholine receptors. Here I present a brief history of our collaboration during this period.</p>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142550079","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-26DOI: 10.1016/j.neuroscience.2024.10.047
Erin B Rousseau, Hannah D Jackson, Suman Guha, Sydney S Sherman, Michael Cima, Elena H Chartoff
Targeted intracranial delivery of molecularly-specific therapies within intricate brain structures poses a formidable challenge due to the heterogeneity of neuronal phenotypes and functions. Here we report the use of an implantable, miniaturized neural drug delivery system permitting dynamic adjustment of pharmacotherapies. Specifically, we exploit the spatial accuracy afforded by this method for targeting modulation of neuronal microstructures. Kappa opioid receptors (KOR) within the dorsal medial nucleus accumbens shell (NASh) are selectively activated through micro infusions of the KOR agonist, U-50488. Remarkably, we demonstrate that micro infusions of U-50488 into the dorsal NASh induces reward-like conditioned place preferences, whereas a mere 1 mm shift ventrally results in conditioned place aversions. The striking precision afforded by this method may prove useful in other neurotherapeutic interventions.
由于神经元表型和功能的异质性,在错综复杂的大脑结构中进行分子特异性疗法的颅内靶向给药是一项艰巨的挑战。在这里,我们报告了一种可植入的微型神经给药系统的使用情况,该系统允许动态调整药物疗法。具体来说,我们利用这种方法所提供的空间精确性,对神经元微结构进行靶向调节。通过微量注射 KOR 激动剂 U-50488 来选择性激活背内侧核团外壳(NASh)内的卡巴阿片受体(KOR)。值得注意的是,我们证明将 U-50488 微量注入背侧纳森氏核诱导出类似奖赏的条件性场所偏好,而仅向腹侧移动 1 毫米就会导致条件性场所厌恶。这种方法的惊人精确性可能会被证明有助于其他神经治疗干预。
{"title":"Microdosing of a kappa opioid receptor agonist within proximal nucleus accumbens shell microstructures revealing opposing behavioral outcomes.","authors":"Erin B Rousseau, Hannah D Jackson, Suman Guha, Sydney S Sherman, Michael Cima, Elena H Chartoff","doi":"10.1016/j.neuroscience.2024.10.047","DOIUrl":"https://doi.org/10.1016/j.neuroscience.2024.10.047","url":null,"abstract":"<p><p>Targeted intracranial delivery of molecularly-specific therapies within intricate brain structures poses a formidable challenge due to the heterogeneity of neuronal phenotypes and functions. Here we report the use of an implantable, miniaturized neural drug delivery system permitting dynamic adjustment of pharmacotherapies. Specifically, we exploit the spatial accuracy afforded by this method for targeting modulation of neuronal microstructures. Kappa opioid receptors (KOR) within the dorsal medial nucleus accumbens shell (NASh) are selectively activated through micro infusions of the KOR agonist, U-50488. Remarkably, we demonstrate that micro infusions of U-50488 into the dorsal NASh induces reward-like conditioned place preferences, whereas a mere 1 mm shift ventrally results in conditioned place aversions. The striking precision afforded by this method may prove useful in other neurotherapeutic interventions.</p>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567509","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-26DOI: 10.1016/j.neuroscience.2024.10.018
Background
Stem-cell derived extracellular vesicles (EVs) have shown promise in preclinical spinal cord injury (SCI) models but lack a comprehensive literature review for clinical translation guidance.
Methods
This meta-analysis with trial sequential analysis systematically search PubMed, Web of Science, Embase, and Cochrane Library databases. Prespecified inclusion criteria were studies reporting on measurable outcomes relevant to SCI repair. Risk of bias and quality of reporting were assessed. Random-effects meta-analyses and subgroup analyses comparing natural and bio-engineered EVs were performed. The study was registered with PROSPERO (CRD42024512122).
Findings
The search identified 3935 records, of which 39 studies were included, totaling 1801 animals. Administration of EVs significantly improved locomotor function as measured by Basso-Beattie-Bresnahan or Basso-Mouse-Scale scores at 1 week (natural EVs: SMD 1.50, 95 % CI 1.06–1.95; bio-engineered EVs: SMD 1.93, 95 % CI 1.34–2.52) and 3 weeks (natural EVs: SMD 2.57, 95 % CI 1.96–3.17; bio-engineered EVs: SMD 3.16, 95 % CI 2.29–4.02) post-injury. Subgroup analyses indicated surface modification approaches were most effective among bio-engineered EV strategies. EVs also promoted nerve growth (SMD 2.95, 95 % CI 2.12–3.78), enhanced neuron conductivity (MD 0.75, 95 %CI 0.59–0.90), alleviated inflammation (SMD −3.12, 95 % CI −4.15--2.10), and reduced lesion size (SMD −2.90, 95 % CI −3.87--1.93).
Conclusions
Both natural and bio-engineered EVs improve functional and pathological outcomes in animal models of SCI. The enhanced benefits observed with bio-engineered EVs, particularly those utilizing surface modification approaches, highlight the importance of continued exploration into bio-engineering techniques to optimize EVs’ therapeutic efficacy for SCI repair.
Protocol Registration
CRD42024512122.
背景:干细胞衍生的细胞外囊泡(EVs干细胞衍生的细胞外囊泡(EVs)在临床前脊髓损伤(SCI)模型中显示出前景,但缺乏全面的文献综述来指导临床转化:这项荟萃分析和试验序列分析系统地检索了 PubMed、Web of Science、Embase 和 Cochrane Library 数据库。预设的纳入标准是报告与 SCI 修复相关的可测量结果的研究。对偏倚风险和报告质量进行了评估。对天然 EV 和生物工程 EV 进行了随机效应荟萃分析和亚组分析。该研究已在 PROSPERO(CRD42024512122)注册:研究结果:检索到 3935 条记录,其中包括 39 项研究,共计 1801 只动物。根据巴索-巴蒂-布雷斯纳汉(Basso-Beattie-Bresnahan)或巴索-小鼠量表(Basso-Mouse-Scale)的评分,服用EVs能明显改善动物1周后的运动功能(天然EVs:SMD 1.50,95 %):SMD:1.50,95 % CI 1.06-1.95;生物工程EVs:1.93,95 % CI 1.34-2.52)和 3 周(天然 EVs:SMD为2.57,95 % CI为1.96-3.17;生物工程EVs:损伤后 3 周(天然 EV:SMD 2.57,95 % CI 1.96-3.17;生物工程 EV:SMD 3.16,95 % CI 2.29-4.02)。分组分析表明,在生物工程EV策略中,表面修饰方法最为有效。EVs还能促进神经生长(SMD 2.95,95 % CI 2.12-3.78)、增强神经元传导性(MD 0.75,95 %CI 0.59-0.90)、减轻炎症(SMD -3.12,95 % CI -4.15--2.10)并缩小病变范围(SMD -2.90,95 % CI -3.87--1.93):结论:天然EVs和生物工程EVs都能改善SCI动物模型的功能和病理结果。结论:天然EVs和生物工程EVs都能改善SCI动物模型的功能和病理结果。观察到生物工程EVs,尤其是利用表面修饰方法的EVs,能带来更多益处,这凸显了继续探索生物工程技术以优化EVs对SCI修复疗效的重要性。协议注册号:CRD42024512122。
{"title":"Natural and bio-engineered stem cell-derived extracellular vesicles for spinal cord injury repair: A meta-analysis with trial sequential analysis","authors":"","doi":"10.1016/j.neuroscience.2024.10.018","DOIUrl":"10.1016/j.neuroscience.2024.10.018","url":null,"abstract":"<div><h3>Background</h3><div>Stem-cell derived extracellular vesicles (EVs) have shown promise in preclinical spinal cord injury (SCI) models but lack a comprehensive literature review for clinical translation guidance.</div></div><div><h3>Methods</h3><div>This <em>meta</em>-analysis with trial sequential analysis systematically search PubMed, Web of Science, Embase, and Cochrane Library databases. Prespecified inclusion criteria were studies reporting on measurable outcomes relevant to SCI repair. Risk of bias and quality of reporting were assessed. Random-effects <em>meta</em>-analyses and subgroup analyses comparing natural and bio-engineered EVs were performed. The study was registered with PROSPERO (CRD42024512122).</div></div><div><h3>Findings</h3><div>The search identified 3935 records, of which 39 studies were included, totaling 1801 animals. Administration of EVs significantly improved locomotor function as measured by Basso-Beattie-Bresnahan or Basso-Mouse-Scale scores at 1 week (natural EVs: SMD 1.50, 95 % CI 1.06–1.95; bio-engineered EVs: SMD 1.93, 95 % CI 1.34–2.52) and 3 weeks (natural EVs: SMD 2.57, 95 % CI 1.96–3.17; bio-engineered EVs: SMD 3.16, 95 % CI 2.29–4.02) post-injury. Subgroup analyses indicated surface modification approaches were most effective among bio-engineered EV strategies. EVs also promoted nerve growth (SMD 2.95, 95 % CI 2.12–3.78), enhanced neuron conductivity (MD 0.75, 95 %CI 0.59–0.90), alleviated inflammation (SMD −3.12, 95 % CI −4.15--2.10), and reduced lesion size (SMD −2.90, 95 % CI −3.87--1.93).</div></div><div><h3>Conclusions</h3><div>Both natural and bio-engineered EVs improve functional and pathological outcomes in animal models of SCI. The enhanced benefits observed with bio-engineered EVs, particularly those utilizing surface modification approaches, highlight the importance of continued exploration into bio-engineering techniques to optimize EVs’ therapeutic efficacy for SCI repair.</div><div><strong>Protocol Registration</strong></div><div>CRD42024512122.</div></div>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-26DOI: 10.1016/j.neuroscience.2024.10.044
{"title":"Corrigendum to “Assessing the location, relative expression and subclass of dopamine receptors in the cerebellum of hemi-parkinsonian rats” [Neuroscience 521 (2023) 1–19]","authors":"","doi":"10.1016/j.neuroscience.2024.10.044","DOIUrl":"10.1016/j.neuroscience.2024.10.044","url":null,"abstract":"","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142552573","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-26DOI: 10.1016/j.neuroscience.2024.10.046
Neuroprobes that use nanostructured photonic interfaces are capable of multimodal sensing, stimulation, and imaging with unprecedented spatio-temporal resolution. In addition to electrical recording, optogenetic modulation, high-resolution optical imaging, and molecular sensing, these advanced probes combine nanophotonic waveguides, optical transducers, nanostructured electrodes, and biochemical sensors. The potential of this technology lies in unraveling the mysteries of neural coding principles, mapping functional connectivity in complex brain circuits, and developing new therapeutic interventions for neurological disorders. Nevertheless, achieving the full potential of nanostructured photonic neural probes requires overcoming challenges such as ensuring long-term biocompatibility, integrating nanoscale components at high density, and developing robust data-analysis pipelines. In this review, we summarize and discuss the role of photonics in neural probes, trends in electrode diameter for neural interface technologies, nanophotonic technologies using nanostructured materials, advances in nanofabrication photonics interface engineering, and challenges and opportunities. Finally, interdisciplinary efforts are required to unlock the transformative potential of next-generation neuroscience therapies.
{"title":"Nanostructured Photonics Probes: A Transformative Approach in Neurotherapeutics and Brain Circuitry","authors":"","doi":"10.1016/j.neuroscience.2024.10.046","DOIUrl":"10.1016/j.neuroscience.2024.10.046","url":null,"abstract":"<div><div>Neuroprobes that use nanostructured photonic interfaces are capable of multimodal sensing, stimulation, and imaging with unprecedented spatio-temporal resolution. In addition to electrical recording, optogenetic modulation, high-resolution optical imaging, and molecular sensing, these advanced probes combine nanophotonic waveguides, optical transducers, nanostructured electrodes, and biochemical sensors. The potential of this technology lies in unraveling the mysteries of neural coding principles, mapping functional connectivity in complex brain circuits, and developing new therapeutic interventions for neurological disorders. Nevertheless, achieving the full potential of nanostructured photonic neural probes requires overcoming challenges such as ensuring long-term biocompatibility, integrating nanoscale components at high density, and developing robust data-analysis pipelines. In this review, we summarize and discuss the role of photonics in neural probes, trends in electrode diameter for neural interface technologies, nanophotonic technologies using nanostructured materials, advances in nanofabrication photonics interface engineering, and challenges and opportunities. Finally, interdisciplinary efforts are required to unlock the transformative potential of next-generation neuroscience therapies.</div></div>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142560751","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-24DOI: 10.1016/j.neuroscience.2024.10.033
The study aimed to validate the protective effect of neuroglobin (Ngb) in a cell model of Parkinson’s disease (PD) and explore its therapeutic potential. Lentivirus-Ngb (LvNgb) and siRNA-Ngb (siNgb) were used to achieve Ngb overexpression and knockdown, respectively, in a sporadic PD cell model. Apoptosis was evaluated by flow cytometry-based Annexin V/propidium iodide assays. Activation of the pro-apoptotic factor, Caspase-9, was detected by immunoblotting, and Complex I activities were detected by using enzyme-linked immunosorbent assay (ELISA). Mitochondrial dysfunction was examined by measuring the mitochondrial membrane potential (MMP), NAD+/NADH ratios, and reactive oxygen species (ROS) levels. Additionally, coimmunoprecipitation (Co-IP) assays were conducted in mouse neuroblastoma cell line 9D (MN9D) cells to determine the interactions of Ngb with the Complex I subunit NDUFA10. The results showed that Ngb overexpression reduced the percentages of apoptotic cells, total caspase-9 levels and restored Complex I activities in the PD cell model. Conversely, knockdown of Ngb resulted in an increase in apoptotic cells, higher total caspase-9 levels, and decreased Complex I activities. Furthermore, Ngb overexpression restored MMP and NAD+/NADH ratios and alleviated ROS-mediated oxidative stress in MN9D cells. Finally, Co-IP confirmed the interaction between Ngb and NDUFA10 in MN9D cells. In conclusion, Ngb protects MN9D cells against apoptosis by interacting with Complex I subunit NDUFA10, rescuing its activity and inhibiting the mitochondrial pathway of apoptosis in the MPP+-mediated PD model.
{"title":"Neuroglobin protects dopaminergic neurons in a Parkinson’s cell model by interacting with mitochondrial complex NDUFA10","authors":"","doi":"10.1016/j.neuroscience.2024.10.033","DOIUrl":"10.1016/j.neuroscience.2024.10.033","url":null,"abstract":"<div><div>The study aimed to validate the protective effect of neuroglobin (Ngb) in a cell model of Parkinson’s disease (PD) and explore its therapeutic potential. Lentivirus-Ngb (LvNgb) and siRNA-Ngb (siNgb) were used to achieve Ngb overexpression and knockdown, respectively, in a sporadic PD cell model. Apoptosis was evaluated by flow cytometry-based Annexin V/propidium iodide assays. Activation of the pro-apoptotic factor, Caspase-9, was detected by immunoblotting, and Complex I activities were detected by using enzyme-linked immunosorbent assay (ELISA). Mitochondrial dysfunction was examined by measuring the mitochondrial membrane potential (MMP), NAD<sup>+</sup>/NADH ratios, and reactive oxygen species (ROS) levels. Additionally, coimmunoprecipitation (Co-IP) assays were conducted in mouse neuroblastoma cell line 9D (MN9D) cells to determine the interactions of Ngb with the Complex I subunit NDUFA10. The results showed that Ngb overexpression reduced the percentages of apoptotic cells, total caspase-9 levels and restored Complex I activities in the PD cell model. Conversely, knockdown of Ngb resulted in an increase in apoptotic cells, higher total caspase-9 levels, and decreased Complex I activities. Furthermore, Ngb overexpression restored MMP and NAD<sup>+</sup>/NADH ratios and alleviated ROS-mediated oxidative stress in MN9D cells. Finally, Co-IP confirmed the interaction between Ngb and NDUFA10 in MN9D cells. In conclusion, Ngb protects MN9D cells against apoptosis by interacting with Complex I subunit NDUFA10, rescuing its activity and inhibiting the mitochondrial pathway of apoptosis in the MPP<sup>+</sup>-mediated PD model.</div></div>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504862","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-24DOI: 10.1016/j.neuroscience.2024.10.043
Mecamylamine, a noncompetitive blocker of nicotinic acetylcholine receptors (nAChRs), is the racemic mixture of two stereoisomers: S-(+)-mecamylamine (S-mec) and R-(−)-mecamylamine (R-mec), with distinct interactions with α4β2 nAChRs. It has been shown that mecamylamine increases glutamate release and excites serotonergic (5-HT) neurons in the dorsal raphe nucleus (DRN). In this study, we separately evaluated the effects of S-mec and R-mec on 5-HT neuron excitability. S-mec (3 μM) increased firing frequency by 40 %, while R-mec (3 μM) raised it by only 22 %. S-mec acts as a positive allosteric modulator on high-sensitivity (HS) α4β2 nAChRs at glutamate terminals, enhancing spontaneous excitatory postsynaptic currents (sEPSCs) in 5-HT neurons. Conversely, R-mec decreased sEPSCs by blocking HS α4β2 nAChRs and reduced GABA-mediated inhibitory currents (sIPSCs) by blocking α7 nAChRs at GABAergic terminals. These mechanisms make S-mec more effective than R-mec in enhancing 5-HT neuron firing. Moreover, combining S-mec with TC-2559, a selective agonist of HS α4β2 nAChRs, increased firing frequency by 65 %, exceeding the effect of S-mec alone. To validate these findings, we evaluated the antidepressant effects of S-mec (1 mg/kg) combined with TC-2559 or RJR-2403, another α4β2 nAChR agonist. This combination successfully reduced depression-like behaviors, suggesting a potential treatment strategy for patients resistant to conventional antidepressants.
{"title":"S-(+)-mecamylamine increases the firing rate of serotonin neurons and diminishes depressive-like behaviors in an animal model of stress","authors":"","doi":"10.1016/j.neuroscience.2024.10.043","DOIUrl":"10.1016/j.neuroscience.2024.10.043","url":null,"abstract":"<div><div>Mecamylamine, a noncompetitive blocker of nicotinic acetylcholine receptors (nAChRs), is the racemic mixture of two stereoisomers: S-(+)-mecamylamine (S-mec) and R-(−)-mecamylamine (R-mec), with distinct interactions with α4β2 nAChRs. It has been shown that mecamylamine increases glutamate release and excites serotonergic (5-HT) neurons in the dorsal raphe nucleus (DRN). In this study, we separately evaluated the effects of S-mec and R-mec on 5-HT neuron excitability. S-mec (3 μM) increased firing frequency by 40 %, while R-mec (3 μM) raised it by only 22 %. S-mec acts as a positive allosteric modulator on high-sensitivity (HS) α4β2 nAChRs at glutamate terminals, enhancing spontaneous excitatory postsynaptic currents (sEPSCs) in 5-HT neurons. Conversely, R-mec decreased sEPSCs by blocking HS α4β2 nAChRs and reduced GABA-mediated inhibitory currents (sIPSCs) by blocking α7 nAChRs at GABAergic terminals. These mechanisms make S-mec more effective than R-mec in enhancing 5-HT neuron firing. Moreover, combining S-mec with TC-2559, a selective agonist of HS α4β2 nAChRs, increased firing frequency by 65 %, exceeding the effect of S-mec alone. To validate these findings, we evaluated the antidepressant effects of S-mec (1 mg/kg) combined with TC-2559 or RJR-2403, another α4β2 nAChR agonist. This combination successfully reduced depression-like behaviors, suggesting a potential treatment strategy for patients resistant to conventional antidepressants.</div></div>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504864","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}
Pub Date : 2024-10-24DOI: 10.1016/j.neuroscience.2024.10.038
Xili Jiang, Wei Zhang, Shucai Xie
Background: Neonatal hypoxia-ischemia encephalopathy (HIE) is a significant reason for neonatal mortality and prolonged disability. We have previously revealed that GPR39 activation attenuates neuroinflammation in a neonatal HIE rat model. This study aimed to investigate whether GPR39 affected microglial pyroptosis post-HIE.
Methods: A neonatal rat model of HIE and a microglia cell model of oxygen-glucose deprivation (OGD) were established. Neuronal loss and cerebral infarction were assessed by using TTC, H&E staining, and Nissl staining. Pyroptosis was evaluated with western blot, LDH assay kit, ELISA, and flow cytometry. Total m6A level and GPR39 m6A modification were determined using m6A dot blot and MeRIP. The interaction between METTL3/HuR/GSK3β and GPR39 was analyzed by performing molecular interaction experiments. GPR39 mRNA stability was examined with actinomycin D.
Results: The level of GPR39 was increased in neonatal HIE rats and OGD-treated microglia. Brain injury and neuronal loss were significantly increased in the HIE model when GPR39 was knocked down. GPR39 knockdown aggravated NLRP3 inflammasome-mediated microglial pyroptosis. METTL3 upregulated GPR39 expression in an m6A-dependent manner. METTL3 enhanced the interaction of HuR and GPR39. In OGD-exposed microglia, METTL3 elevated GPR39 expression and mRNA stability, which declined after HuR depletion. METTL3 knockdown promoted microglial pyroptosis, which was reversed by GPR39 agonist. Furthermore, microglial pyroptosis was inhibited by GPR39 upregulation, but the outcome was reverted by GSK3β activator SNP.
Conclusion: METTL3 inhibits microglial pyroptosis in neonatal HIE via regulating m6A-HuR dependent stabilization of GPR39, which contributes to therapeutics development for neonatal HIE.
背景:新生儿缺氧缺血性脑病(HIE新生儿缺氧缺血性脑病(HIE)是导致新生儿死亡和长期残疾的重要原因。我们以前曾发现,在新生儿 HIE 大鼠模型中,GPR39 激活可减轻神经炎症。本研究旨在探讨 GPR39 是否会影响 HIE 后的小胶质细胞脓毒症:方法:建立了新生大鼠 HIE 模型和氧-葡萄糖剥夺(OGD)小胶质细胞模型。通过TTC、H&E染色和Nissl染色评估神经元损失和脑梗塞。用 Western 印迹、LDH 检测试剂盒、ELISA 和流式细胞术评估了嗜热症。总 m6A 水平和 GPR39 m6A 修饰是通过 m6A dot 印迹和 MeRIP 测定的。通过分子相互作用实验分析了 METTL3/HuR/GSK3β 与 GPR39 之间的相互作用。用放线菌素 D 检验了 GPR39 mRNA 的稳定性:结果:在新生 HIE 大鼠和 OGD 处理的小胶质细胞中,GPR39 水平升高。在 HIE 模型中,当 GPR39 被敲除时,脑损伤和神经元损失显著增加。敲除 GPR39 会加重 NLRP3 炎性体介导的小胶质细胞热解。METTL3 以 m6A 依赖性方式上调 GPR39 的表达。METTL3 增强了 HuR 和 GPR39 的相互作用。在暴露于 OGD 的小胶质细胞中,METTL3 提高了 GPR39 的表达和 mRNA 稳定性,而在 HuR 消耗后,这种表达和 mRNA 稳定性下降。敲除 METTL3 可促进小胶质细胞脓毒症,而 GPR39 激动剂可逆转这种现象。此外,GPR39上调可抑制小胶质细胞的脓毒症,但GSK3β激活剂SNP可逆转这一结果:结论:METTL3通过调控m6A-HuR依赖性GPR39的稳定来抑制新生儿HIE中的小胶质细胞脓毒症,这有助于新生儿HIE治疗药物的开发。
{"title":"METTL3 inhibits microglial pyroptosis in neonatal hypoxia-ischemia encephalopathy by regulating GPR39 expression in an m6A-HuR-dependent manner.","authors":"Xili Jiang, Wei Zhang, Shucai Xie","doi":"10.1016/j.neuroscience.2024.10.038","DOIUrl":"https://doi.org/10.1016/j.neuroscience.2024.10.038","url":null,"abstract":"<p><strong>Background: </strong>Neonatal hypoxia-ischemia encephalopathy (HIE) is a significant reason for neonatal mortality and prolonged disability. We have previously revealed that GPR39 activation attenuates neuroinflammation in a neonatal HIE rat model. This study aimed to investigate whether GPR39 affected microglial pyroptosis post-HIE.</p><p><strong>Methods: </strong>A neonatal rat model of HIE and a microglia cell model of oxygen-glucose deprivation (OGD) were established. Neuronal loss and cerebral infarction were assessed by using TTC, H&E staining, and Nissl staining. Pyroptosis was evaluated with western blot, LDH assay kit, ELISA, and flow cytometry. Total m6A level and GPR39 m6A modification were determined using m6A dot blot and MeRIP. The interaction between METTL3/HuR/GSK3β and GPR39 was analyzed by performing molecular interaction experiments. GPR39 mRNA stability was examined with actinomycin D.</p><p><strong>Results: </strong>The level of GPR39 was increased in neonatal HIE rats and OGD-treated microglia. Brain injury and neuronal loss were significantly increased in the HIE model when GPR39 was knocked down. GPR39 knockdown aggravated NLRP3 inflammasome-mediated microglial pyroptosis. METTL3 upregulated GPR39 expression in an m6A-dependent manner. METTL3 enhanced the interaction of HuR and GPR39. In OGD-exposed microglia, METTL3 elevated GPR39 expression and mRNA stability, which declined after HuR depletion. METTL3 knockdown promoted microglial pyroptosis, which was reversed by GPR39 agonist. Furthermore, microglial pyroptosis was inhibited by GPR39 upregulation, but the outcome was reverted by GSK3β activator SNP.</p><p><strong>Conclusion: </strong>METTL3 inhibits microglial pyroptosis in neonatal HIE via regulating m6A-HuR dependent stabilization of GPR39, which contributes to therapeutics development for neonatal HIE.</p>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-24DOI: 10.1016/j.neuroscience.2024.10.031
The blood–brain barrier’s limited permeability to tenofovir restricts its ability to clear HIV from the brain. Probenecid acting as an adjuvant increases tenofovir concentrations in plasma and the kidneys thereby enhancing its therapeutic effect. However, the probenecid effect on brain tenofovir concentration and possible adverse effects remains poorly understood. We investigated the effect of probenecid co-administered tenofovir on tenofovir brain concentration, interleukin-1β (IL-1β) and dopamine concentration in the prefrontal cortex (PFC) and the cerebellum. Ninety-six male BALB/c mice were divided into four groups viz: a control group, Tenofovir disoproxil fumarate (TDF) treated, probenecid treated, and TDF + probenecid treated. We orally administered a single dose of TDF (5 mg/kg), and probenecid (8.3 mg/kg), and sacrificed six mice per group after 1 h, 4 h, and 6 h post-treatment to collect plasma, PFC, and cerebellar tissue. Co-administered tenofovir increased tenofovir concentration, peaking at 6 h with the cerebellum having the highest concentration. This suggests that probenecid enhanced the entry of tenofovir into the brain. Tenofovir alone increased IL-1β concentration at all intervals post-administration, while probenecid alone had no impact on IL-1β concentration. Co-administered tenofovir also increased IL-1β concentration. Probenecid’s limited impact on IL-1β concentration following co-administration suggests that its anti-inflammatory properties may require more than 6 h to have an effect. Furthermore, neither tenofovir nor probenecid affected dopamine concentration. In conclusion, probenecid enhances the concentration and retention of tenofovir in the brain, making it a possible pharmacokinetic enhancer. However, its anti-inflammatory effects may require a longer duration to fully manifest.
{"title":"Modulation of tenofovir by probenecid: Impact on drug, interleukin-1β, and dopamine concentration in the prefrontal cortex and cerebellum","authors":"","doi":"10.1016/j.neuroscience.2024.10.031","DOIUrl":"10.1016/j.neuroscience.2024.10.031","url":null,"abstract":"<div><div>The blood–brain barrier’s limited permeability to tenofovir restricts its ability to clear HIV from the brain. Probenecid acting as an adjuvant increases tenofovir concentrations in plasma and the kidneys thereby enhancing its therapeutic effect. However, the probenecid effect on brain tenofovir concentration and possible adverse effects remains poorly understood. We investigated the effect of probenecid co-administered tenofovir on tenofovir brain concentration, interleukin-1β (IL-1β) and dopamine concentration in the prefrontal cortex (PFC) and the cerebellum. Ninety-six male BALB/c mice were divided into four groups viz: a control group, Tenofovir disoproxil fumarate (TDF) treated, probenecid treated, and TDF + probenecid treated. We orally administered a single dose of TDF (5 mg/kg), and probenecid (8.3 mg/kg), and sacrificed six mice per group after 1 h, 4 h, and 6 h post-treatment to collect plasma, PFC, and cerebellar tissue. Co-administered tenofovir increased tenofovir concentration, peaking at 6 h with the cerebellum having the highest concentration. This suggests that probenecid enhanced the entry of tenofovir into the brain. Tenofovir alone increased IL-1β concentration at all intervals post-administration, while probenecid alone had no impact on IL-1β concentration. Co-administered tenofovir also increased IL-1β concentration. Probenecid’s limited impact on IL-1β concentration following co-administration suggests that its anti-inflammatory properties may require more than 6 h to have an effect. Furthermore, neither tenofovir nor probenecid affected dopamine concentration. In conclusion, probenecid enhances the concentration and retention of tenofovir in the brain, making it a possible pharmacokinetic enhancer. However, its anti-inflammatory effects may require a longer duration to fully manifest.</div></div>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504861","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}
Pub Date : 2024-10-24DOI: 10.1016/j.neuroscience.2024.10.040
Juan Carlos López-Ramos, Esther Martínez-Lara, Julia Serrano, Patricia Fernández, Gloria G Parras, Antonio Ruiz-Marcos, José Rodrigo
Thyroid hormones play an important morphogenetic role during the fetal and neonatal periods and regulate numerous metabolic processes. In the central nervous system, they control myelination and overall brain development, regional gene expression, and regulation of oxygen consumption. Their deficiency in the fetal and neonatal periods causes severe mental retardation, due to lack of thyroid function, or to iodine deficiency. At the same time, nitric oxide is an atypical neurotransmitter that also has special relevance in neuronal development and plasticity and functions as a vasodilator, regulating cerebral blood flow. Although under physiological conditions it functions as a neuroprotector, in excess it can be neurotoxic. We have studied, by immunocytochemical and Western blot techniques, the evolution of the expression of neuronal and inducible isoforms of the enzyme nitric oxide synthase, and of nitrotyrosine as a marker of protein nitration produced by the presence of nitric oxide, during the early stages of postnatal brain development. We induced hypothyroidism by administering mercaptomethylimidazole to pregnant mothers, from the seventh day of gestation until the sacrifice of the offspring. The results show a delay in the evolution of the expression of the two isoforms of the enzyme nitric oxide synthase in hypothyroid animals, followed by an anomalous overexpression in later stages. Finally, the expression of nitrotyrosine follows an evolution that is synchronized with that shown by both isoenzymes in control and hypothyroid animals.
甲状腺激素在胎儿期和新生儿期发挥着重要的形态发生作用,并调节着许多新陈代谢过程。在中枢神经系统中,它们控制着髓鞘化和大脑的整体发育、区域基因表达以及耗氧量的调节。在胎儿期和新生儿期,由于缺乏甲状腺功能或碘缺乏,它们的缺乏会导致严重的智力迟钝。同时,一氧化氮是一种非典型神经递质,在神经元的发育和可塑性方面也有特殊意义,并具有血管扩张剂的功能,可调节脑血流量。虽然在生理条件下,它具有保护神经的功能,但过量时也会对神经产生毒性。我们通过免疫细胞化学和 Western 印迹技术,研究了一氧化氮合酶的神经元和诱导型同工酶,以及一氧化氮存在时蛋白质硝化的标志物硝基酪氨酸在出生后大脑发育早期阶段的表达变化。我们从妊娠第七天开始给孕妇注射巯甲基咪唑,诱发甲状腺功能减退症,直至后代被处死。结果显示,在甲状腺功能减退的动物体内,一氧化氮合酶的两种同工酶的表达延迟演变,随后在后期出现异常过度表达。最后,硝基酪氨酸的表达与对照组和甲状腺机能减退动物体内两种同工酶的表达同步进行。
{"title":"Nitric oxide synthase system in the brain development of neonatal hypothyroid rats.","authors":"Juan Carlos López-Ramos, Esther Martínez-Lara, Julia Serrano, Patricia Fernández, Gloria G Parras, Antonio Ruiz-Marcos, José Rodrigo","doi":"10.1016/j.neuroscience.2024.10.040","DOIUrl":"https://doi.org/10.1016/j.neuroscience.2024.10.040","url":null,"abstract":"<p><p>Thyroid hormones play an important morphogenetic role during the fetal and neonatal periods and regulate numerous metabolic processes. In the central nervous system, they control myelination and overall brain development, regional gene expression, and regulation of oxygen consumption. Their deficiency in the fetal and neonatal periods causes severe mental retardation, due to lack of thyroid function, or to iodine deficiency. At the same time, nitric oxide is an atypical neurotransmitter that also has special relevance in neuronal development and plasticity and functions as a vasodilator, regulating cerebral blood flow. Although under physiological conditions it functions as a neuroprotector, in excess it can be neurotoxic. We have studied, by immunocytochemical and Western blot techniques, the evolution of the expression of neuronal and inducible isoforms of the enzyme nitric oxide synthase, and of nitrotyrosine as a marker of protein nitration produced by the presence of nitric oxide, during the early stages of postnatal brain development. We induced hypothyroidism by administering mercaptomethylimidazole to pregnant mothers, from the seventh day of gestation until the sacrifice of the offspring. The results show a delay in the evolution of the expression of the two isoforms of the enzyme nitric oxide synthase in hypothyroid animals, followed by an anomalous overexpression in later stages. Finally, the expression of nitrotyrosine follows an evolution that is synchronized with that shown by both isoenzymes in control and hypothyroid animals.</p>","PeriodicalId":19142,"journal":{"name":"Neuroscience","volume":null,"pages":null},"PeriodicalIF":2.9,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142504863","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}