Paraventricular Hypothalamic Nucleus

下丘脑室旁核
  • 文章类型: Journal Article
    迷幻药在积极的临床效果后经历了新的兴趣,然而,潜在效应的神经生物学机制仍不清楚.下丘脑室旁核(PVN)在应激反应中起着不可或缺的作用,自主神经功能,社会行为,和其他情感过程。我们研究了psilocin的作用,psilocybin的精神活性代谢产物,SpragueDawley大鼠PVN反应性的研究。通过c-Fos表达在雄性和雌性大鼠中测量,Psilocin增加了不依赖刺激的PVN活性。Psilocin增加了男性而不是女性的PVN对厌恶性空气刺激的反应性。在注射后2天和7天恢复反应性,没有组差异。此外,先前注射psilocin不会影响急性束缚应激后的PVN反应性。通过基线威胁响应进行分类的实验组表明,男性PVN反应性的增加是由主动威胁响应者驱动的。这些发现将PVN确定为迷幻药作用的重要部位,并暗示了威胁响应行为。
    Psychedelics have experienced renewed interest following positive clinical effects, however the neurobiological mechanisms underlying effects remain unclear. The paraventricular nucleus of the hypothalamus (PVN) plays an integral role in stress response, autonomic function, social behavior, and other affective processes. We investigated the effect of psilocin, the psychoactive metabolite of psilocybin, on PVN reactivity in Sprague Dawley rats. Psilocin increased stimulus-independent PVN activity as measured by c-Fos expression in male and female rats. Psilocin increased PVN reactivity to an aversive air-puff stimulus in males but not females. Reactivity was restored at 2- and 7-days post-injection with no group differences. Additionally, prior psilocin injection did not affect PVN reactivity following acute restraint stress. Experimental groups sub-classified by baseline threat responding indicate that increased male PVN reactivity is driven by active threat responders. These findings identify the PVN as a significant site of psychedelic drug action with implications for threat responding behavior.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:最近的研究已将移情缺陷确定为自闭症谱系障碍患者的核心损害和诊断标准;然而,同理心的改善主要集中在没有目标调节的行为干预上。我们试图比较与类似同理心的恐惧和疼痛行为相关的大脑区域,并探讨催产素-催产素受体系统在恐惧共情中的作用。
    方法:我们使用C57BL小鼠建立了2种恐惧共情和疼痛共情模型。我们采用免疫荧光组织化学技术观察整个大脑中c-Fos的表达,随后定量了不同大脑区域c-Fos阳性细胞的数量。此外,我们利用化学遗传学技术在Oxt-Cret-/+小鼠中选择性操纵这些神经元,以确定催产素在这一过程中的作用.
    结果:恐惧同理心激活的区域是前扣带回皮质,基底外侧杏仁核,伏隔核,室旁核(PVN),外侧罗布,腹侧和背侧海马体。疼痛共情激活的区域是前扣带皮质,基底外侧杏仁核,伏隔核,和外侧罗布。我们发现,增加PVN区域催产素神经元的活性增强了对恐惧移情的反应。这种增强可以通过催产素受体介导。
    结论:这项研究仅包括雄性动物,这限制了对研究结果的更广泛的解释。需要对电路功能进行进一步的研究。
    结论:与恐惧和疼痛共情调节有关的脑区表现出区别;小鼠PVN神经元的活动与共情行为呈正相关。这些发现强调了PVN催产素途径在调节恐惧移情中的作用,并表明催产素信号在介导移情反应中的重要性。
    BACKGROUND: Recent studies have identified empathy deficit as a core impairment and diagnostic criterion for people with autism spectrum disorders; however, the improvement of empathy focuses primarily on behavioural interventions without the target regulation. We sought to compare brain regions associated with empathy-like behaviours of fear and pain, and to explore the role of the oxytocin-oxytocin receptor system in fear empathy.
    METHODS: We used C57BL mice to establish 2 models of fear empathy and pain empathy. We employed immunofluorescence histochemical techniques to observe the expression of c-Fos throughout the entire brain and subsequently quantified the number of c-Fos-positive cells in different brain regions. Furthermore, we employed chemogenetic technology to selectively manipulate these neurons in Oxt-Cre-/+ mice to identify the role of oxytocin in this process.
    RESULTS: The regions activated by fear empathy were the anterior cingulate cortex, basolateral amygdala, nucleus accumbens, paraventricular nucleus (PVN), lateral habenula, and ventral and dorsal hippocampus. The regions activated by pain empathy were the anterior cingulate cortex, basolateral amygdala, nucleus accumbens, and lateral habenula. We found that increasing the activity of oxytocin neurons in the PVN region enhanced the response to fear empathy. This enhancement may be mediated through oxytocin receptors.
    CONCLUSIONS: This study included only male animals, which restricts the broader interpretation of the findings. Further investigations on circuit function need to be conducted.
    CONCLUSIONS: The brain regions implicated in the regulation of fear and pain empathy exhibit distinctions; the activity of PVN neurons was positively correlated with empathic behaviour in mice. These findings highlight the role of the PVN oxytocin pathway in regulating fear empathy and suggest the importance of oxytocin signalling in mediating empathetic responses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    我们先前表明,食欲素神经元被缺氧激活,并促进外周化学反射(PCR)介导的低氧通气反应(HVR),主要是通过促进呼吸频率响应。Orexin神经元投射到孤束核(nTS)和下丘脑室旁核(PVN)。PVN对PCR有重要贡献,并含有nTS-促肾上腺皮质激素释放激素(CRH)神经元。我们假设在雄性大鼠中,食欲素神经元通过激活nTS投射的CRH神经元来促进PCR。我们使用神经元束追踪和免疫组织化学(IHC)来量化缺氧激活PVN投射食欲素神经元的程度。我们将其与食欲素受体(OxR)阻断与suvorexant(Suvo,20mg/kg,i.p.)评估食欲素促进PVN中CRH神经元缺氧诱导激活的程度,包括那些投射到nTS的。在不同的大鼠组中,我们测量了系统性食欲素1受体(Ox1R)阻断(SB-334867;1mg/kg)和PVN中特定Ox1R敲除后的PCR。用Suvo阻断OxR减少了缺氧激活的nTS和PVN神经元的数量,包括那些投射到nTS的CRH神经元。低氧增加了活化的PVN-投射食欲素神经元的数量,但对活化的nTS-投射食欲素神经元的数量没有影响。PVN中的全局Ox1R阻断和部分Ox1R敲除显著降低了PCR。Ox1R敲除还减少了nTS中活化的PVN神经元的数量和活化的酪氨酸-羟化酶神经元的数量。我们的发现表明,食欲素通过表达Ox1R的nTS投射CRH神经元促进PCR。重要性陈述先前我们表明食欲素有助于外周化学反射(PCR),但是这种效应的潜在机制仍然未知。在这里,我们表明:1)食欲素受体阻断减少了PVN和nTS的激活;2)缺氧激活了投射到PVN的食欲素神经元,但不是那些投射到nTS的;3)食欲素受体阻断减少了PVN中nTS-投射促肾上腺皮质激素释放激素(CRH)神经元的激活;4)食欲素1受体(Ox1R)阻断和PVN中特定的Ox1R敲低降低了PCR的强度,和5)Ox1R敲除减少nTS中活化的PVN神经元和酪氨酸羟化酶神经元的数量。这些发现表明,PVN-投射食欲素神经元通过Ox1R促进了nTS-投射CRH神经元上的PCR。
    We previously showed that orexin neurons are activated by hypoxia and facilitate the peripheral chemoreflex (PCR)-mediated hypoxic ventilatory response (HVR), mostly by promoting the respiratory frequency response. Orexin neurons project to the nucleus of the solitary tract (nTS) and the paraventricular nucleus of the hypothalamus (PVN). The PVN contributes significantly to the PCR and contains nTS-projecting corticotropin-releasing hormone (CRH) neurons. We hypothesized that in male rats, orexin neurons contribute to the PCR by activating nTS-projecting CRH neurons. We used neuronal tract tracing and immunohistochemistry (IHC) to quantify the degree that hypoxia activates PVN-projecting orexin neurons. We coupled this with orexin receptor (OxR) blockade with suvorexant (Suvo, 20 mg/kg, i.p.) to assess the degree that orexin facilitates the hypoxia-induced activation of CRH neurons in the PVN, including those projecting to the nTS. In separate groups of rats, we measured the PCR following systemic orexin 1 receptor (Ox1R) blockade (SB-334867; 1 mg/kg) and specific Ox1R knockdown in PVN. OxR blockade with Suvo reduced the number of nTS and PVN neurons activated by hypoxia, including those CRH neurons projecting to nTS. Hypoxia increased the number of activated PVN-projecting orexin neurons but had no effect on the number of activated nTS-projecting orexin neurons. Global Ox1R blockade and partial Ox1R knockdown in the PVN significantly reduced the PCR. Ox1R knockdown also reduced the number of activated PVN neurons and the number of activated tyrosine hydroxylase neurons in the nTS. Our findings suggest orexin facilitates the PCR via nTS-projecting CRH neurons expressing Ox1R.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    下丘脑室旁区(AVPPVN)的精氨酸加压素(AVP)神经元介导大多数物种的性别偏见社会行为,包括哺乳动物。在老鼠身上,神经性别差异被认为是在出生前后的关键窗口(胚胎(E)第18天至出生后(P)第2天)建立的,由此来自胎儿睾丸的循环睾酮在性别二态脑区转化为雌激素.这里,在这个关键窗口之前,我们发现AVPPVN神经元在E15.5之前是性别二态的,妊娠双酚A(BPA)暴露永久男性化女性AVPPVN神经元数量,projects,和电生理特性,导致它们在成年期表现出男性样的表型。此外,我们表明,与对照女性相比,男性和暴露于BPA的女性在E11时出生的神经元数量几乎是P0变成AVP+的两倍,表明AVPPVN神经元男性化发生在E11和P0之间。通过证明从E14.5到E15.5男性化的女性AVPPVN神经元数量的外源性雌激素暴露,我们进一步将这一敏感期缩小到神经发生的时间。而从E13.5到E15.5暴露的泛雌激素受体拮抗剂阻断了男性的男性化。最后,我们表明,限制BPA暴露于E7.5-E15.5导致成年女性表现出更多的社会优势,超过控制女性,与男性行为的获得相一致。我们的研究揭示了E11.5到E15.5的雌激素敏感性窗口影响AVPPVN性别分化,这受到产前BPA暴露的影响。
    Arginine vasopressin (AVP) neurons of the hypothalamic paraventricular region (AVPPVN) mediate sex-biased social behaviors across most species, including mammals. In mice, neural sex differences are thought to be established during a critical window around birth ( embryonic (E) day 18 to postnatal (P) day 2) whereby circulating testosterone from the fetal testis is converted to estrogen in sex-dimorphic brain regions. Here, we found that AVPPVN neurons are sexually dimorphic by E15.5, prior to this critical window, and that gestational bisphenol A (BPA) exposure permanently masculinized female AVPPVN neuronal numbers, projections, and electrophysiological properties, causing them to display male-like phenotypes into adulthood. Moreover, we showed that nearly twice as many neurons that became AVP+ by P0 were born at E11 in males and BPA-exposed females compared to control females, suggesting that AVPPVN neuronal masculinization occurs between E11 and P0. We further narrowed this sensitive period to around the timing of neurogenesis by demonstrating that exogenous estrogen exposure from E14.5 to E15.5 masculinized female AVPPVN neuronal numbers, whereas a pan-estrogen receptor antagonist exposed from E13.5 to E15.5 blocked masculinization of males. Finally, we showed that restricting BPA exposure to E7.5-E15.5 caused adult females to display increased social dominance over control females, consistent with an acquisition of male-like behaviors. Our study reveals an E11.5 to E15.5 window of estrogen sensitivity impacting AVPPVN sex differentiation, which is impacted by prenatal BPA exposure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    高血压是钙调磷酸酶抑制剂的主要不良反应,如他克莫司(FK506)和环孢菌素,临床上用作免疫抑制剂。钙调磷酸酶抑制剂诱导的高血压(CIH)与下丘脑室旁核(PVN)的交感神经输出增加有关。GluA2缺乏,Ca2+-渗透性AMPAR受体(CP-AMPAR)是谷氨酸能突触可塑性的关键特征,然而它们在CIH中的作用仍然难以捉摸。这里,我们发现在大鼠中全身给药FK506显著增加PVN突触体中GluA1和GluA2的丝氨酸磷酸化。引人注目的是,FK506处理减少了来自PVN的突触体和内质网富集部分中的GluA1/GluA2异聚体。用IEM-1460阻断CP-AMPAR诱导AMPAR介导的兴奋性突触后电流(AMPAR-EPSC)振幅在逆行标记中更大的降低,FK506处理的大鼠的脊髓突出的PVN神经元比载体处理的大鼠。此外,FK506处理使标记的PVN神经元中AMPAR-EPSC的电流-电压关系从线性整流转变为向内整流。FK506处理大大增强了PVN中α2δ-1与GluA1和GluA2的物理相互作用。加巴喷丁抑制α2δ-1,α2δ-1基因敲除,或破坏α2δ-1-AMPAR与α2δ-1C末端肽的相互作用,可恢复PVN中的GluA1/GluA2异聚体,并减少FK506处理诱导的标记PVN神经元中AMPAR-EPSC的内向整流。此外,将IEM-1460或α2δ-1C末端肽显微注射入PVN可减少FK506治疗的大鼠的肾交感神经放电和动脉血压升高,但在媒介物治疗的大鼠中则没有。因此,下丘脑中的钙调磷酸酶通过控制GluA1/GluA2与α2δ-1的相互作用来组成型调节AMPAR亚基组成和表型。PVN前交感神经元中的突触CP-AMPAR有助于增加CIH中的交感神经流出。关键词:钙调磷酸酶抑制剂的全身治疗可增加PVN中突触GluA1和GluA2的丝氨酸磷酸化。钙调磷酸酶抑制增强PVN前交感神经元突触后Ca2+通透性AMPARs的发生率。钙调磷酸酶抑制增强了α2δ-1与GluA1和GluA2的相互作用,破坏了PVN中GluA1/GluA2异源四聚体的细胞内组装。阻断PVN中的Ca2+通透性AMPAR或α2δ-1-AMPAR相互作用减弱钙调磷酸酶抑制剂增强的交感神经流出。
    Hypertension is a major adverse effect of calcineurin inhibitors, such as tacrolimus (FK506) and cyclosporine, used clinically as immunosuppressants. Calcineurin inhibitor-induced hypertension (CIH) is linked to augmented sympathetic output from the hypothalamic paraventricular nucleus (PVN). GluA2-lacking, Ca2+-permeable AMPA receptors (CP-AMPARs) are a key feature of glutamatergic synaptic plasticity, yet their role in CIH remains elusive. Here, we found that systemic administration of FK506 in rats significantly increased serine phosphorylation of GluA1 and GluA2 in PVN synaptosomes. Strikingly, FK506 treatment reduced GluA1/GluA2 heteromers in both synaptosomes and endoplasmic reticulum-enriched fractions from the PVN. Blocking CP-AMPARs with IEM-1460 induced a larger reduction of AMPAR-mediated excitatory postsynaptic current (AMPAR-EPSC) amplitudes in retrogradely labelled, spinally projecting PVN neurons in FK506-treated rats than in vehicle-treated rats. Furthermore, FK506 treatment shifted the current-voltage relationship of AMPAR-EPSCs from linear to inward rectification in labelled PVN neurons. FK506 treatment profoundly enhanced physical interactions of α2δ-1 with GluA1 and GluA2 in the PVN. Inhibiting α2δ-1 with gabapentin, α2δ-1 genetic knockout, or disrupting α2δ-1-AMPAR interactions with an α2δ-1 C terminus peptide restored GluA1/GluA2 heteromers in the PVN and diminished inward rectification of AMPAR-EPSCs in labelled PVN neurons induced by FK506 treatment. Additionally, microinjection of IEM-1460 or α2δ-1 C terminus peptide into the PVN reduced renal sympathetic nerve discharges and arterial blood pressure elevated in FK506-treated rats but not in vehicle-treated rats. Thus, calcineurin in the hypothalamus constitutively regulates AMPAR subunit composition and phenotypes by controlling GluA1/GluA2 interactions with α2δ-1. Synaptic CP-AMPARs in PVN presympathetic neurons contribute to augmented sympathetic outflow in CIH. KEY POINTS: Systemic treatment with the calcineurin inhibitor increases serine phosphorylation of synaptic GluA1 and GluA2 in the PVN. Calcineurin inhibition enhances the prevalence of postsynaptic Ca2+-permeable AMPARs in PVN presympathetic neurons. Calcineurin inhibition potentiates α2δ-1 interactions with GluA1 and GluA2, disrupting intracellular assembly of GluA1/GluA2 heterotetramers in the PVN. Blocking Ca2+-permeable AMPARs or α2δ-1-AMPAR interactions in the PVN attenuates sympathetic outflow augmented by the calcineurin inhibitor.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    胰高血糖素样肽-1(GLP-1)及其类似物广泛用于治疗糖尿病。室旁核(PVN)对于调节心血管活动至关重要。这项研究旨在确定GLP-1及其受体(GLP-1R)在PVN中调节交感神经流出和血压的作用。实验在雄性正常血压大鼠和自发性高血压大鼠(SHR)中进行。记录肾交感神经活动(RSNA)和平均动脉压(MAP)。GLP-1和GLP-1R表达存在于PVN中。PVN微量注射GLP-1R激动剂重组人GLP-1(rhGLP-1)或EX-4可增加RSNA和MAP,通过GLP-1R拮抗剂EX9-39或GLP-1R拮抗剂1,超氧化物清除剂tempol,抗氧化剂N-乙酰半胱氨酸,NADPH氧化酶抑制剂apocynin,腺苷酸环化酶抑制剂SQ22536或蛋白激酶A(PKA)抑制剂H89。PVN微量注射rhGLP-1增加了超氧化物的产生,NADPH氧化酶活性,cAMP级别,腺苷酸环化酶和PKA活性,由SQ22536或H89阻止。GLP-1和GLP-1R在SHR的PVN中上调。PVN微量注射GLP-1激动剂可增加WKY和SHR的RSNA和MAP,但与WKY相比,GLP-1拮抗剂在SHR降低RSNA和MAP方面的作用更大。GLP-1R激动剂增强了SHRPVN中超氧化物产生和NADPH氧化酶活性的增加,但被GLP-1R拮抗剂减毒。这些结果表明,PVN中GLP-1R的激活通过cAMP-PKA介导的NADPH氧化酶激活和随后的超氧化物产生增加了交感神经流出和血压。PVN中的GLP-1和GLP-1R上调部分导致交感神经过度活跃和高血压。意义声明:GLP-1受体在室旁核(PVN)的激活增加交感神经活动和血压,由cAMP-PKA-NADPH氧化酶-超氧化物的产生介导。在PVN中干预GLP-1受体可能在减轻交感神经过度活跃和高血压方面发挥有益作用。在高血压患者的GLP-1受体激动剂治疗过程中,中枢作用可能会产生重大影响。
    Glucagon-like peptide-1 (GLP-1) and its analogs are widely used for diabetes treatment. The paraventricular nucleus (PVN) is crucial for regulating cardiovascular activity. This study aims to determine the roles of GLP-1 and its receptors (GLP-1R) in the PVN in regulating sympathetic outflow and blood pressure. Experiments were carried out in male normotensive rats and spontaneously hypertensive rats (SHR). Renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) were recorded. GLP-1 and GLP-1R expressions were present in the PVN. PVN microinjection of GLP-1R agonist recombinant human GLP-1 (rhGLP-1) or EX-4 increased RSNA and MAP, which were prevented by GLP-1R antagonist exendin 9-39 (EX9-39) or GLP-1R antagonist 1, superoxide scavenger tempol, antioxidant N-acetylcysteine, NADPH oxidase (NOX) inhibitor apocynin, adenylyl cyclase (AC) inhibitor SQ22536 or protein kinase A (PKA) inhibitor H89. PVN microinjection of rhGLP-1 increased superoxide production, NADPH oxidase activity, cAMP level, AC, and PKA activity, which were prevented by SQ22536 or H89. GLP-1 and GLP-1R were upregulated in the PVN of SHR. PVN microinjection of GLP-1 agonist increased RSNA and MAP in both WKY and SHR, but GLP-1 antagonists caused greater effects in reducing RSNA and MAP in SHR than in WKY. The increased superoxide production and NADPH oxidase activity in the PVN of SHR were augmented by GLP-1R agonists but attenuated by GLP-1R antagonists. These results indicate that activation of GLP-1R in the PVN increased sympathetic outflow and blood pressure via cAMP-PKA-mediated NADPH oxidase activation and subsequent superoxide production. GLP-1 and GLP-1R upregulation in the PVN partially contributes to sympathetic overactivity and hypertension.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    氧合蛋白能传递阻断外周的伤害感受,脊柱,和通过催产素受体(OTR)的脊柱上水平。的确,已经描述了从下丘脑室旁核(PVN)到脊髓和三叉神经尾核(Sp5c)的神经元途径。因此,虽然三叉子宫颈复合体(TCC),跨越Sp5c的解剖区域,C1和C2区域,在与颅面结构相关的一些疼痛障碍中起作用(例如,偏头痛),在这一水平上,催产素能传递在调节伤害性感受中的作用研究甚少。因此,在雄性Wistar大鼠中进行对眶周或脑膜区域刺激敏感的TCC宽动态范围(WDR)细胞的体内电生理记录。PVN电刺激减少了眶周或脑膜电刺激引起的神经元放电;局部施用的OTR拮抗剂可以逆转这种抑制作用。因此,观察到从PVN到充满神经生物素的WDR细胞的神经元投射(使用氟红宝石)。此外,在充满神经生物素的WDR细胞附近发现OTR和CGRP或OTR和GABA之间的共定位。沉积在脑膜(真蓝色)和眶下神经(氟金)的逆行神经元示踪剂显示,在三叉神经节(TG),一些细胞对两种荧光团免疫阳性,表明某些TG细胞通过V1和V2三叉神经分支发送投影。一起,这些数据可能暗示内源性催氧能传递通过CGRPergic(初级传入纤维)和GABA能细胞中的OTR激活抑制了二级神经元的伤害性活动.意义陈述这项研究揭示了三叉神经伤害感受调节所涉及的机制,这对于理解原发性头痛的病理生理学至关重要,比如偏头痛。目前的证据表明,下丘脑在控制三叉神经水平的伤害性活动中起作用。本研究发现,下丘脑室旁核(PVN)的电刺激通过催氧能机制抑制三叉神经二级WDR细胞的伤害性活动。此外,我们发现催产素受体位于初级肽能传入纤维和GABA能细胞中。这些发现支持了PVN和TCC之间的直接催产素能途径的想法,并强调了催产素受体作为偏头痛和其他原发性头痛的靶标的潜力。
    Oxytocinergic transmission blocks nociception at the peripheral, spinal, and supraspinal levels through the oxytocin receptor (OTR). Indeed, a neuronal pathway from the hypothalamic paraventricular nucleus (PVN) to the spinal cord and trigeminal nucleus caudalis (Sp5c) has been described. Hence, although the trigeminocervical complex (TCC), an anatomical area spanning the Sp5c, C1, and C2 regions, plays a role in some pain disorders associated with craniofacial structures (e.g., migraine), the role of oxytocinergic transmission in modulating nociception at this level has been poorly explored. Hence, in vivo electrophysiological recordings of TCC wide dynamic range (WDR) cells sensitive to stimulation of the periorbital or meningeal region were performed in male Wistar rats. PVN electrical stimulation diminished the neuronal firing evoked by periorbital or meningeal electrical stimulation; this inhibition was reversed by OTR antagonists administered locally. Accordingly, neuronal projections (using Fluoro-Ruby) from the PVN to the WDR cells filled with Neurobiotin were observed. Moreover, colocalization between OTR and calcitonin gene-related peptide (CGRP) or OTR and GABA was found near Neurobiotin-filled WDR cells. Retrograde neuronal tracers deposited at the meningeal (True-Blue, TB) and infraorbital nerves (Fluoro-Gold, FG) showed that at the trigeminal ganglion (TG), some cells were immunopositive to both fluorophores, suggesting that some TG cells send projections via the V1 and V2 trigeminal branches. Together, these data may imply that endogenous oxytocinergic transmission inhibits the nociceptive activity of second-order neurons via OTR activation in CGRPergic (primary afferent fibers) and GABAergic cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    通过雄激素受体(AR)和雌激素受体α(ERα)的性腺激素作用调节下丘脑-垂体-肾上腺(HPA)轴反应性和应激相关行为的性别差异。在这里,我们测试了促肾上腺皮质激素释放因子(CRF)是否表达神经元,众所周知,它们可以调节神经内分泌和行为应激反应,共表达AR和ERα作为性腺激素调节这些反应的潜在机制。使用Crh-IRES-Cre::Ai9报告小鼠,我们报告了内侧视前区(MPOA)内CRF神经元中AR的高度共定位,终末纹床核(BST),杏仁核内侧(MeA),和腹内侧下丘脑(VMH),杏仁核中央(CeA)内的中等水平和室旁下丘脑(PVN)中的低水平。在BST(BSTmpl)的主核中发现了CRF/AR共表达的性别差异,CeA,MeA,和VMH(男性>女性)。CRF与ERα的共定位相对于AR共定位通常较低。然而,在MPOA中发现了高共表达,AVPV,和VMH,在弓状核(ARC)中适度共表达,BST,和MeA和低水平的PVN和CeA。在BSTmpl和PVN中发现了CRF/ERα共定位的性别差异(男性>女性)。最后,我们在束缚应激小鼠中评估了CRF神经元的神经激活,并在BSTmpl和导水管周围灰中发现了更大的CRF/c-Fos共表达。而男性在ARC和背侧CA1中的共表达较高。鉴于CRF在调节行为应激反应和HPA轴方面的已知作用,AR/ERα共表达和CRF细胞组的性别特异性激活表明了调节这些功能中性别差异的潜在机制。
    Gonadal hormone actions through androgen receptor (AR) and estrogen receptor alpha (ERα) regulate sex differences in hypothalamic-pituitary-adrenal (HPA) axis responsivity and stress-related behaviors. Here we tested whether corticotropin releasing factor (CRF) expressing neurons, which are widely known to regulate neuroendocrine and behavioral stress responses, co-express AR and ERα as a potential mechanism for gonadal hormone regulation of these responses. Using Crh-IRES-Cre::Ai9 reporter mice we report high co-localization of AR in CRF neurons within the medial preoptic area (MPOA), bed nucleus of the stria terminalis (BST), medial amygdala (MeA), and ventromedial hypothalamus (VMH), moderate levels within the central amygdala (CeA) and low levels in the paraventricular hypothalamus (PVN). Sex differences in CRF/AR co-expression were found in the principal nucleus of the BST (BSTmpl), CeA, MeA, and VMH (males>females). CRF co-localization with ERα was generally lower relative to AR co-localization. However, high co-expression was found within the MPOA, AVPV, and VMH, with moderate co-expression in the arcuate nucleus (ARC), BST, and MeA and low levels in the PVN and CeA. Sex differences in CRF/ERα co-localization were found in the BSTmpl and PVN (males>females). Finally, we assessed neural activation of CRF neurons in restraint-stressed mice and found greater CRF/c-Fos co-expression in females in the BSTmpl and periaqueductal gray, while co-expression was higher in males within the ARC and dorsal CA1. Given the known role of CRF in regulating behavioral stress responses and the HPA axis, AR/ERα co-expression and sex-specific activation of CRF cell groups indicate potential mechanisms for modulating sex differences in these functions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    应激对生殖功能的影响是显着的。下丘脑室旁核(PVN)促肾上腺皮质激素释放激素(CRH)在调节应激反应中起主要作用。了解下丘脑-垂体-肾上腺(HPA)轴和下丘脑-垂体-性腺(HPG)轴如何相互作用对于理解压力如何导致生殖功能障碍至关重要。然而,应激是否通过调节PVNCRH或HPA后遗症影响生殖功能尚未完全阐明。
    在这项研究中,我们研究了PVNCRH神经元的化学激活对生殖功能的影响。我们使用由设计药物(DREADD)病毒构建体专门激活的兴奋性设计受体,长期和选择性地刺激雌性CRH-Cre小鼠中的PVNCRH神经元,两侧注射到PVN中。激动剂化合物-21(C21)通过饮用水递送。我们确定了PVNCRH神经元DREADDs激活对发情周期的影响,动情期和动情期的LH脉冲频率和动情期小鼠的LH激增。还检查了长期C21给药对基础皮质酮分泌的影响以及对动情期急性束缚应激的反应。此外,使用数学模型的计算机模拟来确定PVNCRH神经元DREADDs激活的影响,模拟慢性压力,通过实验检查的生理参数。
    因此,与我们的数学模型预测一致,发情周期的长度延长了,随着发情和动情时间的增加,发情期和发情期减少。此外,动情期LH脉冲的频率降低,但在动情期不受影响。发情期间排卵前LH激增的发生被破坏。动情期皮质酮的基础水平没有受到影响,但长期应用C21后,急性束缚应激反应减弱.
    这些数据表明,PVNCRH神经元在破坏卵巢周期和排卵前LH激增中起着功能性作用,根据我们的数学模型预测,GnRH脉冲发生器的活动在动情期保持相对稳健,而在慢性压力暴露下的动情期则不稳定。
    The impact of stress on reproductive function is significant. Hypothalamic paraventricular nucleus (PVN) corticotrophin-releasing hormone (CRH) plays a major role in regulating the stress response. Understanding how the hypothalamic-pituitary-adrenal (HPA) axis and the hypothalamic-pituitary-gonadal (HPG) axis interact is crucial for comprehending how stress can lead to reproductive dysfunction. However, whether stress influences reproductive function via modulating PVN CRH or HPA sequelae is not fully elucidated.
    In this study, we investigated the impact of chemogenetic activation of PVN CRH neurons on reproductive function. We chronically and selectively stimulated PVN CRH neurons in female CRH-Cre mice using excitatory designer receptor exclusively activated by designer drugs (DREADDs) viral constructs, which were bilaterally injected into the PVN. The agonist compound-21 (C21) was delivered through the drinking water. We determined the effects of DREADDs activation of PVN CRH neurons on the estrous cycles, LH pulse frequency in diestrus and metestrus and LH surge in proestrus mice. The effect of long-term C21 administration on basal corticosterone secretion and the response to acute restraint stress during metestrus was also examined. Additionally, computer simulations of a mathematical model were used to determine the effects of DREADDs activation of PVN CRH neurons, simulating chronic stress, on the physiological parameters examined experimentally.
    As a result, and consistent with our mathematical model predictions, the length of the estrous cycle was extended, with an increase in the time spent in estrus and metestrus, and a decrease in proestrus and diestrus. Additionally, the frequency of LH pulses during metestrus was decreased, but unaffected during diestrus. The occurrence of the preovulatory LH surge during proestrus was disrupted. The basal level of corticosterone during metestrus was not affected, but the response to acute restraint stress was diminished after long-term C21 application.
    These data suggest that PVN CRH neurons play a functional role in disrupting ovarian cyclicity and the preovulatory LH surge, and that the activity of the GnRH pulse generator remains relatively robust during diestrus but not during metestrus under chronic stress exposure in accordance with our mathematical model predictions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:外周炎性细胞因子的升高已被确定为心力衰竭(HF)中神经炎症和交感神经过度活动的积极贡献者。然而,这些细胞因子破坏血脑屏障(BBB)对大脑发挥作用的确切机制仍然难以捉摸。白细胞介素17A与各种神经系统疾病中的BBB破坏有关,在HF的循环和大脑中,其水平显着增加。本研究旨在确定BBB完整性是否在下丘脑室旁核(PVN)内受损,如果是这样,白细胞介素17A是否有助于心肌梗死诱导的HF中的BBB破坏。
    结果:雄性Sprague-Dawley大鼠接受冠状动脉结扎以诱导HF或假手术。一些HF大鼠接受了白介素17受体A小干扰RNA或乱序小干扰RNA腺相关病毒的双侧PVN显微注射。冠状动脉结扎四周后,通过颈动脉内注射荧光染料(异硫氰酸荧光素-葡聚糖10kDa+罗丹明-葡聚糖70kDa)评估BBB的通透性.与假手术大鼠相比,HF大鼠在PVN中表现出异硫氰酸荧光素-葡聚糖10kDa的外渗,但在大脑皮层中没有。血浆白细胞介素17A水平与PVN中异硫氰酸荧光素10kDa外渗呈正相关。caveolin-1的表达,一种胞吞标记,被增强了,而HF大鼠中紧密连接蛋白的表达减少。在PVN微血管的内皮内鉴定出白介素17受体A。用白细胞介素17受体治疗一种小干扰RNA导致PVN中异硫氰酸荧光素10kDa外渗的显着减弱,并逆转PVN中caveolin-1和紧密连接相关蛋白的表达。
    结论:总的来说,这些数据表明,在HF中,PVN内的BBB通透性增强,并且可能归因于BBB内皮中白介素17A/白介素17受体A信号传导的增加,通过促进空穴胞吞作用和紧密连接复合物的降解。
    BACKGROUND: Elevated inflammatory cytokines in the periphery have been identified as active contributors to neuroinflammation and sympathetic overactivity in heart failure (HF). Yet, the exact mechanisms by which these cytokines breach the blood-brain barrier (BBB) to exert their effects on the brain remain elusive. Interleukin 17A has been linked to BBB disruption in various neurologic disorders, and its levels were significantly augmented in circulation and the brain in HF. The present study aimed to determine whether the BBB integrity was compromised within the hypothalamic paraventricular nucleus (PVN), and if so, whether interleukin 17A contributes to BBB disruption in myocardial infarction-induced HF.
    RESULTS: Male Sprague-Dawley rats underwent coronary artery ligation to induce HF or sham surgery. Some HF rats received bilateral PVN microinjections of an interleukin 17 receptor A small interfering RNA or a scrambled small interfering RNA adeno-associated virus. Four weeks after coronary artery ligation, the permeability of the BBB was evaluated by intracarotid injection of fluorescent dyes (fluorescein isothiocyanate-dextran 10 kDa+rhodamine-dextran 70 kDa). Compared with sham-operated rats, HF rats exhibited an elevated extravasation of fluorescein isothiocyanate-dextran 10 kDa within the PVN but not in the brain cortex. The plasma interleukin 17A levels were positively correlated with fluorescein isothiocyanate 10 kDa extravasation in the PVN. The expression of caveolin-1, a transcytosis marker, was augmented, whereas the expression of tight junction proteins was diminished in HF rats. Interleukin 17 receptor A was identified within the endothelium of PVN microvessels. Treatment with interleukin 17 receptor A small interfering RNA led to a significant attenuation of fluorescein isothiocyanate 10 kDa extravasation in the PVN and reversed expression of caveolin-1 and tight junction-associated proteins in the PVN.
    CONCLUSIONS: Collectively, these data indicate that BBB permeability within the PVN is enhanced in HF and is likely attributable to increased interleukin 17A/interleukin 17 receptor A signaling in the BBB endothelium, by promoting caveolar transcytosis and degradation of tight junction complexes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号