首页 > 最新文献

Journal of Neuroinflammation最新文献

英文 中文
Microglia ameliorate delirium-like phenotypes in a murine model of acute ventilator-induced lung injury. 小胶质细胞可改善急性呼吸机诱发肺损伤小鼠模型中的谵妄样表型。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-21 DOI: 10.1186/s12974-024-03260-y
Landon Scott, Kevin D Winzey, Debbie Moreira, Catherine Bresee, Jean-Philippe Vit, Warren G Tourtellotte, S Ananth Karumanchi, Shouri Lahiri

Background: Delirium affects 50-85% of patients on mechanical ventilation and is associated with increased mortality, prolonged hospitalization, and a three-fold higher risk of dementia. Microglia, the resident immune cells of the brain, exhibit both neuroprotective and neurotoxic functions; however, their effects in mechanical ventilation-induced acute lung injury (VILI) are unknown. We hypothesize that in a model of short-term VILI, microglia play a neuroprotective role to ameliorate delirium-like phenotypes.

Methods: Microglia depletion (n = 18) was accomplished using an orally administered colony stimulating factor 1 receptor inhibitor, while controls received a vehicle diet (n = 18). We then compared extent of neuronal injury in the frontal cortex and hippocampus using cleaved caspase-3 (CC3) and multiple delirium-like behaviors in microglia depleted and non-microglia depleted male mice (C57BL/6 J aged 4-9 months) following VILI. Delirium-like behaviors were evaluated using the Open Field, Elevated Plus Maze, and Y-maze assays. We subsequently evaluated whether repopulation of microglia (n = 14 repopulation, 14 vehicle) restored the phenotypes.

Results: Frontal/hippocampal neuronal CC3 levels were significantly higher in microglia depleted VILI mice compared to vehicle-treated VILI controls (p < 0.01, p < 0.01, respectively). These structural changes were accompanied by worse delirium-like behaviors in microglia depleted VILI mice compared to vehicle controls. Specifically, microglia depleted VILI mice demonstrated: (1) significantly increased time in the periphery of the Open Field (p = 0.01), (2) significantly increased coefficient of variation (p = 0.02), (3) trend towards reduced time in the open arms of the Elevated Plus Maze (p = 0.09), and (4) significantly decreased spontaneous alternations on Y-maze (p < 0.01). There was a significant inverse correlation between frontal CC3 and percent spontaneous alternations (R2 = 0.51, p < 0.01). Microglia repopulation showed a near-complete return to vehicle levels of delirium like-behaviors.

Conclusions: This study demonstrates that microglia depletion exacerbates structural and functional delirium-like phenotypes after VILI, while subsequent repopulation of microglia restores these phenotypes. These findings suggest a neuroprotective role for microglia in ameliorating neuronal and functional delirium-like phenotypes and call for consideration of interventions that leverage endogenous microglia physiology to mitigate delirium.

背景:50%-85%的机械通气患者会出现谵妄,这与死亡率增加、住院时间延长以及痴呆风险增加三倍有关。小胶质细胞是大脑中的常驻免疫细胞,具有神经保护和神经毒性两种功能;然而,它们在机械通气诱导的急性肺损伤(VILI)中的作用尚不清楚。我们假设,在短期 VILI 模型中,小胶质细胞可发挥神经保护作用,改善谵妄样表型:方法:使用口服集落刺激因子 1 受体抑制剂来消耗小胶质细胞(n = 18),而对照组则接受车辆饮食(n = 18)。然后,我们使用裂解的 Caspase-3 (CC3) 比较了额叶皮层和海马的神经元损伤程度,并比较了小胶质细胞耗竭和非小胶质细胞耗竭雄性小鼠(C57BL/6 J,4-9 个月)在 VILI 后的多种谵妄样行为。谵妄样行为是通过开阔地、高架正迷宫和Y迷宫试验进行评估的。我们随后评估了小胶质细胞再填充(n = 14 个再填充,14 个车辆)是否能恢复表型:结果:与药物治疗的 VILI 对照组相比,小胶质细胞耗竭的 VILI 小鼠额叶/海马神经元 CC3 水平显著升高(p 2 = 0.51,p 结论:小胶质细胞耗竭的 VILI 小鼠额叶/海马神经元 CC3 水平显著升高:本研究表明,小胶质细胞耗竭会加重 VILI 后的结构性和功能性谵妄样表型,而随后小胶质细胞的重新填充则会恢复这些表型。这些研究结果表明,小胶质细胞在改善神经元和功能性谵妄样表型方面具有神经保护作用,因此需要考虑利用内源性小胶质细胞生理学来减轻谵妄的干预措施。
{"title":"Microglia ameliorate delirium-like phenotypes in a murine model of acute ventilator-induced lung injury.","authors":"Landon Scott, Kevin D Winzey, Debbie Moreira, Catherine Bresee, Jean-Philippe Vit, Warren G Tourtellotte, S Ananth Karumanchi, Shouri Lahiri","doi":"10.1186/s12974-024-03260-y","DOIUrl":"10.1186/s12974-024-03260-y","url":null,"abstract":"<p><strong>Background: </strong>Delirium affects 50-85% of patients on mechanical ventilation and is associated with increased mortality, prolonged hospitalization, and a three-fold higher risk of dementia. Microglia, the resident immune cells of the brain, exhibit both neuroprotective and neurotoxic functions; however, their effects in mechanical ventilation-induced acute lung injury (VILI) are unknown. We hypothesize that in a model of short-term VILI, microglia play a neuroprotective role to ameliorate delirium-like phenotypes.</p><p><strong>Methods: </strong>Microglia depletion (n = 18) was accomplished using an orally administered colony stimulating factor 1 receptor inhibitor, while controls received a vehicle diet (n = 18). We then compared extent of neuronal injury in the frontal cortex and hippocampus using cleaved caspase-3 (CC3) and multiple delirium-like behaviors in microglia depleted and non-microglia depleted male mice (C57BL/6 J aged 4-9 months) following VILI. Delirium-like behaviors were evaluated using the Open Field, Elevated Plus Maze, and Y-maze assays. We subsequently evaluated whether repopulation of microglia (n = 14 repopulation, 14 vehicle) restored the phenotypes.</p><p><strong>Results: </strong>Frontal/hippocampal neuronal CC3 levels were significantly higher in microglia depleted VILI mice compared to vehicle-treated VILI controls (p < 0.01, p < 0.01, respectively). These structural changes were accompanied by worse delirium-like behaviors in microglia depleted VILI mice compared to vehicle controls. Specifically, microglia depleted VILI mice demonstrated: (1) significantly increased time in the periphery of the Open Field (p = 0.01), (2) significantly increased coefficient of variation (p = 0.02), (3) trend towards reduced time in the open arms of the Elevated Plus Maze (p = 0.09), and (4) significantly decreased spontaneous alternations on Y-maze (p < 0.01). There was a significant inverse correlation between frontal CC3 and percent spontaneous alternations (R<sup>2</sup> = 0.51, p < 0.01). Microglia repopulation showed a near-complete return to vehicle levels of delirium like-behaviors.</p><p><strong>Conclusions: </strong>This study demonstrates that microglia depletion exacerbates structural and functional delirium-like phenotypes after VILI, while subsequent repopulation of microglia restores these phenotypes. These findings suggest a neuroprotective role for microglia in ameliorating neuronal and functional delirium-like phenotypes and call for consideration of interventions that leverage endogenous microglia physiology to mitigate delirium.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"270"},"PeriodicalIF":9.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11495074/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467941","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
MOG-specific CAR Tregs: a novel approach to treat multiple sclerosis. MOG 特异性 CAR Tregs:治疗多发性硬化症的新方法。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-20 DOI: 10.1186/s12974-024-03262-w
Jihane Frikeche, Marion David, Xavier Mouska, Damien Treguer, Yue Cui, Sandrine Rouquier, Enora Lecorgne, Emma Proics, Papa Babacar Fall, Audrey Lafon, Gregory Lara, Alexandra Menardi, David Fenard, Tobias Abel, Julie Gertner-Dardenne, Maurus de la Rosa, Celine Dumont

Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system (CNS) with the immune system attacking myelin sheaths leading to neuronal death. While several disease-modifying therapies are available to treat MS, these therapies are not universally effective and do not stop disease progression. More personalized long-term treatment options that target specific aspects of the disease, such as reducing relapse frequency, delaying disability accumulation, and addressing symptoms that impact daily functioning, as well as therapies that can promote neuroprotection and repair are needed. Chimeric Antigen Receptor (CAR) Tcell therapies have revolutionized cancer treatment by intravenously (IV) administering a defined dose of T cells with high specificity provided by the CAR. An autologous CAR T cell therapy using suppressive regulatory T cells (Tregs) inducing long-lasting tolerance would be the ideal treatment for patients. Hence, we expanded the application of CAR-T cells by introducing a CAR into Tregs to treat MS patients. We developed a myelin oligodendrocyte glycoprotein (MOG)-specific CAR Treg cell therapy for patients with MS. MOG is expressed on the outer membrane of the myelin sheath, the insulating layer the forms around nerves, making it an ideal target for CAR Treg therapy. Our lead candidate is a 2nd generation CAR, composed of an anti-MOG scFv screened from a large human library. In vitro, we demonstrated CAR-dependent functionality and showed efficacy in vivo using a passive EAE mouse model. Additionally, the MOG-CAR Tregs have very low tonic signaling with a desirable signal-to-noise ratio resulting in a highly potent CAR. In summary our data suggest that MOG-CAR Tregs are a promising MS treatment option with the potential to induce long-lasting tolerance in patients.

多发性硬化症(MS)是一种影响中枢神经系统(CNS)的自身免疫性疾病,免疫系统攻击髓鞘导致神经元死亡。虽然有几种改变病情的疗法可用于治疗多发性硬化症,但这些疗法并非普遍有效,也不能阻止疾病的发展。我们需要更多针对疾病特定方面的个性化长期治疗方案,如降低复发频率、延缓残疾累积、解决影响日常功能的症状,以及促进神经保护和修复的疗法。嵌合抗原受体(CAR)T细胞疗法通过静脉注射(IV)给药给定剂量的T细胞,CAR具有高度特异性,从而彻底改变了癌症治疗方法。使用抑制性调节 T 细胞(Tregs)诱导持久耐受的自体 CAR T 细胞疗法将是患者的理想治疗方法。因此,我们扩大了CAR-T细胞的应用范围,将CAR引入Tregs治疗多发性硬化症患者。我们为多发性硬化症患者开发了一种髓鞘少突胶质细胞糖蛋白(MOG)特异性 CAR Treg 细胞疗法。MOG表达于神经周围形成的绝缘层--髓鞘的外膜上,因此是CAR Treg疗法的理想靶点。我们的主要候选药物是第二代 CAR,由从大型人类库中筛选出的抗 MOG scFv 组成。我们在体外证明了 CAR 依赖性功能,并利用被动 EAE 小鼠模型在体内显示了疗效。此外,MOG-CAR Tregs 的强直性信号强度很低,具有理想的信噪比,因此是一种高效的 CAR。总之,我们的数据表明,MOG-CAR Tregs 是一种很有前景的多发性硬化症治疗选择,具有诱导患者产生长期耐受的潜力。
{"title":"MOG-specific CAR Tregs: a novel approach to treat multiple sclerosis.","authors":"Jihane Frikeche, Marion David, Xavier Mouska, Damien Treguer, Yue Cui, Sandrine Rouquier, Enora Lecorgne, Emma Proics, Papa Babacar Fall, Audrey Lafon, Gregory Lara, Alexandra Menardi, David Fenard, Tobias Abel, Julie Gertner-Dardenne, Maurus de la Rosa, Celine Dumont","doi":"10.1186/s12974-024-03262-w","DOIUrl":"10.1186/s12974-024-03262-w","url":null,"abstract":"<p><p>Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system (CNS) with the immune system attacking myelin sheaths leading to neuronal death. While several disease-modifying therapies are available to treat MS, these therapies are not universally effective and do not stop disease progression. More personalized long-term treatment options that target specific aspects of the disease, such as reducing relapse frequency, delaying disability accumulation, and addressing symptoms that impact daily functioning, as well as therapies that can promote neuroprotection and repair are needed. Chimeric Antigen Receptor (CAR) Tcell therapies have revolutionized cancer treatment by intravenously (IV) administering a defined dose of T cells with high specificity provided by the CAR. An autologous CAR T cell therapy using suppressive regulatory T cells (Tregs) inducing long-lasting tolerance would be the ideal treatment for patients. Hence, we expanded the application of CAR-T cells by introducing a CAR into Tregs to treat MS patients. We developed a myelin oligodendrocyte glycoprotein (MOG)-specific CAR Treg cell therapy for patients with MS. MOG is expressed on the outer membrane of the myelin sheath, the insulating layer the forms around nerves, making it an ideal target for CAR Treg therapy. Our lead candidate is a 2nd generation CAR, composed of an anti-MOG scFv screened from a large human library. In vitro, we demonstrated CAR-dependent functionality and showed efficacy in vivo using a passive EAE mouse model. Additionally, the MOG-CAR Tregs have very low tonic signaling with a desirable signal-to-noise ratio resulting in a highly potent CAR. In summary our data suggest that MOG-CAR Tregs are a promising MS treatment option with the potential to induce long-lasting tolerance in patients.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"268"},"PeriodicalIF":9.3,"publicationDate":"2024-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11490997/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467943","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
Enhancing Th17 cells drainage through meningeal lymphatic vessels alleviate neuroinflammation after subarachnoid hemorrhage. 加强 Th17 细胞通过脑膜淋巴管的引流可缓解蛛网膜下腔出血后的神经炎症。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-20 DOI: 10.1186/s12974-024-03252-y
Dandan Gao, Bin Zou, Kunyuan Zhu, Shijun Bi, Wenxu Zhang, Xinyu Yang, Jieyu Lai, Guobiao Liang, Pengyu Pan

Background: Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disorder primarily caused by the rupture of aneurysm, which results in a high mortality rate and consequently imposes a significant burden on society. The occurrence of SAH initiates an immune response that further exacerbates brain damage. The acute inflammatory reaction subsequent to SAH plays a crucial role in determining the prognosis. Th17 cells, a subset of T cells, are related to the brain injury following SAH, and it is unclear how Th17 cells are cleared in the brain. Meningeal lymphatic vessels are a newly discovered intracranial fluid transport system that has been shown to drain large molecules and immune cells to deep cervical lymph nodes. There is limited understanding of the role of the meningeal lymphatic system in SAH. The objective of this research is to explore the impact and underlying mechanism of drainage Th17 cells by meningeal lymphatics on SAH.

Methods: Treatments to manipulate meningeal lymphatic function and the CCR7-CCL21 pathway were administered, including laser ablation, injection of VEGF-C geneknockout, and protein injection. Mouse behavior was assessed using the balance beam experiment and the modified Garcia scoring system. Flow cytometry, enzyme-linked immunosorbent assays (ELISA), and immunofluorescence staining were used to study the impact of meningeal lymphatic on SAH drainage. Select patients with unruptured and ruptured aneurysms in our hospital as the control group and the SAH group, with 7 cases in each group. Peripheral blood and cerebrospinal fluid (CSF) samples were assessed by ELISA and flow cytometry.

Results: Mice with SAH showed substantial behavioral abnormalities and brain damage in which immune cells accumulated in the brain. Laser ablation of the meningeal lymphatic system or knockout of the CCR7 gene leads to Th17 cell aggregation in the meninges, resulting in a decreased neurological function score and increased levels of inflammatory factors. Injection of VEGF-C or CCL21 protein promotes Th17 cell drainage to lymph nodes, an increased neurological function score, and decreased levels of inflammatory factors. Clinical blood and CSF results showed that inflammatory factors in SAH group were significantly increased. The number of Th17 cells in the SAH group was significantly higher than the control group. Clinical results confirmed Th17 cells aggravated the level of neuroinflammation after SAH.

Conclusion: This study shows that improving the drainage of Th17 cells by meningeal lymphatics via the CCR7-CCL21 pathway can reduce brain damage and improve behavior in the SAH mouse model. This could lead to new treatment options for SAH.

背景:蛛网膜下腔出血(SAH蛛网膜下腔出血(SAH)是一种严重的脑血管疾病,主要由动脉瘤破裂引起,死亡率很高,因此给社会带来沉重负担。SAH 的发生会引发免疫反应,进一步加重脑损伤。SAH 后的急性炎症反应对预后起着至关重要的作用。Th17 细胞是 T 细胞的一个亚群,与 SAH 后的脑损伤有关,目前尚不清楚 Th17 细胞如何在脑内清除。脑膜淋巴管是一种新发现的颅内液体运输系统,已被证明能将大分子和免疫细胞引流到颈深淋巴结。人们对脑膜淋巴系统在 SAH 中的作用了解有限。本研究旨在探讨脑膜淋巴管引流 Th17 细胞对 SAH 的影响及其内在机制:方法:对脑膜淋巴功能和CCR7-CCL21通路进行治疗,包括激光消融、注射VEGF-C基因敲除和蛋白注射。使用平衡木实验和改进的加西亚评分系统对小鼠行为进行评估。流式细胞术、酶联免疫吸附试验(ELISA)和免疫荧光染色被用来研究脑膜淋巴对SAH引流的影响。选择本院未破裂和破裂动脉瘤患者作为对照组和 SAH 组,每组 7 例。通过ELISA和流式细胞术对外周血和脑脊液样本进行评估:结果:SAH小鼠表现出严重的行为异常和脑损伤,免疫细胞在脑内聚集。激光消融脑膜淋巴系统或敲除 CCR7 基因会导致 Th17 细胞在脑膜聚集,从而导致神经功能评分下降和炎症因子水平升高。注射 VEGF-C 或 CCL21 蛋白可促进 Th17 细胞向淋巴结引流,提高神经功能评分,降低炎症因子水平。临床血液和脑脊液结果显示,SAH 组的炎症因子明显增加。SAH 组 Th17 细胞数量明显高于对照组。临床结果证实 Th17 细胞加重了 SAH 后神经炎症的程度:本研究表明,通过CCR7-CCL21途径改善脑膜淋巴管对Th17细胞的引流可减轻SAH小鼠模型的脑损伤并改善其行为。这可能为 SAH 带来新的治疗方案。
{"title":"Enhancing Th17 cells drainage through meningeal lymphatic vessels alleviate neuroinflammation after subarachnoid hemorrhage.","authors":"Dandan Gao, Bin Zou, Kunyuan Zhu, Shijun Bi, Wenxu Zhang, Xinyu Yang, Jieyu Lai, Guobiao Liang, Pengyu Pan","doi":"10.1186/s12974-024-03252-y","DOIUrl":"10.1186/s12974-024-03252-y","url":null,"abstract":"<p><strong>Background: </strong>Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disorder primarily caused by the rupture of aneurysm, which results in a high mortality rate and consequently imposes a significant burden on society. The occurrence of SAH initiates an immune response that further exacerbates brain damage. The acute inflammatory reaction subsequent to SAH plays a crucial role in determining the prognosis. Th17 cells, a subset of T cells, are related to the brain injury following SAH, and it is unclear how Th17 cells are cleared in the brain. Meningeal lymphatic vessels are a newly discovered intracranial fluid transport system that has been shown to drain large molecules and immune cells to deep cervical lymph nodes. There is limited understanding of the role of the meningeal lymphatic system in SAH. The objective of this research is to explore the impact and underlying mechanism of drainage Th17 cells by meningeal lymphatics on SAH.</p><p><strong>Methods: </strong>Treatments to manipulate meningeal lymphatic function and the CCR7-CCL21 pathway were administered, including laser ablation, injection of VEGF-C geneknockout, and protein injection. Mouse behavior was assessed using the balance beam experiment and the modified Garcia scoring system. Flow cytometry, enzyme-linked immunosorbent assays (ELISA), and immunofluorescence staining were used to study the impact of meningeal lymphatic on SAH drainage. Select patients with unruptured and ruptured aneurysms in our hospital as the control group and the SAH group, with 7 cases in each group. Peripheral blood and cerebrospinal fluid (CSF) samples were assessed by ELISA and flow cytometry.</p><p><strong>Results: </strong>Mice with SAH showed substantial behavioral abnormalities and brain damage in which immune cells accumulated in the brain. Laser ablation of the meningeal lymphatic system or knockout of the CCR7 gene leads to Th17 cell aggregation in the meninges, resulting in a decreased neurological function score and increased levels of inflammatory factors. Injection of VEGF-C or CCL21 protein promotes Th17 cell drainage to lymph nodes, an increased neurological function score, and decreased levels of inflammatory factors. Clinical blood and CSF results showed that inflammatory factors in SAH group were significantly increased. The number of Th17 cells in the SAH group was significantly higher than the control group. Clinical results confirmed Th17 cells aggravated the level of neuroinflammation after SAH.</p><p><strong>Conclusion: </strong>This study shows that improving the drainage of Th17 cells by meningeal lymphatics via the CCR7-CCL21 pathway can reduce brain damage and improve behavior in the SAH mouse model. This could lead to new treatment options for SAH.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"269"},"PeriodicalIF":9.3,"publicationDate":"2024-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11492769/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467937","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
Myeloid gasdermin D drives early-stage T cell immunity and peripheral inflammation in a mouse model of Alzheimer's disease. 在阿尔茨海默病小鼠模型中,髓质气敏素 D 驱动早期 T 细胞免疫和外周炎症。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-19 DOI: 10.1186/s12974-024-03255-9
Wenjuan Rui, Yuqing Wu, Yongbing Yang, Wenting Xie, Dengli Qin, Jie Ming, Zhihan Ye, Liu Lu, Ming Zong, Xianglong Tang, Lieying Fan, Sheng Li

Background: It is now realized that peripheral inflammation and abnormal immune responses, especially T cells, contribute to the development of Alzheimer's disease (AD). Gasdermin D (GSDMD) -mediated pyroptosis has been associated with several neuroinflammatory diseases, but whether GSDMD is involved in the peripheral inflammation and T cell immunity during AD remains unclear.

Methods: We dynamically investigated GSDMD activation in the peripheral and central nervous system of 5×FAD mouse model and dissected the role of myeloid GSDMD using genetic knockout mice, especially its influence on peripheral T cell responses and AD inflammation. RNA sequencing and in vitro coculture were used to elucidate the underlying immune mechanisms involved. Targeted inhibitor experiments and clinical correlation analysis were used to further verify the function of GSDMD in AD.

Results: In the present study, caspase activated GSDMD in the spleen of 5×FAD mice earlier than in the brain during disease progression. Loss of myeloid cell GSDMD was shown to impair early-stage effector T cell activation in the periphery and prevent T cell infiltration into the brain, with an overall reduction in neuroinflammation. Furthermore, myeloid cell GSDMD induced T cell PD-1 expression through the IL-1β/NF-κB pathway, restricting regulatory T cells. The administration of a GSDMD inhibitor combined with an anti-PD-1 antibody was found to mitigate the development of AD-associated inflammation. In some AD patients, plasma sPD-1 is positively correlated with IL-Iβ and clinical features.

Conclusions: Our study systematically identified a role for GSDMD in the AD-related peripheral inflammation and early-stage T cell immunity. These findings also suggest the therapeutic potential of targeting GSDMD for the early intervention in AD.

背景:现在人们已经意识到,外周炎症和异常免疫反应,尤其是 T 细胞,是阿尔茨海默病(AD)发病的原因之一。由Gasdermin D(GSDMD)介导的热蛋白沉积与多种神经炎症性疾病有关,但GSDMD是否参与了AD期间的外周炎症和T细胞免疫仍不清楚:方法: 我们动态研究了 5×FAD 小鼠模型外周和中枢神经系统中 GSDMD 的活化情况,并利用基因敲除小鼠剖析了骨髓 GSDMD 的作用,尤其是其对外周 T 细胞反应和 AD 炎症的影响。研究人员利用 RNA 测序和体外共培养技术阐明了相关的免疫机制。利用靶向抑制剂实验和临床相关性分析进一步验证了GSDMD在AD中的功能:结果:在本研究中,5×FAD小鼠脾脏中caspase激活的GSDMD早于大脑中的GSDMD。研究表明,髓系细胞 GSDMD 的缺失会损害外周早期效应 T 细胞的活化,并阻止 T 细胞浸润大脑,从而全面减轻神经炎症。此外,髓系细胞 GSDMD 通过 IL-1β/NF-κB 途径诱导 T 细胞 PD-1 的表达,从而限制调节性 T 细胞。研究发现,服用 GSDMD 抑制剂和抗 PD-1 抗体可减轻 AD 相关炎症的发展。在一些AD患者中,血浆sPD-1与IL-Iβ和临床特征呈正相关:我们的研究系统地确定了 GSDMD 在 AD 相关外周炎症和早期 T 细胞免疫中的作用。结论:我们的研究系统地确定了 GSDMD 在与 AD 相关的外周炎症和早期 T 细胞免疫中的作用,这些发现还表明,以 GSDMD 为靶点对 AD 进行早期干预具有治疗潜力。
{"title":"Myeloid gasdermin D drives early-stage T cell immunity and peripheral inflammation in a mouse model of Alzheimer's disease.","authors":"Wenjuan Rui, Yuqing Wu, Yongbing Yang, Wenting Xie, Dengli Qin, Jie Ming, Zhihan Ye, Liu Lu, Ming Zong, Xianglong Tang, Lieying Fan, Sheng Li","doi":"10.1186/s12974-024-03255-9","DOIUrl":"10.1186/s12974-024-03255-9","url":null,"abstract":"<p><strong>Background: </strong>It is now realized that peripheral inflammation and abnormal immune responses, especially T cells, contribute to the development of Alzheimer's disease (AD). Gasdermin D (GSDMD) -mediated pyroptosis has been associated with several neuroinflammatory diseases, but whether GSDMD is involved in the peripheral inflammation and T cell immunity during AD remains unclear.</p><p><strong>Methods: </strong>We dynamically investigated GSDMD activation in the peripheral and central nervous system of 5×FAD mouse model and dissected the role of myeloid GSDMD using genetic knockout mice, especially its influence on peripheral T cell responses and AD inflammation. RNA sequencing and in vitro coculture were used to elucidate the underlying immune mechanisms involved. Targeted inhibitor experiments and clinical correlation analysis were used to further verify the function of GSDMD in AD.</p><p><strong>Results: </strong>In the present study, caspase activated GSDMD in the spleen of 5×FAD mice earlier than in the brain during disease progression. Loss of myeloid cell GSDMD was shown to impair early-stage effector T cell activation in the periphery and prevent T cell infiltration into the brain, with an overall reduction in neuroinflammation. Furthermore, myeloid cell GSDMD induced T cell PD-1 expression through the IL-1β/NF-κB pathway, restricting regulatory T cells. The administration of a GSDMD inhibitor combined with an anti-PD-1 antibody was found to mitigate the development of AD-associated inflammation. In some AD patients, plasma sPD-1 is positively correlated with IL-Iβ and clinical features.</p><p><strong>Conclusions: </strong>Our study systematically identified a role for GSDMD in the AD-related peripheral inflammation and early-stage T cell immunity. These findings also suggest the therapeutic potential of targeting GSDMD for the early intervention in AD.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"266"},"PeriodicalIF":9.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11491014/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467944","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
Anti-CD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8+ T-cell cytotoxicity after traumatic brain injury. 抗 CD49d Ab 治疗可改善年龄相关的炎症反应,并减轻脑外伤后 CD8+ T 细胞的细胞毒性。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-19 DOI: 10.1186/s12974-024-03257-7
Zhangying Chen, Kacie P Ford, Mecca B A R Islam, Hanxiao Wan, Hyebin Han, Abhirami Ramakrishnan, Ryan J Brown, Veronica Villanueva, Yidan Wang, Booker T Davis, Craig Weiss, Weiguo Cui, David Gate, Steven J Schwulst

Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8+ T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8+ T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8+ T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people.

在创伤性脑损伤(TBI)患者中,65 岁及以上的患者所占比例越来越大。与年轻人相比,老年创伤性脑损伤患者的发病率和死亡率更高。我们之前的数据表明,通过阻断α4整合素,抗CD49d抗体(aCD49d Ab)可抑制CD8+ T细胞向损伤脑部的浸润,提高存活率并减轻神经认知障碍。在这里,我们旨在揭示 aCD49d Ab 治疗如何改变老化小鼠大脑中的局部细胞反应。因此,小鼠在创伤后长期会产生与年龄相关的毒性细胞因子和趋化因子反应。aCD49d 抗体可减轻这种反应,同时减轻 T 辅助细胞(Th)1/Th17 的免疫学转变,并修复 CD8+ T 细胞的整体细胞毒性。此外,aCD49d Ab 还能减少表现出较高效应状态的 CD8+ T 细胞,从而减少慢性创伤性脑损伤老龄小鼠(而非年轻小鼠)大脑中的克隆扩增。总之,aCD49d Ab是治疗老年人创伤性脑损伤的一种很有前景的治疗策略。
{"title":"Anti-CD49d Ab treatment ameliorates age-associated inflammatory response and mitigates CD8<sup>+</sup> T-cell cytotoxicity after traumatic brain injury.","authors":"Zhangying Chen, Kacie P Ford, Mecca B A R Islam, Hanxiao Wan, Hyebin Han, Abhirami Ramakrishnan, Ryan J Brown, Veronica Villanueva, Yidan Wang, Booker T Davis, Craig Weiss, Weiguo Cui, David Gate, Steven J Schwulst","doi":"10.1186/s12974-024-03257-7","DOIUrl":"10.1186/s12974-024-03257-7","url":null,"abstract":"<p><p>Patients aged 65 years and older account for an increasing proportion of patients with traumatic brain injury (TBI). Older TBI patients experience increased morbidity and mortality compared to their younger counterparts. Our prior data demonstrated that by blocking α4 integrin, anti-CD49d antibody (aCD49d Ab) abrogates CD8<sup>+</sup> T-cell infiltration into the injured brain, improves survival, and attenuates neurocognitive deficits. Here, we aimed to uncover how aCD49d Ab treatment alters local cellular responses in the aged mouse brain. Consequently, mice incur age-associated toxic cytokine and chemokine responses long-term post-TBI. aCD49d Ab attenuates this response along with a T helper (Th)1/Th17 immunological shift and remediation of overall CD8<sup>+</sup> T cell cytotoxicity. Furthermore, aCD49d Ab reduces CD8<sup>+</sup> T cells exhibiting higher effector status, leading to reduced clonal expansion in aged, but not young, mouse brains with chronic TBI. Together, aCD49d Ab is a promising therapeutic strategy for treating TBI in the older people.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"267"},"PeriodicalIF":9.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11491007/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467925","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
Compromised endothelial Wnt/β-catenin signaling mediates the blood-brain barrier disruption and leads to neuroinflammation in endotoxemia. 内皮细胞 Wnt/β-catenin 信号转导受损介导了血脑屏障破坏,并导致内毒素血症中的神经炎症。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-19 DOI: 10.1186/s12974-024-03261-x
Xiaowen Huang, Pengju Wei, Cheng Fang, Min Yu, Shilun Yang, Linhui Qiu, Yu Wang, Aimin Xu, Ruby Lai Chong Hoo, Junlei Chang

The blood-brain barrier (BBB) is a critical interface that maintains the central nervous system homeostasis by controlling the exchange of substances between the blood and the brain. Disruption of the BBB plays a vital role in the development of neuroinflammation and neurological dysfunction in sepsis, but the mechanisms by which the BBB becomes disrupted during sepsis are not well understood. Here, we induced endotoxemia, a major type of sepsis, in mice by intraperitoneal injection of lipopolysaccharide (LPS). LPS acutely increased BBB permeability, activated microglia, and heightened inflammatory responses in brain endothelium and parenchyma. Concurrently, LPS or proinflammatory cytokines activated the NF-κB pathway, inhibiting Wnt/β-catenin signaling in brain endothelial cells in vitro and in vivo. Cell culture study revealed that NF-κB p65 directly interacted with β-catenin to suppress Wnt/β-catenin signaling. Pharmacological NF-κB pathway inhibition restored brain endothelial Wnt/β-catenin signaling activity and mitigated BBB disruption and neuroinflammation in septic mice. Furthermore, genetic or pharmacological activation of brain endothelial Wnt/β-catenin signaling substantially alleviated LPS-induced BBB leakage and neuroinflammation, while endothelial conditional ablation of the Wnt7a/7b co-receptor Gpr124 exacerbated the BBB leakage caused by LPS. Mechanistically, Wnt/β-catenin signaling activation rectified the reduced expression levels of tight junction protein ZO-1 and transcytosis suppressor Mfsd2a in brain endothelial cells of mice with endotoxemia, inhibiting both paracellular and transcellular permeability of the BBB. Our findings demonstrate that endotoxemia-associated systemic inflammation decreases endothelial Wnt/β-catenin signaling through activating NF-κB pathway, resulting in acute BBB disruption and neuroinflammation. Targeting the endothelial Wnt/β-catenin signaling may offer a promising therapeutic strategy for preserving BBB integrity and treating neurological dysfunction in sepsis.

血脑屏障(BBB)是一个重要的界面,它通过控制血液和大脑之间的物质交换来维持中枢神经系统的平衡。血脑屏障的破坏在脓毒症神经炎症和神经功能障碍的发展过程中起着至关重要的作用,但人们对脓毒症期间血脑屏障破坏的机制还不甚了解。在这里,我们通过腹腔注射脂多糖(LPS)诱导小鼠发生内毒素血症(败血症的一种主要类型)。LPS 急性增加了 BBB 的通透性,激活了小胶质细胞,并增强了脑内皮细胞和实质细胞的炎症反应。同时,LPS 或促炎细胞因子激活了 NF-κB 通路,抑制了体外和体内脑内皮细胞的 Wnt/β-catenin 信号传导。细胞培养研究发现,NF-κB p65 与 β-catenin 直接相互作用,抑制了 Wnt/β-catenin 信号转导。药理NF-κB通路抑制恢复了脓毒症小鼠脑内皮Wnt/β-catenin信号活性,减轻了BBB破坏和神经炎症。此外,遗传或药物激活脑内皮 Wnt/β-catenin 信号大大缓解了 LPS 引起的 BBB 渗漏和神经炎症,而内皮条件性消减 Wnt7a/7b 共受体 Gpr124 则加剧了 LPS 引起的 BBB 渗漏。从机制上讲,Wnt/β-catenin 信号激活纠正了内毒素血症小鼠脑内皮细胞中紧密连接蛋白 ZO-1 和转细胞抑制因子 Mfsd2a 表达水平的降低,从而抑制了 BBB 的细胞旁通透性和转细胞通透性。我们的研究结果表明,内毒素血症相关的全身性炎症会通过激活NF-κB通路降低内皮Wnt/β-catenin信号传导,从而导致急性BBB破坏和神经炎症。以内皮 Wnt/β-catenin 信号为靶点可能会为保护败血症中的 BBB 完整性和治疗神经功能障碍提供一种有前景的治疗策略。
{"title":"Compromised endothelial Wnt/β-catenin signaling mediates the blood-brain barrier disruption and leads to neuroinflammation in endotoxemia.","authors":"Xiaowen Huang, Pengju Wei, Cheng Fang, Min Yu, Shilun Yang, Linhui Qiu, Yu Wang, Aimin Xu, Ruby Lai Chong Hoo, Junlei Chang","doi":"10.1186/s12974-024-03261-x","DOIUrl":"10.1186/s12974-024-03261-x","url":null,"abstract":"<p><p>The blood-brain barrier (BBB) is a critical interface that maintains the central nervous system homeostasis by controlling the exchange of substances between the blood and the brain. Disruption of the BBB plays a vital role in the development of neuroinflammation and neurological dysfunction in sepsis, but the mechanisms by which the BBB becomes disrupted during sepsis are not well understood. Here, we induced endotoxemia, a major type of sepsis, in mice by intraperitoneal injection of lipopolysaccharide (LPS). LPS acutely increased BBB permeability, activated microglia, and heightened inflammatory responses in brain endothelium and parenchyma. Concurrently, LPS or proinflammatory cytokines activated the NF-κB pathway, inhibiting Wnt/β-catenin signaling in brain endothelial cells in vitro and in vivo. Cell culture study revealed that NF-κB p65 directly interacted with β-catenin to suppress Wnt/β-catenin signaling. Pharmacological NF-κB pathway inhibition restored brain endothelial Wnt/β-catenin signaling activity and mitigated BBB disruption and neuroinflammation in septic mice. Furthermore, genetic or pharmacological activation of brain endothelial Wnt/β-catenin signaling substantially alleviated LPS-induced BBB leakage and neuroinflammation, while endothelial conditional ablation of the Wnt7a/7b co-receptor Gpr124 exacerbated the BBB leakage caused by LPS. Mechanistically, Wnt/β-catenin signaling activation rectified the reduced expression levels of tight junction protein ZO-1 and transcytosis suppressor Mfsd2a in brain endothelial cells of mice with endotoxemia, inhibiting both paracellular and transcellular permeability of the BBB. Our findings demonstrate that endotoxemia-associated systemic inflammation decreases endothelial Wnt/β-catenin signaling through activating NF-κB pathway, resulting in acute BBB disruption and neuroinflammation. Targeting the endothelial Wnt/β-catenin signaling may offer a promising therapeutic strategy for preserving BBB integrity and treating neurological dysfunction in sepsis.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"265"},"PeriodicalIF":9.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11491032/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467926","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
Transient CSF1R inhibition ameliorates behavioral deficits in Cntnap2 knockout and valproic acid-exposed mouse models of autism. 瞬时 CSF1R 抑制可改善 Cntnap2 基因敲除小鼠和丙戊酸暴露小鼠自闭症模型的行为缺陷。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-18 DOI: 10.1186/s12974-024-03259-5
Jiao Meng, Pengming Pan, Gengshuo Guo, Anqi Chen, Xiangbao Meng, Heli Liu

Microglial abnormality and heterogeneity are observed in autism spectrum disorder (ASD) patients and animal models of ASD. Microglial depletion by colony stimulating factor 1-receptor (CSF1R) inhibition has been proved to improve autism-like behaviors in maternal immune activation mouse offspring. However, it is unclear whether CSF1R inhibition has extensive effectiveness and pharmacological heterogeneity in treating autism models caused by genetic and environmental risk factors. Here, we report pharmacological functions and cellular mechanisms of PLX5622, a small-molecule CSF1R inhibitor, in treating Cntnap2 knockout and valproic acid (VPA)-exposed autism model mice. For the Cntnap2 knockout mice, PLX5622 can improve their social ability and reciprocal social behavior, slow down their hyperactivity in open field and repetitive grooming behavior, and enhance their nesting ability. For the VPA model mice, PLX5622 can enhance their social ability and social novelty, and alleviate their anxiety behavior, repetitive and stereotyped autism-like behaviors such as grooming and marble burying. At the cellular level, PLX5622 restores the morphology and/or number of microglia in the somatosensory cortex, striatum, and hippocampal CA1 regions of the two models. Specially, PLX5622 corrects neurophysiological abnormalities in the striatum of the Cntnap2 knockout mice, and in the somatosensory cortex, striatum, and hippocampal CA1 regions of the VPA model mice. Incidentally, microglial dynamic changes in the VPA model mice are also reported. Our study demonstrates that microglial depletion and repopulation by transient CSF1R inhibition is effective, and however, has differential pharmacological functions and cellular mechanisms in rescuing behavioral deficits in the two autism models.

在自闭症谱系障碍(ASD)患者和自闭症谱系障碍动物模型中观察到小胶质细胞异常和异质性。事实证明,通过抑制集落刺激因子 1-受体(CSF1R)来消耗小胶质细胞,可以改善母体免疫激活小鼠后代的自闭症样行为。然而,CSF1R抑制在治疗由遗传和环境风险因素引起的自闭症模型方面是否具有广泛的有效性和药理学异质性尚不清楚。在此,我们报告了小分子CSF1R抑制剂PLX5622治疗Cntnap2基因敲除和丙戊酸(VPA)暴露自闭症模型小鼠的药理功能和细胞机制。对于Cntnap2基因敲除小鼠,PLX5622能提高它们的社交能力和互惠社交行为,减缓它们在空旷场地的多动和重复梳理行为,并增强它们的筑巢能力。对于VPA模型小鼠,PLX5622可提高其社交能力和社交新奇感,减轻其焦虑行为、重复和刻板的自闭症样行为,如梳理和埋弹珠。在细胞水平上,PLX5622能恢复两个模型的躯体感觉皮层、纹状体和海马CA1区小胶质细胞的形态和/或数量。特别是,PLX5622 能纠正 Cntnap2 基因敲除小鼠纹状体的神经生理学异常,以及 VPA 模型小鼠躯体感觉皮层、纹状体和海马 CA1 区的神经生理学异常。此外,我们还报告了 VPA 模型小鼠的小胶质细胞动态变化。我们的研究表明,通过瞬时 CSF1R 抑制来消耗和重新填充小胶质细胞是有效的,但在挽救两种自闭症模型的行为缺陷方面,其药理功能和细胞机制有所不同。
{"title":"Transient CSF1R inhibition ameliorates behavioral deficits in Cntnap2 knockout and valproic acid-exposed mouse models of autism.","authors":"Jiao Meng, Pengming Pan, Gengshuo Guo, Anqi Chen, Xiangbao Meng, Heli Liu","doi":"10.1186/s12974-024-03259-5","DOIUrl":"10.1186/s12974-024-03259-5","url":null,"abstract":"<p><p>Microglial abnormality and heterogeneity are observed in autism spectrum disorder (ASD) patients and animal models of ASD. Microglial depletion by colony stimulating factor 1-receptor (CSF1R) inhibition has been proved to improve autism-like behaviors in maternal immune activation mouse offspring. However, it is unclear whether CSF1R inhibition has extensive effectiveness and pharmacological heterogeneity in treating autism models caused by genetic and environmental risk factors. Here, we report pharmacological functions and cellular mechanisms of PLX5622, a small-molecule CSF1R inhibitor, in treating Cntnap2 knockout and valproic acid (VPA)-exposed autism model mice. For the Cntnap2 knockout mice, PLX5622 can improve their social ability and reciprocal social behavior, slow down their hyperactivity in open field and repetitive grooming behavior, and enhance their nesting ability. For the VPA model mice, PLX5622 can enhance their social ability and social novelty, and alleviate their anxiety behavior, repetitive and stereotyped autism-like behaviors such as grooming and marble burying. At the cellular level, PLX5622 restores the morphology and/or number of microglia in the somatosensory cortex, striatum, and hippocampal CA1 regions of the two models. Specially, PLX5622 corrects neurophysiological abnormalities in the striatum of the Cntnap2 knockout mice, and in the somatosensory cortex, striatum, and hippocampal CA1 regions of the VPA model mice. Incidentally, microglial dynamic changes in the VPA model mice are also reported. Our study demonstrates that microglial depletion and repopulation by transient CSF1R inhibition is effective, and however, has differential pharmacological functions and cellular mechanisms in rescuing behavioral deficits in the two autism models.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"262"},"PeriodicalIF":9.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487716/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467955","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
MAVS signaling shapes microglia responses to neurotropic virus infection. MAVS 信号塑造了小胶质细胞对神经病毒感染的反应。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-18 DOI: 10.1186/s12974-024-03258-6
Olivia Luise Gern, Andreas Pavlou, Felix Mulenge, Lena Mareike Busker, Luca Ghita, Angela Aringo, Bibiana Costa, Julia Spanier, Inken Waltl, Martin Stangel, Ulrich Kalinke

Viral encephalitis is characterized by a series of immunological reactions that can control virus infection in the brain, but dysregulated responses may cause excessive inflammation and brain damage. Microglia are brain-resident myeloid cells that are specialized in surveilling the local CNS environment and in case of viral brain infection they contribute to the control of the infection and to restriction of viral dissemination. Here, we report that after exposure to neurotropic vesicular stomatitis virus (VSV), murine in vitro microglia cultures showed rapid upregulation of a broad range of pro-inflammatory and antiviral genes, which were stably expressed over the entire 8 h infection period. Additionally, a set of immunomodulatory genes was upregulated between 6 and 8 h post infection. In microglia cultures, the induction of several immune response pathways including cytokine responses was dependent on mitochondrial antiviral-signaling protein (MAVS). Consequently, in Mavs-deficient microglia the control of virus propagation failed as indicated by augmented virus titers and the accumulation of viral transcripts. Thus, in the analyzed in vitro system, MAVS signaling is critically required to achieve full microglia activation and to mediate profound antiviral effects. In Mavs-deficient mice, intranasal VSV instillation caused higher disease severity than in WT mice and virus dissemination was noticed beyond the olfactory bulb. Virus spread to inner regions of the olfactory bulb, i.e., the granular cell layer, correlated with the recruitment of highly inflammatory non-microglia myeloid cells into the olfactory bulb in Mavs-/- mice. Furthermore, increased cytokine levels were detected in the nasal cavity, the olfactory bulb and in other brain regions. Thus, microglial MAVS signaling is critically needed for virus sensing, full microglia activation, and for orchestration of protective immunity in the virus-infected CNS.

病毒性脑炎的特征是一系列免疫反应,这些反应可以控制病毒在大脑中的感染,但失调的反应可能导致过度炎症和脑损伤。小胶质细胞是大脑驻留的髓系细胞,专门监视中枢神经系统的局部环境,在脑部病毒感染的情况下,它们有助于控制感染和限制病毒传播。在这里,我们报告了小鼠体外小胶质细胞培养物暴露于神经性水泡性口炎病毒(VSV)后,一系列促炎和抗病毒基因迅速上调,并在整个 8 小时感染期内稳定表达。此外,一组免疫调节基因在感染后 6 至 8 小时内上调。在小胶质细胞培养物中,包括细胞因子反应在内的多种免疫反应途径的诱导都依赖于线粒体抗病毒信号蛋白(MAVS)。因此,在缺乏 MAVS 的小胶质细胞中,病毒传播的控制失效,表现为病毒滴度升高和病毒转录物的积累。因此,在所分析的体外系统中,MAVS 信号传导是实现小胶质细胞完全激活和介导深远抗病毒效应的关键所在。与 WT 小鼠相比,在 MAVs 缺失的小鼠中,鼻内灌注 VSV 导致的疾病严重程度更高,而且病毒传播范围超出了嗅球。病毒传播到嗅球内部区域(即颗粒细胞层)与Mavs-/-小鼠嗅球中高度炎症性非小胶质细胞髓样细胞的招募有关。此外,在鼻腔、嗅球和其他脑区也检测到细胞因子水平升高。因此,在病毒感染的中枢神经系统中,小胶质细胞 MAVS 信号传导对于病毒感染、小胶质细胞全面激活和协调保护性免疫至关重要。
{"title":"MAVS signaling shapes microglia responses to neurotropic virus infection.","authors":"Olivia Luise Gern, Andreas Pavlou, Felix Mulenge, Lena Mareike Busker, Luca Ghita, Angela Aringo, Bibiana Costa, Julia Spanier, Inken Waltl, Martin Stangel, Ulrich Kalinke","doi":"10.1186/s12974-024-03258-6","DOIUrl":"10.1186/s12974-024-03258-6","url":null,"abstract":"<p><p>Viral encephalitis is characterized by a series of immunological reactions that can control virus infection in the brain, but dysregulated responses may cause excessive inflammation and brain damage. Microglia are brain-resident myeloid cells that are specialized in surveilling the local CNS environment and in case of viral brain infection they contribute to the control of the infection and to restriction of viral dissemination. Here, we report that after exposure to neurotropic vesicular stomatitis virus (VSV), murine in vitro microglia cultures showed rapid upregulation of a broad range of pro-inflammatory and antiviral genes, which were stably expressed over the entire 8 h infection period. Additionally, a set of immunomodulatory genes was upregulated between 6 and 8 h post infection. In microglia cultures, the induction of several immune response pathways including cytokine responses was dependent on mitochondrial antiviral-signaling protein (MAVS). Consequently, in Mavs-deficient microglia the control of virus propagation failed as indicated by augmented virus titers and the accumulation of viral transcripts. Thus, in the analyzed in vitro system, MAVS signaling is critically required to achieve full microglia activation and to mediate profound antiviral effects. In Mavs-deficient mice, intranasal VSV instillation caused higher disease severity than in WT mice and virus dissemination was noticed beyond the olfactory bulb. Virus spread to inner regions of the olfactory bulb, i.e., the granular cell layer, correlated with the recruitment of highly inflammatory non-microglia myeloid cells into the olfactory bulb in Mavs<sup>-/-</sup> mice. Furthermore, increased cytokine levels were detected in the nasal cavity, the olfactory bulb and in other brain regions. Thus, microglial MAVS signaling is critically needed for virus sensing, full microglia activation, and for orchestration of protective immunity in the virus-infected CNS.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"264"},"PeriodicalIF":9.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11490141/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467940","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
Intestinal homeostasis disrupted by Periodontitis exacerbates Alzheimer's Disease in APP/PS1 mice. 牙周炎破坏的肠道稳态会加重 APP/PS1 小鼠的阿尔茨海默病。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-18 DOI: 10.1186/s12974-024-03256-8
Xueshen Qian, Xuxin Lin, Weiqiang Hu, Lu Zhang, Wenqian Chen, Shuang Zhang, Song Ge, Xiongcheng Xu, Kai Luo

Periodontitis exacerbates Alzheimer's disease (AD) through multiple pathways. Both periodontitis and AD are intricately correlated to intestinal homeostasis, yet there is still a lack of direct evidence regarding whether periodontitis can regulate the progression of AD by modulating intestinal homeostasis. The current study induced experimental periodontitis in AD mice by bilaterally ligating the maxillary second molars with silk and administering Pg-LPS injections in APPswe/PS1ΔE9 (APP/PS1) mice. Behavioral tests and histological analyses of brain tissue were conducted after 8 weeks. Gut microbiota was analyzed and colon tissue were also evaluated. Then, fecal microbiota from mice with periodontitis was transplanted into antibiotic-treated mice to confirm the effects of periodontitis on AD and the potential mechanism was explored. The results indicated periodontitis exacerbated cognitive impairment and anxious behaviour in APP/PS1 mice, with increased Aβ deposition, microglial overactivation and neuroinflammation in brain. Moreover, the intestinal homeostasis of AD mice was altered by periodontitis, including affecting gut microbiota composition, causing colon inflammation and destroyed intestinal epithelial barrier. Furthermore, AD mice that underwent fecal transplantation from mice with periodontitis exhibited worsened AD progression and disrupted intestinal homeostasis. It also impaired intestinal barrier function, elevated peripheral inflammation, damaged blood-brain barrier (BBB) and caused neuroinflammation and synapses impairment. Taken together, the current study demonstrated that periodontitis could disrupt intestinal homeostasis to exacerbate AD progression potential via causing gut microbial dysbiosis, intestinal inflammation and intestinal barrier impairment to induce peripheral inflammation and damage BBB, ultimately leading to neuroinflammation and synapse impairment. It underscores the importance of maintaining both periodontal health and intestinal homeostasis to reduce the risk of AD.

牙周炎会通过多种途径加剧阿尔茨海默病(AD)。牙周炎和阿尔茨海默病都与肠道稳态密切相关,但关于牙周炎是否能通过调节肠道稳态来调控阿尔茨海默病的进展,目前仍缺乏直接证据。本研究通过用丝线结扎上颌第二磨牙,并给APPswe/PS1ΔE9(APP/PS1)小鼠注射Pg-LPS,诱导AD小鼠发生实验性牙周炎。8 周后进行行为测试和脑组织组织学分析。同时还分析了肠道微生物群并评估了结肠组织。然后,将牙周炎小鼠的粪便微生物群移植到抗生素治疗的小鼠体内,以证实牙周炎对AD的影响,并探讨其潜在机制。结果表明,牙周炎加剧了APP/PS1小鼠的认知障碍和焦虑行为,增加了脑内Aβ沉积、小胶质细胞过度激活和神经炎症。此外,牙周炎还改变了AD小鼠的肠道稳态,包括影响肠道微生物群组成、引起结肠炎症和破坏肠道上皮屏障。此外,接受牙周炎小鼠粪便移植的AD小鼠表现出AD恶化和肠道平衡被破坏。它还损害了肠道屏障功能,加剧了外周炎症,破坏了血脑屏障(BBB),并导致神经炎症和突触受损。综上所述,本研究表明,牙周炎可通过引起肠道微生物菌群失调、肠道炎症和肠道屏障受损,诱发外周炎症和损伤血脑屏障,最终导致神经炎症和突触受损,从而破坏肠道平衡,加剧AD进展的可能性。该研究强调了保持牙周健康和肠道平衡对降低 AD 风险的重要性。
{"title":"Intestinal homeostasis disrupted by Periodontitis exacerbates Alzheimer's Disease in APP/PS1 mice.","authors":"Xueshen Qian, Xuxin Lin, Weiqiang Hu, Lu Zhang, Wenqian Chen, Shuang Zhang, Song Ge, Xiongcheng Xu, Kai Luo","doi":"10.1186/s12974-024-03256-8","DOIUrl":"https://doi.org/10.1186/s12974-024-03256-8","url":null,"abstract":"<p><p>Periodontitis exacerbates Alzheimer's disease (AD) through multiple pathways. Both periodontitis and AD are intricately correlated to intestinal homeostasis, yet there is still a lack of direct evidence regarding whether periodontitis can regulate the progression of AD by modulating intestinal homeostasis. The current study induced experimental periodontitis in AD mice by bilaterally ligating the maxillary second molars with silk and administering Pg-LPS injections in APP<sup>swe</sup>/PS1<sup>ΔE9</sup> (APP/PS1) mice. Behavioral tests and histological analyses of brain tissue were conducted after 8 weeks. Gut microbiota was analyzed and colon tissue were also evaluated. Then, fecal microbiota from mice with periodontitis was transplanted into antibiotic-treated mice to confirm the effects of periodontitis on AD and the potential mechanism was explored. The results indicated periodontitis exacerbated cognitive impairment and anxious behaviour in APP/PS1 mice, with increased Aβ deposition, microglial overactivation and neuroinflammation in brain. Moreover, the intestinal homeostasis of AD mice was altered by periodontitis, including affecting gut microbiota composition, causing colon inflammation and destroyed intestinal epithelial barrier. Furthermore, AD mice that underwent fecal transplantation from mice with periodontitis exhibited worsened AD progression and disrupted intestinal homeostasis. It also impaired intestinal barrier function, elevated peripheral inflammation, damaged blood-brain barrier (BBB) and caused neuroinflammation and synapses impairment. Taken together, the current study demonstrated that periodontitis could disrupt intestinal homeostasis to exacerbate AD progression potential via causing gut microbial dysbiosis, intestinal inflammation and intestinal barrier impairment to induce peripheral inflammation and damage BBB, ultimately leading to neuroinflammation and synapse impairment. It underscores the importance of maintaining both periodontal health and intestinal homeostasis to reduce the risk of AD.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"263"},"PeriodicalIF":9.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11489998/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467939","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
Glutamine metabolism modulates microglial NLRP3 inflammasome activity through mitophagy in Alzheimer's disease. 谷氨酰胺代谢在阿尔茨海默病中通过丝裂吞噬调节小胶质细胞NLRP3炎性体的活性
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-15 DOI: 10.1186/s12974-024-03254-w
Zhixin Zhang, Miao Li, Xiang Li, Zhiyang Feng, Gan Luo, Ying Wang, Xiaoyan Gao

The NLR family pyrin domain containing 3 (NLRP3) inflammasome in microglia is intimately linked to the pathogenesis of Alzheimer's disease (AD). Although NLRP3 inflammasome activity is regulated by cellular metabolism, the underlying mechanism remains elusive. Here, we found that under the pathological conditions of AD, the activation of NLRP3 inflammasome in microglia is accompanied by increased glutamine metabolism. Suppression of glutaminase, the rate limiting enzyme in glutamine metabolism, attenuated the NLRP3 inflammasome activation both in the microglia of AD mice and cultured inflammatory microglia. Mechanistically, inhibiting glutaminase blocked the anaplerotic flux of glutamine to the tricarboxylic acid cycle and amino acid synthesis, down-regulated mTORC1 signaling by phosphorylating AMPK, which stimulated mitophagy and limited the accumulation of intracellular reactive oxygen species, ultimately prevented the activation of NLRP3 inflammasomes in activated microglia during AD. Taken together, our findings suggest that glutamine metabolism regulates the activation of NLRP3 inflammasome through mitophagy in microglia, thus providing a potential therapeutic target for AD treatment.

小胶质细胞中的 NLR 家族含吡啶域 3(NLRP3)炎性体与阿尔茨海默病(AD)的发病机制密切相关。尽管NLRP3炎性体的活性受细胞代谢的调控,但其潜在机制仍然难以捉摸。在这里,我们发现在 AD 的病理条件下,小胶质细胞中 NLRP3 炎症小体的激活伴随着谷氨酰胺代谢的增加。谷氨酰胺酶是谷氨酰胺代谢的限速酶,抑制谷氨酰胺酶可减轻AD小鼠小胶质细胞和培养的炎性小胶质细胞中NLRP3炎性体的激活。从机理上讲,抑制谷氨酰胺酶可阻断谷氨酰胺向三羧酸循环和氨基酸合成的无机通路,通过磷酸化AMPK下调mTORC1信号传导,从而刺激有丝分裂并限制细胞内活性氧的积累,最终阻止AD期间激活的小胶质细胞中NLRP3炎性体的活化。综上所述,我们的研究结果表明,谷氨酰胺代谢通过小胶质细胞中的有丝分裂来调节NLRP3炎性体的激活,从而为AD治疗提供了一个潜在的治疗靶点。
{"title":"Glutamine metabolism modulates microglial NLRP3 inflammasome activity through mitophagy in Alzheimer's disease.","authors":"Zhixin Zhang, Miao Li, Xiang Li, Zhiyang Feng, Gan Luo, Ying Wang, Xiaoyan Gao","doi":"10.1186/s12974-024-03254-w","DOIUrl":"https://doi.org/10.1186/s12974-024-03254-w","url":null,"abstract":"<p><p>The NLR family pyrin domain containing 3 (NLRP3) inflammasome in microglia is intimately linked to the pathogenesis of Alzheimer's disease (AD). Although NLRP3 inflammasome activity is regulated by cellular metabolism, the underlying mechanism remains elusive. Here, we found that under the pathological conditions of AD, the activation of NLRP3 inflammasome in microglia is accompanied by increased glutamine metabolism. Suppression of glutaminase, the rate limiting enzyme in glutamine metabolism, attenuated the NLRP3 inflammasome activation both in the microglia of AD mice and cultured inflammatory microglia. Mechanistically, inhibiting glutaminase blocked the anaplerotic flux of glutamine to the tricarboxylic acid cycle and amino acid synthesis, down-regulated mTORC1 signaling by phosphorylating AMPK, which stimulated mitophagy and limited the accumulation of intracellular reactive oxygen species, ultimately prevented the activation of NLRP3 inflammasomes in activated microglia during AD. Taken together, our findings suggest that glutamine metabolism regulates the activation of NLRP3 inflammasome through mitophagy in microglia, thus providing a potential therapeutic target for AD treatment.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"261"},"PeriodicalIF":9.3,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11481753/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467938","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
期刊
Journal of Neuroinflammation
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