Journal of Neuroinflammation最新文献

筛选
英文 中文
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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation Pub Date : 2024-10-18 DOI: 10.1186/s12974-024-03259-5
Jiao Meng, Pengming Pan, Gengshuo Guo, Anqi Chen, Xiangbao Meng, Heli Liu
{"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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation 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
{"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
Journal of Neuroinflammation 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
{"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
Minocycline prevents early age-related cognitive decline in a mouse model of intellectual disability caused by ZBTB18/RP58 haploinsufficiency. 米诺环素可预防ZBTB18/RP58单倍体缺失导致的智力残疾小鼠模型中与年龄相关的早期认知能力衰退。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-10-12 DOI: 10.1186/s12974-024-03217-1
Tomoko Tanaka, Shinobu Hirai, Hiroyuki Manabe, Kentaro Endo, Hiroko Shimbo, Yasumasa Nishito, Junjiro Horiuchi, Hikari Yoshitane, Haruo Okado
{"title":"Minocycline prevents early age-related cognitive decline in a mouse model of intellectual disability caused by ZBTB18/RP58 haploinsufficiency.","authors":"Tomoko Tanaka, Shinobu Hirai, Hiroyuki Manabe, Kentaro Endo, Hiroko Shimbo, Yasumasa Nishito, Junjiro Horiuchi, Hikari Yoshitane, Haruo Okado","doi":"10.1186/s12974-024-03217-1","DOIUrl":"https://doi.org/10.1186/s12974-024-03217-1","url":null,"abstract":"<p><p>Haploinsufficiency of the transcriptional repressor ZBTB18/RP58 is associated with intellectual disability. However, the mechanisms causing this disability are unknown, and preventative measures and treatments are not available. Here, we assessed multiple behaviors in Zbtb18/Rp58 heterozygous-knockout mice, and examined local field potentials, DNA fragmentation, mitochondrial morphology, and performed histochemical and transcriptome analyses in the hippocampus to evaluate chronic inflammation. In wild-type mice, object location memory was present at a similar level at 2 and 4-5 months of age, and became impaired at 12-18 months. In contrast, Zbtb18/Rp58 heterozygous-knockout mice displayed early onset impairments in object location memory by 4-5 months of age. These mice also exhibited earlier accumulation of DNA and mitochondrial damage, and activated microglia in the dentate gyrus, which are associated with defective DNA repair. Notably, chronic minocycline therapy, which has neuroprotective and anti-inflammatory effects, attenuated age-related phenotypes, including accumulation of DNA damage, increased microglial activation, and impairment of object location memory. Our results suggest that Zbtb18/Rp58 activity is required for DNA repair and its reduction results in DNA and mitochondrial damage, increased activation of microglia, and inflammation, leading to accelerated declines in cognitive functions. Minocycline has potential as a therapeutic agent for the treatment of ZBTB18/RP58 haploinsufficiency-associated cognitive dysfunction.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"260"},"PeriodicalIF":9.3,"publicationDate":"2024-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11471036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142467942","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信