Journal of Neuroinflammation最新文献

筛选
英文 中文
Protecting effects of 4-octyl itaconate on neonatal hypoxic-ischemic encephalopathy via Nrf2 pathway in astrocytes. 伊它康酸 4-辛酯通过星形胶质细胞中的 Nrf2 通路对新生儿缺氧缺血性脑病的保护作用
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-17 DOI: 10.1186/s12974-024-03121-8
Yanping Yang, Yang Li, Wenyi Yang, Xueying Yang, Man Luo, Ling Qin, Junchao Zhu
{"title":"Protecting effects of 4-octyl itaconate on neonatal hypoxic-ischemic encephalopathy via Nrf2 pathway in astrocytes.","authors":"Yanping Yang, Yang Li, Wenyi Yang, Xueying Yang, Man Luo, Ling Qin, Junchao Zhu","doi":"10.1186/s12974-024-03121-8","DOIUrl":"10.1186/s12974-024-03121-8","url":null,"abstract":"<p><strong>Background: </strong>Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the most common neurological problems occurring in the perinatal period. However, there still is not a promising approach to reduce long-term neurodevelopmental outcomes of HIE. Recently, itaconate has been found to exhibit anti-oxidative and anti-inflammatory effects. However, the therapeutic efficacy of itaconate in HIE remains inconclusive. Therefore, this study attempts to explore the pathophysiological mechanisms of oxidative stress and inflammatory responses in HIE as well as the potential therapeutic role of a derivative of itaconate, 4-octyl itaconate (4OI).</p><p><strong>Methods: </strong>We used 7-day-old mice to induce hypoxic-ischemic (HI) model by right common carotid artery ligation followed by 1 h of hypoxia. Behavioral experiments including the Y-maze and novel object recognition test were performed on HI mice at P60 to evaluate long-term neurodevelopmental outcomes. We employed an approach combining non-targeted metabolomics with transcriptomics to screen alterations in metabolic profiles and gene expression in the hippocampal tissue of the mice at 8 h after hypoxia. Immunofluorescence staining and RT-PCR were used to evaluate the pathological changes in brain tissue cells and the expression of mRNA and proteins. 4OI was intraperitoneally injected into HI model mice to assess its anti-inflammatory and antioxidant effects. BV2 and C8D1A cells were cultured in vitro to study the effect of 4OI on the expression and nuclear translocation of Nrf2. We also used Nrf2-siRNA to further validate 4OI-induced Nrf2 pathway in astrocytes.</p><p><strong>Results: </strong>We found that in the acute phase of HI, there was an accumulation of pyruvate and lactate in the hippocampal tissue, accompanied by oxidative stress and pro-inflammatory, as well as increased expression of antioxidative stress and anti-inflammatory genes. Treatment of 4OI could inhibit activation and proliferation of microglial cells and astrocytes, reduce neuronal death and relieve cognitive dysfunction in HI mice. Furthermore, 4OI enhanced nuclear factor erythroid-2-related factor (Nfe2l2; Nrf2) expression and nuclear translocation in astrocytes, reduced pro-inflammatory cytokine production, and increased antioxidant enzyme expression.</p><p><strong>Conclusion: </strong>Our study demonstrates that 4OI has a potential therapeutic effect on neuronal damage and cognitive deficits in HIE, potentially through the modulation of inflammation and oxidative stress pathways by Nrf2 in astrocytes.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11102208/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140958166","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
Therapeutic effects of orexin-A in sepsis-associated encephalopathy in mice. 奥曲肽-A 对小鼠败血症相关脑病的治疗效果。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-17 DOI: 10.1186/s12974-024-03111-w
Jing Guo, Zhuo Kong, Sha Yang, Jingjing Da, Liangzhao Chu, Guoqiang Han, Jian Liu, Ying Tan, Jiqin Zhang
{"title":"Therapeutic effects of orexin-A in sepsis-associated encephalopathy in mice.","authors":"Jing Guo, Zhuo Kong, Sha Yang, Jingjing Da, Liangzhao Chu, Guoqiang Han, Jian Liu, Ying Tan, Jiqin Zhang","doi":"10.1186/s12974-024-03111-w","DOIUrl":"10.1186/s12974-024-03111-w","url":null,"abstract":"<p><strong>Background: </strong>Sepsis-associated encephalopathy (SAE) causes acute and long-term cognitive deficits. However, information on the prevention and treatment of cognitive dysfunction after sepsis is limited. The neuropeptide orexin-A (OXA) has been shown to play a protective role against neurological diseases by modulating the inflammatory response through the activation of OXR1 and OXR2 receptors. However, the role of OXA in mediating the neuroprotective effects of SAE has not yet been reported.</p><p><strong>Methods: </strong>A mouse model of SAE was induced using cecal ligation perforation (CLP) and treated via intranasal administration of exogenous OXA after surgery. Mouse survival, in addition to cognitive and anxiety behaviors, were assessed. Changes in neurons, cerebral edema, blood-brain barrier (BBB) permeability, and brain ultrastructure were monitored. Levels of pro-inflammatory factors (IL-1β, TNF-α) and microglial activation were also measured. The underlying molecular mechanisms were investigated by proteomics analysis and western blotting.</p><p><strong>Results: </strong>Intranasal OXA treatment reduced mortality, ameliorated cognitive and emotional deficits, and attenuated cerebral edema, BBB disruption, and ultrastructural brain damage in mice. In addition, OXA significantly reduced the expression of the pro-inflammatory factors IL-1β and TNF-α, and inhibited microglial activation. In addition, OXA downregulated the expression of the Rras and RAS proteins, and reduced the phosphorylation of P-38 and JNK, thus inhibiting activation of the MAPK pathway. JNJ-10,397,049 (an OXR2 blocker) reversed the effect of OXA, whereas SB-334,867 (an OXR1 blocker) did not.</p><p><strong>Conclusion: </strong>This study demonstrated that the intranasal administration of moderate amounts of OXA protects the BBB and inhibits the activation of the OXR2/RAS/MAPK pathway to attenuate the outcome of SAE, suggesting that OXA may be a promising therapeutic approach for the management of SAE.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11102217/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140958126","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
Overexpression of pathogenic tau in astrocytes causes a reduction in AQP4 and GLT1, an immunosuppressed phenotype and unique transcriptional responses to repetitive mild TBI without appreciable changes in tauopathy. 致病性 tau 在星形胶质细胞中的过度表达会导致 AQP4 和 GLT1 的减少、免疫抑制表型以及对重复性轻度创伤性脑损伤的独特转录反应,而 tauopathy 却没有明显变化。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-15 DOI: 10.1186/s12974-024-03117-4
Camila Ortiz, Andrew Pearson, Robyn McCartan, Shawn Roche, Nolan Carothers, Mackenzie Browning, Sylvia Perez, Bin He, Stephen D Ginsberg, Michael Mullan, Elliott J Mufson, Fiona Crawford, Joseph Ojo
{"title":"Overexpression of pathogenic tau in astrocytes causes a reduction in AQP4 and GLT1, an immunosuppressed phenotype and unique transcriptional responses to repetitive mild TBI without appreciable changes in tauopathy.","authors":"Camila Ortiz, Andrew Pearson, Robyn McCartan, Shawn Roche, Nolan Carothers, Mackenzie Browning, Sylvia Perez, Bin He, Stephen D Ginsberg, Michael Mullan, Elliott J Mufson, Fiona Crawford, Joseph Ojo","doi":"10.1186/s12974-024-03117-4","DOIUrl":"10.1186/s12974-024-03117-4","url":null,"abstract":"<p><p>Epidemiological studies have unveiled a robust link between exposure to repetitive mild traumatic brain injury (r-mTBI) and elevated susceptibility to develop neurodegenerative disorders, notably chronic traumatic encephalopathy (CTE). The pathogenic lesion in CTE cases is characterized by the accumulation of hyperphosphorylated tau in neurons around small cerebral blood vessels which can be accompanied by astrocytes that contain phosphorylated tau, the latter termed tau astrogliopathy. However, the contribution of tau astrogliopathy to the pathobiology and functional consequences of r-mTBI/CTE or whether it is merely a consequence of aging remains unclear. We addressed these pivotal questions by utilizing a mouse model harboring tau-bearing astrocytes, GFAP<sup>P301L</sup> mice, subjected to our r-mTBI paradigm. Despite the fact that r-mTBI did not exacerbate tau astrogliopathy or general tauopathy, it increased phosphorylated tau in the area underneath the impact site. Additionally, gene ontology analysis of tau-bearing astrocytes following r-mTBI revealed profound alterations in key biological processes including immunological and mitochondrial bioenergetics. Moreover, gene array analysis of microdissected astrocytes accrued from stage IV CTE human brains revealed an immunosuppressed astroglial phenotype similar to tau-bearing astrocytes in the GFAP<sup>P301L</sup> model. Additionally, hippocampal reduction of proteins involved in water transport (AQP4) and glutamate homeostasis (GLT1) was found in the mouse model of tau astrogliopathy. Collectively, these findings reveal the importance of understanding tau astrogliopathy and its role in astroglial pathobiology under normal circumstances and following r-mTBI. The identified mechanisms using this GFAP<sup>P301L</sup> model may suggest targets for therapeutic interventions in r-mTBI pathogenesis in the context of CTE.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11096096/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140943131","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
Interleukin-9 protects from microglia- and TNF-mediated synaptotoxicity in experimental multiple sclerosis. 白细胞介素-9能保护实验性多发性硬化症患者免受小胶质细胞和 TNF 介导的突触毒性的影响。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-14 DOI: 10.1186/s12974-024-03120-9
Livia Guadalupi, Valentina Vanni, Sara Balletta, Silvia Caioli, Francesca De Vito, Diego Fresegna, Krizia Sanna, Monica Nencini, Gloria Donninelli, Elisabetta Volpe, Fabrizio Mariani, Luca Battistini, Mario Stampanoni Bassi, Luana Gilio, Antonio Bruno, Ettore Dolcetti, Fabio Buttari, Georgia Mandolesi, Diego Centonze, Alessandra Musella
{"title":"Interleukin-9 protects from microglia- and TNF-mediated synaptotoxicity in experimental multiple sclerosis.","authors":"Livia Guadalupi, Valentina Vanni, Sara Balletta, Silvia Caioli, Francesca De Vito, Diego Fresegna, Krizia Sanna, Monica Nencini, Gloria Donninelli, Elisabetta Volpe, Fabrizio Mariani, Luca Battistini, Mario Stampanoni Bassi, Luana Gilio, Antonio Bruno, Ettore Dolcetti, Fabio Buttari, Georgia Mandolesi, Diego Centonze, Alessandra Musella","doi":"10.1186/s12974-024-03120-9","DOIUrl":"10.1186/s12974-024-03120-9","url":null,"abstract":"<p><strong>Background: </strong>Multiple sclerosis (MS) is a progressive neurodegenerative disease of the central nervous system characterized by inflammation-driven synaptic abnormalities. Interleukin-9 (IL-9) is emerging as a pleiotropic cytokine involved in MS pathophysiology.</p><p><strong>Methods: </strong>Through biochemical, immunohistochemical, and electrophysiological experiments, we investigated the effects of both peripheral and central administration of IL-9 on C57/BL6 female mice with experimental autoimmune encephalomyelitis (EAE), a model of MS.</p><p><strong>Results: </strong>We demonstrated that both systemic and local administration of IL-9 significantly improved clinical disability, reduced neuroinflammation, and mitigated synaptic damage in EAE. The results unveil an unrecognized central effect of IL-9 against microglia- and TNF-mediated neuronal excitotoxicity. Two main mechanisms emerged: first, IL-9 modulated microglial inflammatory activity by enhancing the expression of the triggering receptor expressed on myeloid cells-2 (TREM2) and reducing TNF release. Second, IL-9 suppressed neuronal TNF signaling, thereby blocking its synaptotoxic effects.</p><p><strong>Conclusions: </strong>The data presented in this work highlight IL-9 as a critical neuroprotective molecule capable of interfering with inflammatory synaptopathy in EAE. These findings open new avenues for treatments targeting the neurodegenerative damage associated with MS, as well as other inflammatory and neurodegenerative disorders of the central nervous system.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11092167/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140922203","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
Western diet increases brain metabolism and adaptive immune responses in a mouse model of amyloidosis. 西式饮食可增加淀粉样变性小鼠模型的脑代谢和适应性免疫反应。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-14 DOI: 10.1186/s12974-024-03080-0
Marilena Poxleitner, Sabrina H L Hoffmann, Georgy Berezhnoy, Tudor M Ionescu, Irene Gonzalez-Menendez, Florian C Maier, Dominik Seyfried, Walter Ehrlichmann, Leticia Quintanilla-Martinez, Andreas M Schmid, Gerald Reischl, Christoph Trautwein, Andreas Maurer, Bernd J Pichler, Kristina Herfert, Nicolas Beziere
{"title":"Western diet increases brain metabolism and adaptive immune responses in a mouse model of amyloidosis.","authors":"Marilena Poxleitner, Sabrina H L Hoffmann, Georgy Berezhnoy, Tudor M Ionescu, Irene Gonzalez-Menendez, Florian C Maier, Dominik Seyfried, Walter Ehrlichmann, Leticia Quintanilla-Martinez, Andreas M Schmid, Gerald Reischl, Christoph Trautwein, Andreas Maurer, Bernd J Pichler, Kristina Herfert, Nicolas Beziere","doi":"10.1186/s12974-024-03080-0","DOIUrl":"10.1186/s12974-024-03080-0","url":null,"abstract":"<p><p>Diet-induced increase in body weight is a growing health concern worldwide. Often accompanied by a low-grade metabolic inflammation that changes systemic functions, diet-induced alterations may contribute to neurodegenerative disorder progression as well. This study aims to non-invasively investigate diet-induced metabolic and inflammatory effects in the brain of an APPPS1 mouse model of Alzheimer's disease. [<sup>18</sup>F]FDG, [<sup>18</sup>F]FTHA, and [<sup>18</sup>F]GE-180 were used for in vivo PET imaging in wild-type and APPPS1 mice. Ex vivo flow cytometry and histology in brains complemented the in vivo findings. <sup>1</sup>H- magnetic resonance spectroscopy in the liver, plasma metabolomics and flow cytometry of the white adipose tissue were used to confirm metaflammatory condition in the periphery. We found disrupted glucose and fatty acid metabolism after Western diet consumption, with only small regional changes in glial-dependent neuroinflammation in the brains of APPPS1 mice. Further ex vivo investigations revealed cytotoxic T cell involvement in the brains of Western diet-fed mice and a disrupted plasma metabolome. <sup>1</sup>H-magentic resonance spectroscopy and immunological results revealed diet-dependent inflammatory-like misbalance in livers and fatty tissue. Our multimodal imaging study highlights the role of the brain-liver-fat axis and the adaptive immune system in the disruption of brain homeostasis in amyloid models of Alzheimer's disease.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11092112/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140922280","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
Extracellular vesicles produced by HIV-1 Nef-expressing cells induce myelin impairment and oligodendrocyte damage in the mouse central nervous system. 表达HIV-1 Nef的细胞产生的胞外囊泡会诱发小鼠中枢神经系统的髓鞘损伤和少突胶质细胞损伤。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-13 DOI: 10.1186/s12974-024-03124-5
Jessica K Schenck, Molly T Karl, Cheryl Clarkson-Paredes, Ashley Bastin, Tatiana Pushkarsky, Beda Brichacek, Robert H Miller, Michael I Bukrinsky
{"title":"Extracellular vesicles produced by HIV-1 Nef-expressing cells induce myelin impairment and oligodendrocyte damage in the mouse central nervous system.","authors":"Jessica K Schenck, Molly T Karl, Cheryl Clarkson-Paredes, Ashley Bastin, Tatiana Pushkarsky, Beda Brichacek, Robert H Miller, Michael I Bukrinsky","doi":"10.1186/s12974-024-03124-5","DOIUrl":"10.1186/s12974-024-03124-5","url":null,"abstract":"<p><p>HIV-associated neurocognitive disorders (HAND) are a spectrum of cognitive impairments that continue to affect approximately half of all HIV-positive individuals despite effective viral suppression through antiretroviral therapy (ART). White matter pathologies have persisted in the ART era, and the degree of white matter damage correlates with the degree of neurocognitive impairment in patients with HAND. The HIV protein Nef has been implicated in HAND pathogenesis, but its effect on white matter damage has not been well characterized. Here, utilizing in vivo, ex vivo, and in vitro methods, we demonstrate that Nef-containing extracellular vesicles (Nef EVs) disrupt myelin sheaths and inflict damage upon oligodendrocytes within the murine central nervous system. Intracranial injection of Nef EVs leads to reduced myelin basic protein (MBP) staining and a decreased number of CC1 + oligodendrocytes in the corpus callosum. Moreover, cerebellar slice cultures treated with Nef EVs exhibit diminished MBP expression and increased presence of unmyelinated axons. Primary mixed brain cultures and enriched oligodendrocyte precursor cell cultures exposed to Nef EVs display a decreased number of O4 + cells, indicative of oligodendrocyte impairment. These findings underscore the potential contribution of Nef EV-mediated damage to oligodendrocytes and myelin maintenance in the pathogenesis of HAND.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11090814/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140915774","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
Thymic gene expression analysis reveals a potential link between HIF-1A and Th17/Treg imbalance in thymoma associated myasthenia gravis. 胸腺基因表达分析揭示了HIF-1A与胸腺瘤相关性肌萎缩症中Th17/Treg失衡之间的潜在联系。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-11 DOI: 10.1186/s12974-024-03095-7
İlayda Altınönder, Mustafa Kaya, Sibel P Yentür, Arman Çakar, Hacer Durmuş, Gülçin Yegen, Berker Özkan, Yeşim Parman, Amr H Sawalha, Guher Saruhan-Direskeneli
{"title":"Thymic gene expression analysis reveals a potential link between HIF-1A and Th17/Treg imbalance in thymoma associated myasthenia gravis.","authors":"İlayda Altınönder, Mustafa Kaya, Sibel P Yentür, Arman Çakar, Hacer Durmuş, Gülçin Yegen, Berker Özkan, Yeşim Parman, Amr H Sawalha, Guher Saruhan-Direskeneli","doi":"10.1186/s12974-024-03095-7","DOIUrl":"10.1186/s12974-024-03095-7","url":null,"abstract":"<p><p>Myasthenia gravis (MG) is an immune-mediated disease frequently associated with thymic changes. Increased T helper 17 (Th17) cell activity and dysfunctional regulatory T (Treg) cells have been demonstrated in subgroups of MG. On the other hand, hypoxia-inducible factor 1 (HIF-1) has been shown to regulate the Th17/Treg balance by inducing Th17 differentiation while attenuating Treg development. To identify the underlying mechanisms of different thymic pathologies in MG development, we evaluated thymic samples from thymoma-associated myasthenia gravis (TAMG), MG with hyperplasia (TFH-MG) and thymoma without MG (TOMA) patients. Differential gene expression analysis revealed that TAMG and TFH-MG cells are associated with different functional pathways. A higher RORC/FOXP3 ratio provided evidence for Th17/Treg imbalance in TAMG potentially related to increased HIF1A. The hypoxic microenvironment in thymoma may be a driver of TAMG by increasing HIF1A. These findings may lead to new therapeutic approaches targeting HIF1A in the development of TAMG.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11088784/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140908974","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
Dysregulated brain-gut axis in the setting of traumatic brain injury: review of mechanisms and anti-inflammatory pharmacotherapies. 脑外伤情况下的脑-肠轴失调:机制和抗炎药物疗法综述。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-10 DOI: 10.1186/s12974-024-03118-3
Mahmoud G El Baassiri, Zachariah Raouf, Sarah Badin, Alejandro Escobosa, Chhinder P Sodhi, Isam W Nasr
{"title":"Dysregulated brain-gut axis in the setting of traumatic brain injury: review of mechanisms and anti-inflammatory pharmacotherapies.","authors":"Mahmoud G El Baassiri, Zachariah Raouf, Sarah Badin, Alejandro Escobosa, Chhinder P Sodhi, Isam W Nasr","doi":"10.1186/s12974-024-03118-3","DOIUrl":"10.1186/s12974-024-03118-3","url":null,"abstract":"<p><p>Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11083845/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140903538","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
Monocytes release cystatin F dimer to associate with Aβ and aggravate amyloid pathology and cognitive deficits in Alzheimer's disease. 单核细胞释放胱抑素 F 二聚体与 Aβ 结合,加重阿尔茨海默病的淀粉样病理和认知障碍。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-10 DOI: 10.1186/s12974-024-03119-2
Qiang Li, Bing Li, Li Liu, Kang-Ji Wang, Ming-Yue Liu, Yu Deng, Ze Li, Wei-Dong Zhao, Li-Yong Wu, Yu-Hua Chen, Ke Zhang
{"title":"Monocytes release cystatin F dimer to associate with Aβ and aggravate amyloid pathology and cognitive deficits in Alzheimer's disease.","authors":"Qiang Li, Bing Li, Li Liu, Kang-Ji Wang, Ming-Yue Liu, Yu Deng, Ze Li, Wei-Dong Zhao, Li-Yong Wu, Yu-Hua Chen, Ke Zhang","doi":"10.1186/s12974-024-03119-2","DOIUrl":"10.1186/s12974-024-03119-2","url":null,"abstract":"<p><strong>Background: </strong>Understanding the molecular mechanisms of Alzheimer's disease (AD) has important clinical implications for guiding therapy. Impaired amyloid beta (Aβ) clearance is critical in the pathogenesis of sporadic AD, and blood monocytes play an important role in Aβ clearance in the periphery. However, the mechanism underlying the defective phagocytosis of Aβ by monocytes in AD remains unclear.</p><p><strong>Methods: </strong>Initially, we collected whole blood samples from sporadic AD patients and isolated the monocytes for RNA sequencing analysis. By establishing APP/PS1 transgenic model mice with monocyte-specific cystatin F overexpression, we assessed the influence of monocyte-derived cystatin F on AD development. We further used a nondenaturing gel to identify the structure of the secreted cystatin F in plasma. Flow cytometry, enzyme-linked immunosorbent assays and laser scanning confocal microscopy were used to analyse the internalization of Aβ by monocytes. Pull down assays, bimolecular fluorescence complementation assays and total internal reflection fluorescence microscopy were used to determine the interactions and potential interactional amino acids between the cystatin F protein and Aβ. Finally, the cystatin F protein was purified and injected via the tail vein into 5XFAD mice to assess AD pathology.</p><p><strong>Results: </strong>Our results demonstrated that the expression of the cystatin F protein was specifically increased in the monocytes of AD patients. Monocyte-derived cystatin F increased Aβ deposition and exacerbated cognitive deficits in APP/PS1 mice. Furthermore, secreted cystatin F in the plasma of AD patients has a dimeric structure that is closely related to clinical signs of AD. Moreover, we noted that the cystatin F dimer blocks the phagocytosis of Aβ by monocytes. Mechanistically, the cystatin F dimer physically interacts with Aβ to inhibit its recognition and internalization by monocytes through certain amino acid interactions between the cystatin F dimer and Aβ. We found that high levels of the cystatin F dimer protein in blood contributed to amyloid pathology and cognitive deficits as a risk factor in 5XFAD mice.</p><p><strong>Conclusions: </strong>Our findings highlight that the cystatin F dimer plays a crucial role in regulating Aβ metabolism via its peripheral clearance pathway, providing us with a potential biomarker for diagnosis and potential target for therapeutic intervention.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11088181/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140903541","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
Deficiency of leucine-rich repeat kinase 2 aggravates thioacetamide-induced acute liver failure and hepatic encephalopathy in mice. 富亮氨酸重复激酶 2 缺乏会加重硫代乙酰胺诱导的小鼠急性肝衰竭和肝性脑病。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-05-09 DOI: 10.1186/s12974-024-03125-4
Dan Li, Shu-Fang Yu, Lin Lin, Jie-Ru Guo, Si-Mei Huang, Xi-Lin Wu, Han-Lin You, Xiao-Juan Cheng, Qiu-Yang Zhang, Yu-Qi Zeng, Xiao-Dong Pan
{"title":"Deficiency of leucine-rich repeat kinase 2 aggravates thioacetamide-induced acute liver failure and hepatic encephalopathy in mice.","authors":"Dan Li, Shu-Fang Yu, Lin Lin, Jie-Ru Guo, Si-Mei Huang, Xi-Lin Wu, Han-Lin You, Xiao-Juan Cheng, Qiu-Yang Zhang, Yu-Qi Zeng, Xiao-Dong Pan","doi":"10.1186/s12974-024-03125-4","DOIUrl":"10.1186/s12974-024-03125-4","url":null,"abstract":"<p><strong>Background: </strong>Hepatic encephalopathy (HE) is closely associated with inflammatory responses. However, as a crucial regulator of the immune and inflammatory responses, the role of leucine-rich repeat kinase 2 (LRRK2) in the pathogenesis of HE remains unraveled. Herein, we investigated this issue in thioacetamide (TAA)-induced HE following acute liver failure (ALF).</p><p><strong>Methods: </strong>TAA-induced HE mouse models of LRRK2 wild type (WT), LRRK2 G2019S mutation (Lrrk2<sup>G2019S</sup>) and LRRK2 knockout (Lrrk2<sup>-/-</sup>) were established. A battery of neurobehavioral experiments was conducted. The biochemical indexes and pro-inflammatory cytokines were detected. The prefrontal cortex (PFC), striatum (STR), hippocampus (HIP), and liver were examined by pathology and electron microscopy. The changes of autophagy-lysosomal pathway and activity of critical Rab GTPases were analyzed.</p><p><strong>Results: </strong>The Lrrk2<sup>-/-</sup>-HE model reported a significantly lower survival rate than the other two models (24% vs. 48%, respectively, p < 0.05), with no difference found between the WT-HE and Lrrk2<sup>G2019S</sup>-HE groups. Compared with the other groups, after the TAA injection, the Lrrk2<sup>-/-</sup> group displayed a significant increase in ammonium and pro-inflammatory cytokines, aggravated hepatic inflammation/necrosis, decreased autophagy, and abnormal phosphorylation of lysosomal Rab10. All three models reported microglial activation, neuronal loss, disordered vesicle transmission, and damaged myelin structure. The Lrrk2<sup>-/-</sup>-HE mice presented no severer neuronal injury than the other genotypes.</p><p><strong>Conclusions: </strong>LRRK2 deficiency may exacerbate TAA-induced ALF and HE in mice, in which inflammatory response is evident in the brain and aggravated in the liver. These novel findings indicate a need of sufficient clinical awareness of the adverse effects of LRRK2 inhibitors on the liver.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11084037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140898710","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