首页 > 最新文献

Journal of Neuroinflammation最新文献

英文 中文
Tyro3 and Gas6 are associated with white matter and myelin integrity in multiple sclerosis. Tyro3和Gas6与多发性硬化症患者的白质和髓鞘完整性有关。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-13 DOI: 10.1186/s12974-024-03315-0
Igal Rosenstein, Lenka Novakova, Hlin Kvartsberg, Anna Nordin, Sofia Rasch, Elzbieta Rembeza, Sofia Sandgren, Clas Malmeström, Stefanie Fruhwürth, Markus Axelsson, Kaj Blennow, Henrik Zetterberg, Jan Lycke

Background: The Gas6/TAM (Tyro3, Axl, and Mer) receptor system has been implicated in demyelination and delayed remyelination in experimental animal models, but data in humans are scarce. We aimed to investigate the role of Gas6/TAM in neurodegenerative processes in multiple sclerosis (MS).

Methods: From a prospective 5-year follow-up study, soluble Gas6/TAM biomarkers were analyzed in cerebrospinal fluid (CSF) by enzyme-linked immunosorbent assay (ELISA) at baseline in patients with relapsing-remitting MS (RRMS) (n = 40), progressive MS (PMS) (n = 20), and healthy controls (HC) (n = 25). Brain volumes, including myelin content (MyC) and white matter (WM) were measured by synthetic magnetic resonance imaging at baseline, 12 months, and 60-month follow-up. Associations with brain volume changes were investigated in multivariable linear regression models. Gas6/TAM concentrations were also determined at 12 months follow-up in RRMS to assess treatment response.

Results: Baseline concentrations of Tyro3, Axl, and Gas6 were significantly higher in PMS vs. RRMS and HC. Mer was higher in PMS vs. HC. Tyro3 and Gas6 were associated with reduced WM (β = 25.5, 95% confidence interval [CI] [6.11-44.96, p = 0.012; β = 11.4, 95% CI [0.42-22.4], p = 0.042, respectively) and MyC (β = 7.95, 95%CI [1.84-14.07], p = 0.012; β = 4.4, 95%CI [1.04-7.75], p = 0.012 respectively) at 60 months. Patients with evidence of remyelination at last follow-up had lower baseline soluble Tyro3 (p = 0.033) and Gas6 (p = 0.014). Except Mer, Gas6/TAM concentrations did not change with treatment in RRMS.

Discussion: Our data indicate a potential role for the Gas6/TAM receptor system in neurodegenerative processes influencing demyelination and ineffective remyelination.

背景:Gas6/TAM (Tyro3, Axl和Mer)受体系统在实验动物模型中与脱髓鞘和延迟的再髓鞘形成有关,但在人类中的数据很少。我们旨在研究Gas6/TAM在多发性硬化症(MS)神经退行性过程中的作用。方法:通过一项前瞻性5年随访研究,采用酶联免疫吸附试验(ELISA)分析了复发-缓解型多发性硬化症(RRMS) (n = 40)、进展型多发性硬化症(PMS) (n = 20)和健康对照(HC) (n = 25)患者基线时脑脊液(CSF)中可溶性Gas6/TAM生物标志物。在基线、12个月和60个月的随访中,通过合成磁共振成像测量脑容量,包括髓磷脂含量(MyC)和白质(WM)。在多变量线性回归模型中研究了与脑容量变化的关系。在RRMS随访12个月时测定Gas6/TAM浓度,以评估治疗反应。结果:PMS组Tyro3、Axl和Gas6的基线浓度明显高于RRMS和HC组。经前症候群的Mer高于HC。Tyro3和Gas6与WM降低相关(β = 25.5, 95%可信区间[CI] [6.11-44.96, p = 0.012;β= 11.4,95% CI(0.42 - -22.4),分别为p = 0.042)和MyC(β= 7.95,95% CI [1.84 - -14.07], p = 0.012;β= 4.4,95% ci(1.04 - -7.75),在60个月分别p = 0.012)。在最后一次随访中,有证据表明髓鞘再生的患者有较低的基线可溶性Tyro3 (p = 0.033)和Gas6 (p = 0.014)。除Mer外,RRMS中Gas6/TAM浓度不随治疗变化。讨论:我们的数据表明,Gas6/TAM受体系统在影响脱髓鞘和无效髓鞘再生的神经退行性过程中的潜在作用。
{"title":"Tyro3 and Gas6 are associated with white matter and myelin integrity in multiple sclerosis.","authors":"Igal Rosenstein, Lenka Novakova, Hlin Kvartsberg, Anna Nordin, Sofia Rasch, Elzbieta Rembeza, Sofia Sandgren, Clas Malmeström, Stefanie Fruhwürth, Markus Axelsson, Kaj Blennow, Henrik Zetterberg, Jan Lycke","doi":"10.1186/s12974-024-03315-0","DOIUrl":"10.1186/s12974-024-03315-0","url":null,"abstract":"<p><strong>Background: </strong>The Gas6/TAM (Tyro3, Axl, and Mer) receptor system has been implicated in demyelination and delayed remyelination in experimental animal models, but data in humans are scarce. We aimed to investigate the role of Gas6/TAM in neurodegenerative processes in multiple sclerosis (MS).</p><p><strong>Methods: </strong>From a prospective 5-year follow-up study, soluble Gas6/TAM biomarkers were analyzed in cerebrospinal fluid (CSF) by enzyme-linked immunosorbent assay (ELISA) at baseline in patients with relapsing-remitting MS (RRMS) (n = 40), progressive MS (PMS) (n = 20), and healthy controls (HC) (n = 25). Brain volumes, including myelin content (MyC) and white matter (WM) were measured by synthetic magnetic resonance imaging at baseline, 12 months, and 60-month follow-up. Associations with brain volume changes were investigated in multivariable linear regression models. Gas6/TAM concentrations were also determined at 12 months follow-up in RRMS to assess treatment response.</p><p><strong>Results: </strong>Baseline concentrations of Tyro3, Axl, and Gas6 were significantly higher in PMS vs. RRMS and HC. Mer was higher in PMS vs. HC. Tyro3 and Gas6 were associated with reduced WM (β = 25.5, 95% confidence interval [CI] [6.11-44.96, p = 0.012; β = 11.4, 95% CI [0.42-22.4], p = 0.042, respectively) and MyC (β = 7.95, 95%CI [1.84-14.07], p = 0.012; β = 4.4, 95%CI [1.04-7.75], p = 0.012 respectively) at 60 months. Patients with evidence of remyelination at last follow-up had lower baseline soluble Tyro3 (p = 0.033) and Gas6 (p = 0.014). Except Mer, Gas6/TAM concentrations did not change with treatment in RRMS.</p><p><strong>Discussion: </strong>Our data indicate a potential role for the Gas6/TAM receptor system in neurodegenerative processes influencing demyelination and ineffective remyelination.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"320"},"PeriodicalIF":9.3,"publicationDate":"2024-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11645787/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142822172","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
Glyphosate exposure exacerbates neuroinflammation and Alzheimer's disease-like pathology despite a 6-month recovery period in mice. 草甘膦暴露加剧了小鼠的神经炎症和阿尔茨海默病样病理,尽管有6个月的恢复期。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-04 DOI: 10.1186/s12974-024-03290-6
Samantha K Bartholomew, Wendy Winslow, Ritin Sharma, Khyatiben V Pathak, Savannah Tallino, Jessica M Judd, Hector Leon, Julie Turk, Patrick Pirrotte, Ramon Velazquez

Background: Glyphosate use in the United States (US) has increased each year since the introduction of glyphosate-tolerant crops in 1996, yet little is known about its effects on the brain. We recently found that C57BL/6J mice dosed with glyphosate for 14 days showed glyphosate and its major metabolite aminomethylphosphonic acid present in brain tissue, with corresponding increases in pro-inflammatory cytokine tumor necrosis factor-⍺ (TNF-⍺) in the brain and peripheral blood plasma. Since TNF-⍺ is elevated in neurodegenerative disorders such as Alzheimer's Disease (AD), in this study, we asked whether glyphosate exposure serves as an accelerant of AD pathogenesis. Additionally, whether glyphosate and aminomethylphosphonic acid remain in the brain after a recovery period has yet to be examined.

Methods: We hypothesized that glyphosate exposure would induce neuroinflammation in control mice, while exacerbating neuroinflammation in AD mice, causing elevated Amyloid-β and tau pathology and worsening spatial cognition after recovery. We dosed 4.5-month-old 3xTg-AD and non-transgenic (NonTg) control mice with either 0, 50 or 500 mg/kg of glyphosate daily for 13 weeks followed by a 6-month recovery period.

Results: We found that aminomethylphosphonic acid was detectable in the brains of 3xTg-AD and NonTg glyphosate-dosed mice despite the 6-month recovery. Glyphosate-dosed 3xTg-AD mice showed reduced survival, increased thigmotaxia in the Morris water maze, significant increases in the beta secretase enzyme (BACE-1) of amyloidogenic processing, amyloid-β (Aβ) 42 insoluble fractions, Aβ 42 plaque load and plaque size, and phosphorylated tau (pTau) at epitopes Threonine 181, Serine 396, and AT8 (Serine 202, Threonine 205). Notably, we found increased pro- and anti-inflammatory cytokines and chemokines persisting in both 3xTg-AD and NonTg brain tissue and in 3xTg-AD peripheral blood plasma.

Conclusion: Taken together, our results are the first to demonstrate that despite an extended recovery period, exposure to glyphosate elicits long-lasting pathological consequences. As glyphosate use continues to rise, more research is needed to elucidate the impact of this herbicide and its metabolites on the human brain, and their potential to contribute to dysfunctions observed in neurodegenerative diseases.

背景:自1996年引进抗草甘膦作物以来,草甘膦在美国的使用量每年都在增加,但对其对大脑的影响知之甚少。我们最近发现,C57BL/6J小鼠给予草甘膦14天后,脑组织中出现草甘膦及其主要代谢物氨基甲基膦酸,脑组织和外周血中促炎细胞因子肿瘤坏死因子- TNF-升高。由于TNF-在神经退行性疾病如阿尔茨海默病(AD)中升高,在本研究中,我们询问草甘膦暴露是否作为AD发病机制的促进剂。此外,草甘膦和氨基甲基膦酸在恢复期后是否仍留在大脑中还有待研究。方法:我们假设草甘膦暴露会诱发对照小鼠的神经炎症,同时加重AD小鼠的神经炎症,导致淀粉样蛋白-β和tau病理升高,恢复后的空间认知能力下降。我们给4.5个月大的3xTg-AD和非转基因(NonTg)对照小鼠每天分别给予0、50或500 mg/kg的草甘膦,持续13周,然后进行6个月的恢复期。结果:我们发现3xTg-AD和NonTg草甘膦剂量小鼠的大脑中尽管恢复了6个月,但仍可检测到氨基甲基膦酸。草甘磷剂量的3xTg-AD小鼠表现出存活率降低,Morris水迷宫中thigmotaxia增加,淀粉样蛋白加工的β分泌酶(BACE-1),淀粉样蛋白-β (Aβ) 42不溶性部分,Aβ 42斑块负荷和斑块大小显著增加,磷酸化tau蛋白(pTau)在表位苏氨酸181,丝氨酸396和AT8(丝氨酸202,苏氨酸205)。值得注意的是,我们发现3xTg-AD和非tg脑组织以及3xTg-AD外周血血浆中亲炎性和抗炎性细胞因子和趋化因子持续增加。结论:综上所述,我们的研究结果首次证明,尽管恢复期延长,暴露于草甘膦会引起长期的病理后果。随着草甘膦使用量的持续增加,需要更多的研究来阐明这种除草剂及其代谢物对人类大脑的影响,以及它们可能导致神经退行性疾病中观察到的功能障碍。
{"title":"Glyphosate exposure exacerbates neuroinflammation and Alzheimer's disease-like pathology despite a 6-month recovery period in mice.","authors":"Samantha K Bartholomew, Wendy Winslow, Ritin Sharma, Khyatiben V Pathak, Savannah Tallino, Jessica M Judd, Hector Leon, Julie Turk, Patrick Pirrotte, Ramon Velazquez","doi":"10.1186/s12974-024-03290-6","DOIUrl":"10.1186/s12974-024-03290-6","url":null,"abstract":"<p><strong>Background: </strong>Glyphosate use in the United States (US) has increased each year since the introduction of glyphosate-tolerant crops in 1996, yet little is known about its effects on the brain. We recently found that C57BL/6J mice dosed with glyphosate for 14 days showed glyphosate and its major metabolite aminomethylphosphonic acid present in brain tissue, with corresponding increases in pro-inflammatory cytokine tumor necrosis factor-⍺ (TNF-⍺) in the brain and peripheral blood plasma. Since TNF-⍺ is elevated in neurodegenerative disorders such as Alzheimer's Disease (AD), in this study, we asked whether glyphosate exposure serves as an accelerant of AD pathogenesis. Additionally, whether glyphosate and aminomethylphosphonic acid remain in the brain after a recovery period has yet to be examined.</p><p><strong>Methods: </strong>We hypothesized that glyphosate exposure would induce neuroinflammation in control mice, while exacerbating neuroinflammation in AD mice, causing elevated Amyloid-β and tau pathology and worsening spatial cognition after recovery. We dosed 4.5-month-old 3xTg-AD and non-transgenic (NonTg) control mice with either 0, 50 or 500 mg/kg of glyphosate daily for 13 weeks followed by a 6-month recovery period.</p><p><strong>Results: </strong>We found that aminomethylphosphonic acid was detectable in the brains of 3xTg-AD and NonTg glyphosate-dosed mice despite the 6-month recovery. Glyphosate-dosed 3xTg-AD mice showed reduced survival, increased thigmotaxia in the Morris water maze, significant increases in the beta secretase enzyme (BACE-1) of amyloidogenic processing, amyloid-β (Aβ) 42 insoluble fractions, Aβ 42 plaque load and plaque size, and phosphorylated tau (pTau) at epitopes Threonine 181, Serine 396, and AT8 (Serine 202, Threonine 205). Notably, we found increased pro- and anti-inflammatory cytokines and chemokines persisting in both 3xTg-AD and NonTg brain tissue and in 3xTg-AD peripheral blood plasma.</p><p><strong>Conclusion: </strong>Taken together, our results are the first to demonstrate that despite an extended recovery period, exposure to glyphosate elicits long-lasting pathological consequences. As glyphosate use continues to rise, more research is needed to elucidate the impact of this herbicide and its metabolites on the human brain, and their potential to contribute to dysfunctions observed in neurodegenerative diseases.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"316"},"PeriodicalIF":9.3,"publicationDate":"2024-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11619132/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142780330","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
Autoantibody profiles in Alzheimer´s, Parkinson´s, and dementia with Lewy bodies: altered IgG affinity and IgG/IgM/IgA responses to alpha-synuclein, amyloid-beta, and tau in disease-specific pathological patterns. 阿尔茨海默病、帕金森病和路易体痴呆的自身抗体谱:在疾病特异性病理模式中,对α -突触核蛋白、β -淀粉样蛋白和tau蛋白的IgG亲和力和IgG/IgM/IgA反应发生改变
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-03 DOI: 10.1186/s12974-024-03293-3
Luisa Knecht, Katrine Dalsbøl, Anja Hviid Simonsen, Falk Pilchner, Jean Alexander Ross, Kristian Winge, Lisette Salvesen, Sara Bech, Anne-Mette Hejl, Annemette Løkkegaard, Steen G Hasselbalch, Richard Dodel, Susana Aznar, Gunhild Waldemar, Tomasz Brudek, Jonas Folke

Background: Alzheimer's disease (AD) and Parkinson's disease (PD) are leading neurodegenerative disorders marked by protein aggregation, with AD featuring amyloid-beta (Aβ) and tau proteins, and PD alpha-synuclein (αSyn). Dementia with Lewy bodies (DLB) often presents with a mix of these pathologies. This study explores naturally occurring autoantibodies (nAbs), including Immunoglobulin (Ig)G, IgM, and IgA, which target αSyn, Aβ and tau to maintain homeostasis and were previously found altered in AD and PD patients, among others.

Main text: We extended this investigation across AD, PD and DLB patients investigating both the affinities of IgGs and levels of IgGs, IgMs and IgAs towards αSyn, Aβ and tau utilizing chemiluminescence assays. We confirmed that AD and PD patients exhibited lower levels of high-affinity anti-Aβ and anti-αSyn IgGs, respectively, than healthy controls. AD patients also showed diminished levels of high-affinity anti-αSyn IgGs, while anti-tau IgG affinities did not differ significantly across groups. However, DLB patients exhibited increased anti-αSyn IgG but decreased anti-αSyn IgM levels compared to controls and PD patients, with AD patients showing a similar pattern. Interestingly, AD patients had higher anti-Aβ IgG but lower anti-Aβ IgA levels than DLB patients. DLB patients had reduced anti-Aβ IgM levels compared to controls, and anti-tau IgG levels were lower in AD than PD patients, who had reduced anti-tau IgM levels compared to controls. AD patients uniquely showed higher anti-tau IgA levels. Significant correlations were observed between clinical measures and nAbs, with negative correlations between anti-αSyn IgG affinity and levels in DLB patients and a positive correlation with anti-αSyn IgA levels in PD patients. Disease-specific changes in nAb levels and affinity correlations were identified, highlighting altered immune responses.

Conclusion: This study reveals distinctive nAb profiles in AD, DLB, and PD, pinpointing specific immune deficiencies against pathological proteins. These insights into the autoreactive immune system's role in neurodegeneration suggest nAbs as potential markers for vulnerability to protein aggregation, offering new avenues for understanding and possibly diagnosing these conditions.

背景:阿尔茨海默病(AD)和帕金森病(PD)是主要的以蛋白聚集为特征的神经退行性疾病,AD以淀粉样蛋白- β (Aβ)和tau蛋白以及PD α -突触核蛋白(αSyn)为特征。路易体痴呆(DLB)通常表现为这些病理的混合。本研究探索了天然存在的自身抗体(nab),包括免疫球蛋白(Ig)G, IgM和IgA,它们靶向αSyn, Aβ和tau来维持体内平衡,并且先前发现在AD和PD患者中发生改变。我们将这项研究扩展到AD、PD和DLB患者,利用化学发光法研究了igg的亲和力以及igg、IgMs和IgAs对αSyn、Aβ和tau的水平。我们证实,AD和PD患者分别表现出低于健康对照组的高亲和力抗a β和抗α syn igg水平。AD患者的高亲和力抗α - syn IgG水平也下降,而抗tau IgG的亲和力在组间无显著差异。然而,与对照组和PD患者相比,DLB患者抗α syn IgG水平升高,而抗α syn IgM水平降低,AD患者也表现出类似的模式。有趣的是,AD患者的抗β IgG水平高于DLB患者,而抗β IgA水平低于DLB患者。与对照组相比,DLB患者的抗β IgM水平降低,AD患者的抗tau IgG水平低于PD患者,PD患者的抗tau IgM水平低于对照组。AD患者独有地显示出更高的抗tau IgA水平。临床指标与nab呈显著相关,DLB患者抗α syn IgG亲和力与水平呈负相关,PD患者抗α syn IgA水平呈正相关。确定了nAb水平和亲和相关性的疾病特异性变化,突出了免疫反应的改变。结论:本研究揭示了AD, DLB和PD中独特的nAb谱,确定了针对病理蛋白的特异性免疫缺陷。这些对自身反应性免疫系统在神经退行性疾病中的作用的见解表明,nab是蛋白质聚集易感性的潜在标记物,为理解和可能诊断这些疾病提供了新的途径。
{"title":"Autoantibody profiles in Alzheimer´s, Parkinson´s, and dementia with Lewy bodies: altered IgG affinity and IgG/IgM/IgA responses to alpha-synuclein, amyloid-beta, and tau in disease-specific pathological patterns.","authors":"Luisa Knecht, Katrine Dalsbøl, Anja Hviid Simonsen, Falk Pilchner, Jean Alexander Ross, Kristian Winge, Lisette Salvesen, Sara Bech, Anne-Mette Hejl, Annemette Løkkegaard, Steen G Hasselbalch, Richard Dodel, Susana Aznar, Gunhild Waldemar, Tomasz Brudek, Jonas Folke","doi":"10.1186/s12974-024-03293-3","DOIUrl":"10.1186/s12974-024-03293-3","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) and Parkinson's disease (PD) are leading neurodegenerative disorders marked by protein aggregation, with AD featuring amyloid-beta (Aβ) and tau proteins, and PD alpha-synuclein (αSyn). Dementia with Lewy bodies (DLB) often presents with a mix of these pathologies. This study explores naturally occurring autoantibodies (nAbs), including Immunoglobulin (Ig)G, IgM, and IgA, which target αSyn, Aβ and tau to maintain homeostasis and were previously found altered in AD and PD patients, among others.</p><p><strong>Main text: </strong>We extended this investigation across AD, PD and DLB patients investigating both the affinities of IgGs and levels of IgGs, IgMs and IgAs towards αSyn, Aβ and tau utilizing chemiluminescence assays. We confirmed that AD and PD patients exhibited lower levels of high-affinity anti-Aβ and anti-αSyn IgGs, respectively, than healthy controls. AD patients also showed diminished levels of high-affinity anti-αSyn IgGs, while anti-tau IgG affinities did not differ significantly across groups. However, DLB patients exhibited increased anti-αSyn IgG but decreased anti-αSyn IgM levels compared to controls and PD patients, with AD patients showing a similar pattern. Interestingly, AD patients had higher anti-Aβ IgG but lower anti-Aβ IgA levels than DLB patients. DLB patients had reduced anti-Aβ IgM levels compared to controls, and anti-tau IgG levels were lower in AD than PD patients, who had reduced anti-tau IgM levels compared to controls. AD patients uniquely showed higher anti-tau IgA levels. Significant correlations were observed between clinical measures and nAbs, with negative correlations between anti-αSyn IgG affinity and levels in DLB patients and a positive correlation with anti-αSyn IgA levels in PD patients. Disease-specific changes in nAb levels and affinity correlations were identified, highlighting altered immune responses.</p><p><strong>Conclusion: </strong>This study reveals distinctive nAb profiles in AD, DLB, and PD, pinpointing specific immune deficiencies against pathological proteins. These insights into the autoreactive immune system's role in neurodegeneration suggest nAbs as potential markers for vulnerability to protein aggregation, offering new avenues for understanding and possibly diagnosing these conditions.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"317"},"PeriodicalIF":9.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11613470/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769854","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
Ketogenic diet modulates immune cell transcriptional landscape and ameliorates experimental autoimmune uveitis in mice. 生酮饮食调节免疫细胞转录景观和改善实验性自身免疫性葡萄膜炎小鼠。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-03 DOI: 10.1186/s12974-024-03308-z
Runping Duan, Tianfu Wang, Zhaohuai Li, Loujing Jiang, Xiaoyang Yu, Daquan He, Tianyu Tao, Xiuxing Liu, Zhaohao Huang, Lei Feng, Wenru Su

Background: Uveitis manifests as immune-mediated inflammatory disorders within the eye, posing a serious threat to vision. The ketogenic diet (KD) has emerged as a promising dietary intervention, yet its impact on the immune microenvironments and role in uveitis remains unclear.

Methods: Utilizing single-cell RNA sequencing (scRNA-seq) data from lymph node and retina of mice, we conduct a comprehensive investigation into the effects of KD on immune microenvironments. Flow cytometry is conducted to verify the potential mechanisms.

Results: This study demonstrates that KD alters the composition and function of immune profiles. Specifically, KD promotes the differentiation of Treg cells and elevates its proportion in heathy mice. In response to experimental autoimmune uveitis challenges, KD alleviates the inflammatory symptoms, lowers CD4+ T cell pathogenicity, and corrects the Th17/Treg imbalance. Additionally, KD decreases the proportion of Th17 cell and increases Treg cells in the retina. Analysis of combined retinal and CDLN immune cells reveals that retinal immune cells, particularly CD4+ T cells, exhibit heightened inflammatory responses, which KD partially reverses.

Conclusions: The KD induces inhibitory structural and functional alterations in immune cells from lymph nodes to retina, suggesting its potential as a therapy for uveitis.

背景:葡萄膜炎表现为眼内免疫介导的炎症性疾病,对视力构成严重威胁。生酮饮食(KD)已成为一种有前景的饮食干预,但其对免疫微环境的影响及其在葡萄膜炎中的作用尚不清楚。方法:利用小鼠淋巴结和视网膜的单细胞RNA测序(scRNA-seq)数据,全面研究KD对免疫微环境的影响。流式细胞术验证了潜在的机制。结果:本研究表明KD改变了免疫谱的组成和功能。具体而言,KD促进Treg细胞的分化,并提高其在健康小鼠中的比例。在应对实验性自身免疫性葡萄膜炎挑战时,KD可缓解炎症症状,降低CD4+ T细胞致病性,并纠正Th17/Treg失衡。此外,KD降低了Th17细胞的比例,增加了视网膜中Treg细胞的比例。对视网膜和ccdn联合免疫细胞的分析显示,视网膜免疫细胞,特别是CD4+ T细胞,表现出增强的炎症反应,KD部分逆转了这一反应。结论:KD诱导免疫细胞从淋巴结到视网膜的抑制性结构和功能改变,提示其治疗葡萄膜炎的潜力。
{"title":"Ketogenic diet modulates immune cell transcriptional landscape and ameliorates experimental autoimmune uveitis in mice.","authors":"Runping Duan, Tianfu Wang, Zhaohuai Li, Loujing Jiang, Xiaoyang Yu, Daquan He, Tianyu Tao, Xiuxing Liu, Zhaohao Huang, Lei Feng, Wenru Su","doi":"10.1186/s12974-024-03308-z","DOIUrl":"10.1186/s12974-024-03308-z","url":null,"abstract":"<p><strong>Background: </strong>Uveitis manifests as immune-mediated inflammatory disorders within the eye, posing a serious threat to vision. The ketogenic diet (KD) has emerged as a promising dietary intervention, yet its impact on the immune microenvironments and role in uveitis remains unclear.</p><p><strong>Methods: </strong>Utilizing single-cell RNA sequencing (scRNA-seq) data from lymph node and retina of mice, we conduct a comprehensive investigation into the effects of KD on immune microenvironments. Flow cytometry is conducted to verify the potential mechanisms.</p><p><strong>Results: </strong>This study demonstrates that KD alters the composition and function of immune profiles. Specifically, KD promotes the differentiation of Treg cells and elevates its proportion in heathy mice. In response to experimental autoimmune uveitis challenges, KD alleviates the inflammatory symptoms, lowers CD4<sup>+</sup> T cell pathogenicity, and corrects the Th17/Treg imbalance. Additionally, KD decreases the proportion of Th17 cell and increases Treg cells in the retina. Analysis of combined retinal and CDLN immune cells reveals that retinal immune cells, particularly CD4<sup>+</sup> T cells, exhibit heightened inflammatory responses, which KD partially reverses.</p><p><strong>Conclusions: </strong>The KD induces inhibitory structural and functional alterations in immune cells from lymph nodes to retina, suggesting its potential as a therapy for uveitis.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"319"},"PeriodicalIF":9.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11613848/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769856","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
Metformin attenuates central sensitization by regulating neuroinflammation through the TREM2-SYK signaling pathway in a mouse model of chronic migraine. 在慢性偏头痛小鼠模型中,二甲双胍通过TREM2-SYK信号通路调节神经炎症,从而减轻中枢致敏。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-03 DOI: 10.1186/s12974-024-03313-2
Zhenzhen Fan, Dandan Su, Zi Chao Li, Songtang Sun, Zhaoming Ge

Background: Chronic migraine (CM) is a serious neurological disorder. Central sensitization is one of the important pathophysiological mechanisms underlying CM, and microglia-induced neuroinflammation conduces to central sensitization. Triggering receptor expressed on myeloid cells 2 (TREM2) is presented solely in microglia residing within the central nervous system and plays a key role in neuroinflammation. Metformin has been shown to regulate inflammatory responses and exert analgesic effects, but its relationship with CM remains unclear. In the study, we investigated whether metformin modulates TREM2 to improve central sensitization of CM and clarified the potential molecular mechanisms.

Methods: A CM mouse model was induced by administration of nitroglycerin (NTG). Behavioral evaluations were conducted using von Frey filaments and hot plate experiments. Western blot and immunofluorescence techniques were employed to investigate the molecular mechanisms. Metformin and the SYK inhibitor R406 were administered to mice to assess their regulatory effects on neuroinflammation and central sensitization. To explore the role of TREM2-SYK in regulating neuroinflammation with metformin, a lentivirus encoding TREM2 was injected into the trigeminal nucleus caudalis (TNC). In vitro experiments were conducted to evaluate the regulation of TREM2-SYK by metformin, involving interventions with LPS, metformin, R406, siTREM2, and TREM2 plasmids.

Results: Metformin and R406 pretreatment can effectively improve hyperalgesia in CM mice. Both metformin and R406 significantly inhibit c-fos and CGRP expression in CM mice, effectively suppressing the activation of microglia and NLRP3 inflammasome induced by NTG. With the administration of NTG, TREM2 expression gradually increased in TNC microglia. Additionally, we observed that metformin significantly inhibits TREM2 and SYK expression in CM mice. Lv-TREM2 attenuated metformin-mediated anti-inflammatory responses. In vitro experiments, knockdown of TREM2 inhibited LPS-induced SYK pathway activation and alleviated inflammatory responses. After the sole overexpression of TREM2, the SYK signaling pathway is activated, resulting in the activation of the NLRP3 inflammasome and an increased expression of pro-inflammatory cytokines; nevertheless, this consequence can be reversed by R406. The overexpression of TREM2 attenuates the inhibition of SYK activity mediated by metformin, and this effect can be reversed by R406.

Conclusions: Our findings suggest that metformin attenuates central sensitization in CM by regulating the activation of microglia and NLRP3 inflammasome through the TREM2-SYK pathway.

背景:慢性偏头痛是一种严重的神经系统疾病。中枢致敏是CM的重要病理生理机制之一,小胶质细胞诱导的神经炎症有助于中枢致敏。髓样细胞2触发受体(TREM2)仅存在于中枢神经系统的小胶质细胞中,在神经炎症中起关键作用。二甲双胍已被证明可调节炎症反应并发挥镇痛作用,但其与CM的关系尚不清楚。在这项研究中,我们研究了二甲双胍是否通过调节TREM2来改善CM的中枢致敏,并阐明了潜在的分子机制。方法:用硝酸甘油(NTG)诱导CM小鼠模型。采用von Frey细丝和热板实验进行行为评价。采用Western blot和免疫荧光技术研究其分子机制。给小鼠二甲双胍和SYK抑制剂R406,以评估其对神经炎症和中枢致敏的调节作用。为了探讨TREM2- syk在二甲双胍调节神经炎症中的作用,将编码TREM2的慢病毒注射到三叉神经尾核(TNC)中。体外实验评估二甲双胍对TREM2- syk的调控作用,包括LPS、二甲双胍、R406、siTREM2和TREM2质粒的干预。结果:二甲双胍和R406预处理能有效改善CM小鼠痛觉过敏。二甲双胍和R406均能显著抑制CM小鼠c-fos和CGRP的表达,有效抑制NTG诱导的小胶质细胞和NLRP3炎性体的活化。随着NTG的给药,TREM2在TNC小胶质细胞中的表达逐渐升高。此外,我们观察到二甲双胍显著抑制CM小鼠TREM2和SYK的表达。Lv-TREM2减弱二甲双胍介导的抗炎反应。在体外实验中,敲低TREM2可抑制lps诱导的SYK通路激活,减轻炎症反应。TREM2单独过表达后,SYK信号通路被激活,导致NLRP3炎性小体激活,促炎细胞因子表达增加;然而,这种结果可以通过R406逆转。TREM2过表达可减弱二甲双胍介导的SYK活性抑制,R406可逆转这一作用。结论:我们的研究结果表明,二甲双胍通过TREM2-SYK途径调节小胶质细胞和NLRP3炎性体的激活,从而减轻CM的中枢致敏。
{"title":"Metformin attenuates central sensitization by regulating neuroinflammation through the TREM2-SYK signaling pathway in a mouse model of chronic migraine.","authors":"Zhenzhen Fan, Dandan Su, Zi Chao Li, Songtang Sun, Zhaoming Ge","doi":"10.1186/s12974-024-03313-2","DOIUrl":"10.1186/s12974-024-03313-2","url":null,"abstract":"<p><strong>Background: </strong>Chronic migraine (CM) is a serious neurological disorder. Central sensitization is one of the important pathophysiological mechanisms underlying CM, and microglia-induced neuroinflammation conduces to central sensitization. Triggering receptor expressed on myeloid cells 2 (TREM2) is presented solely in microglia residing within the central nervous system and plays a key role in neuroinflammation. Metformin has been shown to regulate inflammatory responses and exert analgesic effects, but its relationship with CM remains unclear. In the study, we investigated whether metformin modulates TREM2 to improve central sensitization of CM and clarified the potential molecular mechanisms.</p><p><strong>Methods: </strong>A CM mouse model was induced by administration of nitroglycerin (NTG). Behavioral evaluations were conducted using von Frey filaments and hot plate experiments. Western blot and immunofluorescence techniques were employed to investigate the molecular mechanisms. Metformin and the SYK inhibitor R406 were administered to mice to assess their regulatory effects on neuroinflammation and central sensitization. To explore the role of TREM2-SYK in regulating neuroinflammation with metformin, a lentivirus encoding TREM2 was injected into the trigeminal nucleus caudalis (TNC). In vitro experiments were conducted to evaluate the regulation of TREM2-SYK by metformin, involving interventions with LPS, metformin, R406, siTREM2, and TREM2 plasmids.</p><p><strong>Results: </strong>Metformin and R406 pretreatment can effectively improve hyperalgesia in CM mice. Both metformin and R406 significantly inhibit c-fos and CGRP expression in CM mice, effectively suppressing the activation of microglia and NLRP3 inflammasome induced by NTG. With the administration of NTG, TREM2 expression gradually increased in TNC microglia. Additionally, we observed that metformin significantly inhibits TREM2 and SYK expression in CM mice. Lv-TREM2 attenuated metformin-mediated anti-inflammatory responses. In vitro experiments, knockdown of TREM2 inhibited LPS-induced SYK pathway activation and alleviated inflammatory responses. After the sole overexpression of TREM2, the SYK signaling pathway is activated, resulting in the activation of the NLRP3 inflammasome and an increased expression of pro-inflammatory cytokines; nevertheless, this consequence can be reversed by R406. The overexpression of TREM2 attenuates the inhibition of SYK activity mediated by metformin, and this effect can be reversed by R406.</p><p><strong>Conclusions: </strong>Our findings suggest that metformin attenuates central sensitization in CM by regulating the activation of microglia and NLRP3 inflammasome through the TREM2-SYK pathway.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"318"},"PeriodicalIF":9.3,"publicationDate":"2024-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11613737/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769857","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
TRPM7 contributes to pyroptosis and its involvement in status epilepticus. TRPM7参与焦亡并参与癫痫持续状态。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-01 DOI: 10.1186/s12974-024-03292-4
Xin Tong, Yu Tong, Jiahe Zheng, Ruixue Shi, Hongyue Liang, Meixuan Li, Yulu Meng, Jian Shi, Dongyi Zhao, Corey Ray Seehus, Jialu Wang, Xiaoxue Xu, Tomasz Boczek, Sayuri Suzuki, Andrea Fleig, Reinhold Penner, Naining Zhang, Jianjun Xu, Jingjing Duan, Zhiyi Yu, Wuyang Wang, Weidong Zhao, Feng Guo

Background: Pyroptosis, a novel form of programmed cell death, has been implicated in neurodegeneration diseases. However, its role in status epilepticus (SE)-a condition characterized by prolonged or repeated seizures-remains inadequately understood.

Methods: SE were induced by intraperitoneal injection of pilocarpine (PILO). Neuronal excitability was assessed through electroencephalogram (EEG) recordings and patch clamp. Chromatin immunoprecipitation (ChIP) assay was applied to verify the interaction of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) protein with the promoters of Nlrp3 (the gene encoding NOD-like receptor family pyrin domain containing 3) and Trpm7 (transient receptor potential melastatin 7). To further investigate the role of TRPM7 in SE, AAV-sh-TRPM7-EGFP transfected mice and TRPM7 conditional knockout (TRPM7-CKO) mice were utilized.

Results: Our findings revealed elevated levels of IL-18 and IL-1β levels in primary epilepsy patients, along with increased expression level of the TRPM7 in SE models. Knockdown of TRPM7 alleviated neuronal damage and pyroptosis, reversing PILO-treated neuronal hyperexcitability. We demonstrated that p-STAT3 binds to the promoters of both Trpm7 and Nlrp3, modulating their transcriptions in SE. Importantly, inhibition of TRPM7 with NS8593, and inflammasome inhibition with MCC950, alleviated neuronal hyperexcitability and pyroptosis in SE. A new compound, SDUY-225, formulated based on the structure of NS8593 mitigated neuronal damage, pyroptosis, and hyperexcitability.

Conclusions: TRPM7 contributes to pyroptosis in SE, establishing a positive feedback loop involving the p-STAT3/TRPM7/Zn2+/p-STAT3 signaling pathway. Findings in this study raise the possibility that targeting TRPM7 and NLRP3 represents a promising therapeutic approach for SE.

背景:焦亡是一种新的程序性细胞死亡形式,与神经退行性疾病有关。然而,它在癫痫持续状态(SE)中的作用仍未充分了解,SE是一种以长时间或反复发作为特征的疾病。方法:腹腔注射匹罗卡品(pilocarpine, PILO)诱导SE。通过脑电图记录和膜片钳评估神经元兴奋性。采用染色质免疫沉淀法(ChIP)验证磷酸化的信号传导和转录激活因子3 (p-STAT3)蛋白与Nlrp3(编码nod样受体家族pyrin结构域3的基因)和Trpm7(瞬时受体电位美拉他素7)启动子的相互作用。为了进一步研究Trpm7在SE中的作用,我们利用AAV-sh-TRPM7-EGFP转染小鼠和Trpm7条件敲除(Trpm7 - cko)小鼠。结果:我们的研究结果显示,原发性癫痫患者IL-18和IL-1β水平升高,SE模型中TRPM7表达水平升高。TRPM7的下调减轻了神经元损伤和焦亡,逆转了pilo治疗的神经元高兴奋性。我们证明p-STAT3结合Trpm7和Nlrp3的启动子,调节它们在SE中的转录。重要的是,用NS8593抑制TRPM7,用MCC950抑制炎性体,减轻了SE神经元的高兴奋性和焦细胞凋亡。一种基于NS8593结构的新化合物SDUY-225减轻了神经元损伤、焦亡和高兴奋性。结论:TRPM7参与SE的焦亡,建立了p-STAT3/TRPM7/Zn2+/p-STAT3信号通路的正反馈回路。本研究的发现表明,靶向TRPM7和NLRP3可能是治疗SE的一种有希望的方法。
{"title":"TRPM7 contributes to pyroptosis and its involvement in status epilepticus.","authors":"Xin Tong, Yu Tong, Jiahe Zheng, Ruixue Shi, Hongyue Liang, Meixuan Li, Yulu Meng, Jian Shi, Dongyi Zhao, Corey Ray Seehus, Jialu Wang, Xiaoxue Xu, Tomasz Boczek, Sayuri Suzuki, Andrea Fleig, Reinhold Penner, Naining Zhang, Jianjun Xu, Jingjing Duan, Zhiyi Yu, Wuyang Wang, Weidong Zhao, Feng Guo","doi":"10.1186/s12974-024-03292-4","DOIUrl":"https://doi.org/10.1186/s12974-024-03292-4","url":null,"abstract":"<p><strong>Background: </strong>Pyroptosis, a novel form of programmed cell death, has been implicated in neurodegeneration diseases. However, its role in status epilepticus (SE)-a condition characterized by prolonged or repeated seizures-remains inadequately understood.</p><p><strong>Methods: </strong>SE were induced by intraperitoneal injection of pilocarpine (PILO). Neuronal excitability was assessed through electroencephalogram (EEG) recordings and patch clamp. Chromatin immunoprecipitation (ChIP) assay was applied to verify the interaction of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) protein with the promoters of Nlrp3 (the gene encoding NOD-like receptor family pyrin domain containing 3) and Trpm7 (transient receptor potential melastatin 7). To further investigate the role of TRPM7 in SE, AAV-sh-TRPM7-EGFP transfected mice and TRPM7 conditional knockout (TRPM7-CKO) mice were utilized.</p><p><strong>Results: </strong>Our findings revealed elevated levels of IL-18 and IL-1β levels in primary epilepsy patients, along with increased expression level of the TRPM7 in SE models. Knockdown of TRPM7 alleviated neuronal damage and pyroptosis, reversing PILO-treated neuronal hyperexcitability. We demonstrated that p-STAT3 binds to the promoters of both Trpm7 and Nlrp3, modulating their transcriptions in SE. Importantly, inhibition of TRPM7 with NS8593, and inflammasome inhibition with MCC950, alleviated neuronal hyperexcitability and pyroptosis in SE. A new compound, SDUY-225, formulated based on the structure of NS8593 mitigated neuronal damage, pyroptosis, and hyperexcitability.</p><p><strong>Conclusions: </strong>TRPM7 contributes to pyroptosis in SE, establishing a positive feedback loop involving the p-STAT3/TRPM7/Zn<sup>2+</sup>/p-STAT3 signaling pathway. Findings in this study raise the possibility that targeting TRPM7 and NLRP3 represents a promising therapeutic approach for SE.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"315"},"PeriodicalIF":9.3,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11608501/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769871","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
Sitagliptin eye drops prevent the impairment of retinal neurovascular unit in the new Trpv2+/- rat model. 西格列汀滴眼液对Trpv2+/-模型大鼠视网膜神经血管单元损伤的预防作用。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s12974-024-03283-5
Hugo Ramos, Josy Augustine, Burak M Karan, Cristina Hernández, Alan W Stitt, Tim M Curtis, Rafael Simó

Impaired function of the retinal neurovascular unit (NVU) is an early event in diabetic retinopathy (DR). It has been previously shown that topical delivery of the dipeptidyl peptidase-4 (DPP-4) inhibitor sitagliptin can protect against diabetes-mediated dysfunction of the retinal NVU in the db/db mouse. The aim of the present study was to examine whether sitagliptin could prevent the DR-like lesions within the NVU of the new non-diabetic model of DR, the Trpv2 knockout rat (Trpv2+/-). For that purpose, at 3 months of age, Trpv2+/- rats were topically treated twice daily for two weeks with sitagliptin or PBS-vehicle eyedrops. Trpv2+/+ rats treated with vehicle served as the control group. Body weight and glycemia were monitored. Optical coherence tomography recordings, fundus images and retinal samples were obtained to evaluate sitagliptin effects. The results revealed that sitagliptin eye drops had no effect on body weight or glycemia. Vehicle-treated Trpv2+/- rats exhibited retinal thinning and larger diameters of major retinal blood vessels, upregulation of inflammatory factors and oxidative markers, glial activation and formation of acellular capillaries. However, topical administration of sitagliptin significantly prevented all these abnormalities. In conclusion, sitagliptin eye drops exert a protective effect against DR-like lesions in Trpv2+/- rats. Our results suggest that sitagliptin eye drops carry significant potential to treat not only early-stages of DR but also other diseases with impairment of the NVU unrelated to diabetes.

视网膜神经血管单元(NVU)功能受损是糖尿病视网膜病变(DR)的早期事件。先前的研究表明,局部递送二肽基肽酶-4 (DPP-4)抑制剂西格列汀可以预防糖尿病介导的db/db小鼠视网膜NVU功能障碍。本研究的目的是研究西格列汀是否可以预防新型非糖尿病DR模型Trpv2敲除大鼠(Trpv2+/-) NVU内的DR样病变。为此,在3个月大的时候,Trpv2+/-大鼠每天局部治疗两次,连续两周使用西格列汀或pbs载体滴眼液。以Trpv2+/+大鼠为对照组。监测体重和血糖。获得光学相干断层扫描记录、眼底图像和视网膜样本来评估西格列汀的效果。结果显示西格列汀滴眼液对体重和血糖没有影响。Trpv2+/-处理大鼠视网膜变薄,主要视网膜血管直径变大,炎症因子和氧化标志物上调,胶质细胞活化,无细胞毛细血管形成。然而,局部使用西格列汀可显著预防所有这些异常。综上所述,西格列汀滴眼液对Trpv2+/-大鼠dr样病变具有保护作用。我们的研究结果表明,西格列汀滴眼液不仅具有治疗早期DR的巨大潜力,而且还具有治疗与糖尿病无关的其他NVU损害疾病的潜力。
{"title":"Sitagliptin eye drops prevent the impairment of retinal neurovascular unit in the new Trpv2<sup>+/-</sup> rat model.","authors":"Hugo Ramos, Josy Augustine, Burak M Karan, Cristina Hernández, Alan W Stitt, Tim M Curtis, Rafael Simó","doi":"10.1186/s12974-024-03283-5","DOIUrl":"https://doi.org/10.1186/s12974-024-03283-5","url":null,"abstract":"<p><p>Impaired function of the retinal neurovascular unit (NVU) is an early event in diabetic retinopathy (DR). It has been previously shown that topical delivery of the dipeptidyl peptidase-4 (DPP-4) inhibitor sitagliptin can protect against diabetes-mediated dysfunction of the retinal NVU in the db/db mouse. The aim of the present study was to examine whether sitagliptin could prevent the DR-like lesions within the NVU of the new non-diabetic model of DR, the Trpv2 knockout rat (Trpv2<sup>+/-</sup>). For that purpose, at 3 months of age, Trpv2<sup>+/-</sup> rats were topically treated twice daily for two weeks with sitagliptin or PBS-vehicle eyedrops. Trpv2<sup>+/+</sup> rats treated with vehicle served as the control group. Body weight and glycemia were monitored. Optical coherence tomography recordings, fundus images and retinal samples were obtained to evaluate sitagliptin effects. The results revealed that sitagliptin eye drops had no effect on body weight or glycemia. Vehicle-treated Trpv2<sup>+/-</sup> rats exhibited retinal thinning and larger diameters of major retinal blood vessels, upregulation of inflammatory factors and oxidative markers, glial activation and formation of acellular capillaries. However, topical administration of sitagliptin significantly prevented all these abnormalities. In conclusion, sitagliptin eye drops exert a protective effect against DR-like lesions in Trpv2<sup>+/-</sup> rats. Our results suggest that sitagliptin eye drops carry significant potential to treat not only early-stages of DR but also other diseases with impairment of the NVU unrelated to diabetes.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"312"},"PeriodicalIF":9.3,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11607821/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769858","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
Synergistic effects of repeated transcranial magnetic stimulation and mesenchymal stem cells transplantation on alleviating neuroinflammation and PANoptosis in cerebral ischemia. 反复经颅磁刺激与间充质干细胞移植对减轻脑缺血神经炎症和泛光性脑缺血的协同作用。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s12974-024-03302-5
Shimei Cheng, Qiying Lu, Qiuli Liu, Yuanchen Ma, Jinshuo Chen, Di Lu, Mudan Huang, Yinong Huang, Erming Zhao, Jing Luo, Haiqing Zheng
<p><strong>Background: </strong>Neuronal death is the primary cause of poor outcomes in cerebral ischemia. The inflammatory infiltration in the early phase of ischemic stroke plays a vital role in triggering neuronal death. Either transplantation of mesenchymal stem cells (MSCs) derived from humans or repetitive transcranial magnetic stimulation (rTMS) have respectively proved to be neuroprotective and anti-inflammatory in cerebral ischemia. However, either treatment above has its limitations. Whether these two therapies have synergistic effects on improving neurological function and the underlying mechanisms remains unclear. This investigation aims to elucidate the synergistic effects and underlying mechanisms of MSCs combined with rTMS treatment on the neurological function recovery post-ischemia.</p><p><strong>Methods: </strong>A Sprague-Dawley rat model of cerebral infarction was induced via transient middle cerebral artery occlusion (tMCAO). The rats were divided into five groups (n = 50): sham, tMCAO, rTMS, MSCs, and MSCs + rTMS groups. Transplantation of human umbilical cord MSCs and rTMS intervention were performed 24 h post-stroke. Neurological function was further assessed via several behavioral tests and the 2,3,5-triphenyltetrazolium chloride (TTC) staining companied with Nissl staining were used to assess neuronal survival. TUNEL staining, western blotting, immunofluorescence, immunohistochemistry, ELISA, and flow cytometry were employed to measure the levels of neuroinflammation and PANoptosis. The molecular mechanisms underlying the special role of rTMS in the combined therapy were distinguished with transcriptome sequencing via PC12 cells in oxygen-glucose deprivation/reoxygenation (OGD/R) conditions.</p><p><strong>Results: </strong>The combined therapy efficiently reduced lesion volume and improved neuronal survival (P < 0.05), subsequently improving functional recovery after ischemic stroke. MSCs + rTMS treatment ameliorated the PANoptosis in neurons (P < 0.05), accompanied by decreased levels of inflammatory factors in the cerebral tissue and serum during the subacute phase of cerebral infarction. To further explore the roles of either therapy on synergistic effect, we found that the transplanted MSCs primarily localized in the spleen and reduced cerebral inflammatory infiltration after ischemia via suppressed splenic inflammation. Meanwhile, rTMS significantly protects neurons from PANoptosis in MSCs-inhibited inflammatory conditions by downregulating REST unveiled by transcriptome sequencing.</p><p><strong>Conclusions: </strong>Our study elucidates an unidentified mechanism by which the combination of MSCs and rTMS could synergistically promote neuronal survival and suppress neuroinflammation during the subacute phase of cerebral infarction, thus improving neurological outcomes. The downregulating REST induced by rTMS may potentially contribute to the neuroprotective effect against PANoptosis in MSCs-inhibited inflammatory co
背景:神经元死亡是脑缺血预后不良的主要原因。缺血性脑卒中早期的炎症浸润在触发神经元死亡中起着至关重要的作用。来源于人的间充质干细胞(MSCs)移植或重复经颅磁刺激(rTMS)分别被证明对脑缺血具有神经保护和抗炎作用。然而,上述两种治疗方法都有其局限性。这两种疗法是否在改善神经功能方面具有协同作用及其潜在机制尚不清楚。本研究旨在阐明MSCs联合rTMS治疗对缺血后神经功能恢复的协同作用及其机制。方法:采用短暂性大脑中动脉闭塞法(tMCAO)建立脑梗死大鼠模型。将大鼠分为5组(n = 50): sham组、tMCAO组、rTMS组、MSCs组、MSCs + rTMS组。脑卒中后24小时进行人脐带间充质干细胞移植和rTMS干预。通过多项行为测试进一步评估神经功能,并使用2,3,5-三苯四唑氯(TTC)染色和尼氏染色评估神经元存活。采用TUNEL染色、western blotting、免疫荧光、免疫组织化学、ELISA和流式细胞术检测神经炎症和PANoptosis水平。在氧-葡萄糖剥夺/再氧化(OGD/R)条件下,通过PC12细胞转录组测序来区分rTMS在联合治疗中特殊作用的分子机制。结论:我们的研究阐明了MSCs与rTMS联合使用可协同促进脑梗死亚急性期神经元存活和抑制神经炎症,从而改善神经预后的一种尚未明确的机制。rTMS诱导的REST下调可能有助于mscs抑制炎症条件下PANoptosis的神经保护作用。这些结果有望为MSCs和rTMS联合治疗的机制提供新的见解,以协同保护脑缺血损伤和脑卒中早期神经元PANoptosis的潜在靶点。
{"title":"Synergistic effects of repeated transcranial magnetic stimulation and mesenchymal stem cells transplantation on alleviating neuroinflammation and PANoptosis in cerebral ischemia.","authors":"Shimei Cheng, Qiying Lu, Qiuli Liu, Yuanchen Ma, Jinshuo Chen, Di Lu, Mudan Huang, Yinong Huang, Erming Zhao, Jing Luo, Haiqing Zheng","doi":"10.1186/s12974-024-03302-5","DOIUrl":"https://doi.org/10.1186/s12974-024-03302-5","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Neuronal death is the primary cause of poor outcomes in cerebral ischemia. The inflammatory infiltration in the early phase of ischemic stroke plays a vital role in triggering neuronal death. Either transplantation of mesenchymal stem cells (MSCs) derived from humans or repetitive transcranial magnetic stimulation (rTMS) have respectively proved to be neuroprotective and anti-inflammatory in cerebral ischemia. However, either treatment above has its limitations. Whether these two therapies have synergistic effects on improving neurological function and the underlying mechanisms remains unclear. This investigation aims to elucidate the synergistic effects and underlying mechanisms of MSCs combined with rTMS treatment on the neurological function recovery post-ischemia.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;A Sprague-Dawley rat model of cerebral infarction was induced via transient middle cerebral artery occlusion (tMCAO). The rats were divided into five groups (n = 50): sham, tMCAO, rTMS, MSCs, and MSCs + rTMS groups. Transplantation of human umbilical cord MSCs and rTMS intervention were performed 24 h post-stroke. Neurological function was further assessed via several behavioral tests and the 2,3,5-triphenyltetrazolium chloride (TTC) staining companied with Nissl staining were used to assess neuronal survival. TUNEL staining, western blotting, immunofluorescence, immunohistochemistry, ELISA, and flow cytometry were employed to measure the levels of neuroinflammation and PANoptosis. The molecular mechanisms underlying the special role of rTMS in the combined therapy were distinguished with transcriptome sequencing via PC12 cells in oxygen-glucose deprivation/reoxygenation (OGD/R) conditions.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;The combined therapy efficiently reduced lesion volume and improved neuronal survival (P &lt; 0.05), subsequently improving functional recovery after ischemic stroke. MSCs + rTMS treatment ameliorated the PANoptosis in neurons (P &lt; 0.05), accompanied by decreased levels of inflammatory factors in the cerebral tissue and serum during the subacute phase of cerebral infarction. To further explore the roles of either therapy on synergistic effect, we found that the transplanted MSCs primarily localized in the spleen and reduced cerebral inflammatory infiltration after ischemia via suppressed splenic inflammation. Meanwhile, rTMS significantly protects neurons from PANoptosis in MSCs-inhibited inflammatory conditions by downregulating REST unveiled by transcriptome sequencing.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;Our study elucidates an unidentified mechanism by which the combination of MSCs and rTMS could synergistically promote neuronal survival and suppress neuroinflammation during the subacute phase of cerebral infarction, thus improving neurological outcomes. The downregulating REST induced by rTMS may potentially contribute to the neuroprotective effect against PANoptosis in MSCs-inhibited inflammatory co","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"311"},"PeriodicalIF":9.3,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11607903/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769869","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
CD22 blockade exacerbates neuroinflammation in Neuromyelitis optica spectrum disorder. CD22阻断加剧视神经脊髓炎谱系障碍的神经炎症。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s12974-024-03305-2
Wenjun Zhang, Yali Han, Huachen Huang, Yue Su, Honglei Ren, Caiyun Qi, Jinyi Li, Huaijin Yang, Jing Xu, Guoqiang Chang, Wenjin Qiu, Qiang Liu, Ting Chang

Background: Neuromyelitis optica spectrum disorder (NMOSD) is an autoantibody-triggered central nervous system (CNS) demyelinating disease that primarily affects the spinal cord, optic nerves and brainstem. Among the first responders to CNS injury, microglia are prominent players that drive NMOSD lesion formation. However, the key molecular switches controlling the detrimental activity of microglia in NMOSD are poorly understood. CD22 governs the activity of innate and adaptive immunity. In this study, we investigated to what extent and by what mechanisms CD22 may modulate microglial activity, neuroinflammation and CNS lesion formation.

Methods: To determine the expression profile of CD22 in NMOSD, we performed single-cell sequencing and flow cytometry analysis of immune cells from human peripheral blood. We investigated the potential effects and mechanisms of CD22 blockade on microglial activity, leukocyte infiltration and CNS demyelination in a mouse model of NMOSD induced by injection of NMOSD patient serum-derived AQP4-IgG and human complement.

Results: Single-cell sequencing and flow cytometry analysis revealed that CD22 was expressed in B cells, neutrophils, monocytes and microglia-derived exosomes in human peripheral blood from NMOSD patients and controls (n = 5 per group). In a mouse model of NMOSD, CD22 was expressed in B cells, neutrophils, monocytes and microglia (n = 8 per group). In NMOSD mice, CD22 blockade significantly increased the number of CNS lesions, astrocyte loss and demyelination, accompanied by increased inflammatory activity and phagocytosis in microglia. Furthermore, the detrimental effects of CD22 blockade were significantly alleviated in NMOSD mice subjected to depletion of microglia or Gr-1+ myeloid cells, suggesting the involvement of microglia and peripheral Gr-1+ myeloid cells. Additionally, CD22 blockade also led to significantly reduced phosphorylation of SYK and GSK3β in NMOSD. Notably, the detrimental effects of CD22 blockade were greatly diminished in NMOSD mice receiving the phosphorylated SYK inhibitor R406.

Conclusions: Our findings revealed a previously unrecognized role of CD22 as a key molecular switch that governs the detrimental effects of microglia and Gr-1+ myeloid cells in NMOSD, which paves the way for the future design of immune therapies for NMOSD.

背景:视神经脊髓炎谱系障碍(NMOSD)是一种自身抗体引发的中枢神经系统(CNS)脱髓鞘疾病,主要影响脊髓、视神经和脑干。在中枢神经系统损伤的第一反应者中,小胶质细胞是驱动NMOSD病变形成的重要参与者。然而,控制NMOSD中小胶质细胞有害活性的关键分子开关尚不清楚。CD22控制先天免疫和适应性免疫的活性。在这项研究中,我们研究了CD22调节小胶质细胞活性、神经炎症和中枢神经系统病变形成的程度和机制。方法:通过对人外周血免疫细胞进行单细胞测序和流式细胞术分析,确定CD22在NMOSD中的表达谱。在注射NMOSD患者血清源性AQP4-IgG和人补体诱导的NMOSD小鼠模型中,我们研究了CD22阻断对小胶质细胞活性、白细胞浸润和中枢神经系统脱髓鞘的潜在影响和机制。结果:单细胞测序和流式细胞术分析显示,CD22在NMOSD患者和对照组的人外周血B细胞、中性粒细胞、单核细胞和小胶质细胞来源的外泌体中表达(每组n = 5)。在小鼠NMOSD模型中,CD22在B细胞、中性粒细胞、单核细胞和小胶质细胞中表达(每组n = 8)。在NMOSD小鼠中,CD22阻断显著增加了中枢神经系统病变数量、星形胶质细胞损失和脱髓鞘,并伴有小胶质细胞炎症活性和吞噬作用的增加。此外,在小胶质细胞或Gr-1+髓样细胞缺失的NMOSD小鼠中,CD22阻断的有害影响显著减轻,提示小胶质细胞和外周Gr-1+髓样细胞参与其中。此外,CD22阻断也导致NMOSD中SYK和GSK3β磷酸化显著降低。值得注意的是,在接受磷酸化SYK抑制剂R406的NMOSD小鼠中,CD22阻断的有害影响大大减轻。结论:我们的研究结果揭示了CD22作为控制小胶质细胞和Gr-1+髓样细胞在NMOSD中的有害作用的关键分子开关的作用,这为未来设计NMOSD的免疫疗法铺平了道路。
{"title":"CD22 blockade exacerbates neuroinflammation in Neuromyelitis optica spectrum disorder.","authors":"Wenjun Zhang, Yali Han, Huachen Huang, Yue Su, Honglei Ren, Caiyun Qi, Jinyi Li, Huaijin Yang, Jing Xu, Guoqiang Chang, Wenjin Qiu, Qiang Liu, Ting Chang","doi":"10.1186/s12974-024-03305-2","DOIUrl":"https://doi.org/10.1186/s12974-024-03305-2","url":null,"abstract":"<p><strong>Background: </strong>Neuromyelitis optica spectrum disorder (NMOSD) is an autoantibody-triggered central nervous system (CNS) demyelinating disease that primarily affects the spinal cord, optic nerves and brainstem. Among the first responders to CNS injury, microglia are prominent players that drive NMOSD lesion formation. However, the key molecular switches controlling the detrimental activity of microglia in NMOSD are poorly understood. CD22 governs the activity of innate and adaptive immunity. In this study, we investigated to what extent and by what mechanisms CD22 may modulate microglial activity, neuroinflammation and CNS lesion formation.</p><p><strong>Methods: </strong>To determine the expression profile of CD22 in NMOSD, we performed single-cell sequencing and flow cytometry analysis of immune cells from human peripheral blood. We investigated the potential effects and mechanisms of CD22 blockade on microglial activity, leukocyte infiltration and CNS demyelination in a mouse model of NMOSD induced by injection of NMOSD patient serum-derived AQP4-IgG and human complement.</p><p><strong>Results: </strong>Single-cell sequencing and flow cytometry analysis revealed that CD22 was expressed in B cells, neutrophils, monocytes and microglia-derived exosomes in human peripheral blood from NMOSD patients and controls (n = 5 per group). In a mouse model of NMOSD, CD22 was expressed in B cells, neutrophils, monocytes and microglia (n = 8 per group). In NMOSD mice, CD22 blockade significantly increased the number of CNS lesions, astrocyte loss and demyelination, accompanied by increased inflammatory activity and phagocytosis in microglia. Furthermore, the detrimental effects of CD22 blockade were significantly alleviated in NMOSD mice subjected to depletion of microglia or Gr-1<sup>+</sup> myeloid cells, suggesting the involvement of microglia and peripheral Gr-1<sup>+</sup> myeloid cells. Additionally, CD22 blockade also led to significantly reduced phosphorylation of SYK and GSK3β in NMOSD. Notably, the detrimental effects of CD22 blockade were greatly diminished in NMOSD mice receiving the phosphorylated SYK inhibitor R406.</p><p><strong>Conclusions: </strong>Our findings revealed a previously unrecognized role of CD22 as a key molecular switch that governs the detrimental effects of microglia and Gr-1<sup>+</sup> myeloid cells in NMOSD, which paves the way for the future design of immune therapies for NMOSD.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"313"},"PeriodicalIF":9.3,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11607829/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769855","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
A transient blood IL-17 increase triggers neuroinflammation in cerebellum and motor incoordination in hyperammonemic rats. 高氨血症大鼠血液中短暂的IL-17升高可引发小脑神经炎症和运动不协调。
IF 9.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-11-30 DOI: 10.1186/s12974-024-03310-5
Yaiza M Arenas, Carmina Montoliu, Marta Llansola, Vicente Felipo

Patients with liver cirrhosis may show minimal hepatic encephalopathy (MHE) with motor incoordination which is reproduced in hyperammonemic rats. Hyperammonemia induces peripheral inflammation which triggers neuroinflammation and enhanced GABAergic neurotransmission in cerebellum and motor incoordination. The mechanisms involved remain unknown. The aims were to assess if the early increase of peripheral IL-17 triggers motor incoordination in hyperammonemic rats and to identify some underlying mechanisms. We assessed if blocking peripheral IL-17 with anti-IL-17 at 2-4 days of hyperammonemia prevents motor incoordination and analyzed underlying mechanisms. Hyperammonemia induces a transient blood IL-17 increase at days 3-4. This is associated with increased IL-17 receptor membrane expression and activation in cerebellum, leading to NADPH oxidase activation, increased superoxide production and MLCK that induce blood-brain barrier (BBB) permeabilization by reducing occludin and ZO-1. BBB permeabilization facilitates the entry of IL-17, which increases in cerebellum and activates microglia. This increases TNFα and the TNFR1-S1PR2-CCL2-BDNF-TrkB pathway. This enhances GABAergic neurotransmission which impairs motor coordination. Blocking peripheral IL-17 with anti-IL-17 prevents all the above process and prevents motor incoordination. Early treatment to reduce blood IL-17 may be a useful treatment to reverse motor incoordination in patients with MHE.

肝硬化患者可表现为轻度肝性脑病(MHE)伴运动不协调,在高氨血症大鼠中重现。高氨血症引起外周炎症,引发神经炎症,增强小脑gaba能神经传递和运动不协调。所涉及的机制尚不清楚。目的是评估外周血IL-17的早期升高是否会引发高氨血症大鼠的运动不协调,并确定一些潜在的机制。我们评估了在2-4天高氨血症时用抗IL-17阻断外周IL-17是否能预防运动不协调,并分析了潜在的机制。高氨血症诱导血清IL-17在第3-4天短暂升高。这与小脑IL-17受体膜表达和激活增加有关,导致NADPH氧化酶激活,超氧化物产生增加,MLCK通过降低occludin和ZO-1诱导血脑屏障(BBB)通透性。血脑屏障的通透性促进了IL-17的进入,IL-17在小脑中增加并激活小胶质细胞。这增加了TNFα和TNFR1-S1PR2-CCL2-BDNF-TrkB通路。这会增强gaba能神经传递,从而损害运动协调。用抗IL-17阻断外周IL-17可阻止上述所有过程并防止运动不协调。早期治疗降低血液IL-17可能是逆转MHE患者运动不协调的有效治疗方法。
{"title":"A transient blood IL-17 increase triggers neuroinflammation in cerebellum and motor incoordination in hyperammonemic rats.","authors":"Yaiza M Arenas, Carmina Montoliu, Marta Llansola, Vicente Felipo","doi":"10.1186/s12974-024-03310-5","DOIUrl":"https://doi.org/10.1186/s12974-024-03310-5","url":null,"abstract":"<p><p>Patients with liver cirrhosis may show minimal hepatic encephalopathy (MHE) with motor incoordination which is reproduced in hyperammonemic rats. Hyperammonemia induces peripheral inflammation which triggers neuroinflammation and enhanced GABAergic neurotransmission in cerebellum and motor incoordination. The mechanisms involved remain unknown. The aims were to assess if the early increase of peripheral IL-17 triggers motor incoordination in hyperammonemic rats and to identify some underlying mechanisms. We assessed if blocking peripheral IL-17 with anti-IL-17 at 2-4 days of hyperammonemia prevents motor incoordination and analyzed underlying mechanisms. Hyperammonemia induces a transient blood IL-17 increase at days 3-4. This is associated with increased IL-17 receptor membrane expression and activation in cerebellum, leading to NADPH oxidase activation, increased superoxide production and MLCK that induce blood-brain barrier (BBB) permeabilization by reducing occludin and ZO-1. BBB permeabilization facilitates the entry of IL-17, which increases in cerebellum and activates microglia. This increases TNFα and the TNFR1-S1PR2-CCL2-BDNF-TrkB pathway. This enhances GABAergic neurotransmission which impairs motor coordination. Blocking peripheral IL-17 with anti-IL-17 prevents all the above process and prevents motor incoordination. Early treatment to reduce blood IL-17 may be a useful treatment to reverse motor incoordination in patients with MHE.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":"21 1","pages":"314"},"PeriodicalIF":9.3,"publicationDate":"2024-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11608494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142769853","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