Journal of Neuroinflammation最新文献

筛选
英文 中文
CD44 signaling in Müller cells impacts photoreceptor function and survival in healthy and diseased retinas. Müller 细胞中的 CD44 信号影响健康视网膜和患病视网膜中感光器的功能和存活。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-02 DOI: 10.1186/s12974-024-03175-8
Monika Ayten, Tobias Straub, Lew Kaplan, Stefanie M Hauck, Antje Grosche, Susanne F Koch
{"title":"CD44 signaling in Müller cells impacts photoreceptor function and survival in healthy and diseased retinas.","authors":"Monika Ayten, Tobias Straub, Lew Kaplan, Stefanie M Hauck, Antje Grosche, Susanne F Koch","doi":"10.1186/s12974-024-03175-8","DOIUrl":"10.1186/s12974-024-03175-8","url":null,"abstract":"<p><p>Retinitis pigmentosa (RP), an inherited retinal disease, affects 1,5 million people worldwide. The initial mutation-driven photoreceptor degeneration leads to chronic inflammation, characterized by Müller cell activation and upregulation of CD44. CD44 is a cell surface transmembrane glycoprotein and the primary receptor for hyaluronic acid. It is involved in many pathological processes, but little is known about CD44's retinal functions. CD44 expression is also increased in Müller cells from our Pde6b<sup>STOP/STOP</sup> RP mouse model. To gain a more detailed understanding of CD44's role in healthy and diseased retinas, we analyzed Cd44<sup>-/-</sup> and Cd44<sup>-/-</sup>Pde6b<sup>STOP/STOP</sup> mice, respectively. The loss of CD44 led to enhanced photoreceptor degeneration, reduced retinal function, and increased inflammatory response. To understand the underlying mechanism, we performed proteomic analysis on isolated Müller cells from Cd44<sup>-/-</sup> and Cd44<sup>-/-</sup>Pde6b<sup>STOP/STOP</sup> retinas and identified a significant downregulation of glutamate transporter 1 (SLC1A2). This downregulation was accompanied by higher glutamate levels, suggesting impaired glutamate homeostasis. These novel findings indicate that CD44 stimulates glutamate uptake via SLC1A2 in Müller cells, which in turn, supports photoreceptor survival and function.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297696/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878880","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
Toxoplasma infection induces an aged neutrophil population in the CNS that is associated with neuronal protection. 弓形虫感染会诱导中枢神经系统中的中性粒细胞老化,这与神经元保护有关。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-02 DOI: 10.1186/s12974-024-03176-7
Kristina V Bergersen, Bill Kavvathas, Byron D Ford, Emma H Wilson
{"title":"Toxoplasma infection induces an aged neutrophil population in the CNS that is associated with neuronal protection.","authors":"Kristina V Bergersen, Bill Kavvathas, Byron D Ford, Emma H Wilson","doi":"10.1186/s12974-024-03176-7","DOIUrl":"10.1186/s12974-024-03176-7","url":null,"abstract":"<p><strong>Background: </strong>Infection with the protozoan parasite Toxoplasma gondii leads to the formation of lifelong cysts in neurons that can have devastating consequences in the immunocompromised. In the immunocompetent individual, anti-parasitic effector mechanisms and a balanced immune response characterized by pro- and anti-inflammatory cytokine production establishes an asymptomatic infection that rarely leads to neurological symptoms. Several mechanisms are known to play a role in this successful immune response in the brain including T cell production of IFNγ and IL-10 and the involvement of CNS resident cells. This limitation of clinical neuropathology during chronic infection suggests a balance between immune response and neuroprotective mechanisms that collectively prevent clinical manifestations of disease. However, how these two vital mechanisms of protection interact during chronic Toxoplasma infection remains poorly understood.</p><p><strong>Main text: </strong>This study demonstrates a previously undescribed connection between innate neutrophils found chronically in the brain, termed \"chronic brain neutrophils\" (CBNeuts), and neuroprotective mechanisms during Toxoplasma infection. Lack of CBNeuts during chronic infection, accomplished via systemic neutrophil depletion, led to enhanced infection and deleterious effects on neuronal regeneration and repair mechanisms in the brain. Phenotypic and transcriptomic analysis of CBNeuts identified them as distinct from peripheral neutrophils and revealed two main subsets of CBNeuts that display heterogeneity towards both classical effector and neuroprotective functions in an age-dependent manner. Further phenotypic profiling defined expression of the neuroprotective molecules NRG-1 andErbB4 by these cells, and the importance of this signaling pathway during chronic infection was demonstrated via NRG-1 treatment studies.</p><p><strong>Conclusions: </strong>In conclusion, this work identifies CBNeuts as a heterogenous population geared towards both classical immune responses and neuroprotection during chronic Toxoplasma infection and provides the foundation for future mechanistic studies of these cells.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297776/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878904","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
Targeting the immunoproteasome in hypothalamic neurons as a novel therapeutic strategy for high-fat diet-induced obesity and metabolic dysregulation. 以下丘脑神经元中的免疫蛋白酶体为靶点,作为高脂饮食诱发肥胖和代谢失调的新型治疗策略。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-02 DOI: 10.1186/s12974-024-03154-z
Nicolás Albornoz, Javiera Álvarez-Indo, Adely de la Peña, Eloisa Arias-Muñoz, Alanis Coca, Fabián Segovia-Miranda, Bredford Kerr, Mauricio Budini, Alfredo Criollo, María A García-Robles, Eugenia Morselli, Andrea Soza, Patricia V Burgos
{"title":"Targeting the immunoproteasome in hypothalamic neurons as a novel therapeutic strategy for high-fat diet-induced obesity and metabolic dysregulation.","authors":"Nicolás Albornoz, Javiera Álvarez-Indo, Adely de la Peña, Eloisa Arias-Muñoz, Alanis Coca, Fabián Segovia-Miranda, Bredford Kerr, Mauricio Budini, Alfredo Criollo, María A García-Robles, Eugenia Morselli, Andrea Soza, Patricia V Burgos","doi":"10.1186/s12974-024-03154-z","DOIUrl":"10.1186/s12974-024-03154-z","url":null,"abstract":"<p><strong>Objective: </strong>Obesity represents a significant global health challenge characterized by chronic low-grade inflammation and metabolic dysregulation. The hypothalamus, a key regulator of energy homeostasis, is particularly susceptible to obesity's deleterious effects. This study investigated the role of the immunoproteasome, a specialized proteasomal complex implicated in inflammation and cellular homeostasis, during metabolic diseases.</p><p><strong>Methods: </strong>The levels of the immunoproteasome β5i subunit were analyzed by immunostaining, western blotting, and proteasome activity assay in mice fed with either a high-fat diet (HFD) or a regular diet (CHOW). We also characterized the impact of autophagy inhibition on the levels of the immunoproteasome β5i subunit and the activation of the AKT pathway. Finally, through confocal microscopy, we analyzed the contribution of β5i subunit inhibition on mitochondrial function by flow cytometry and mitophagy assay.</p><p><strong>Results: </strong>Using an HFD-fed obese mouse model, we found increased immunoproteasome levels in hypothalamic POMC neurons. Furthermore, we observed that palmitic acid (PA), a major component of saturated fats found in HFD, increased the levels of the β5i subunit of the immunoproteasome in hypothalamic neuronal cells. Notably, the increase in immunoproteasome expression was associated with decreased autophagy, a critical cellular process in maintaining homeostasis and suppressing inflammation. Functionally, PA disrupted the insulin-glucose axis, leading to reduced AKT phosphorylation and increased intracellular glucose levels in response to insulin due to the upregulation of the immunoproteasome. Mechanistically, we identified that the protein PTEN, a key regulator of insulin signaling, was reduced in an immunoproteasome-dependent manner. To further investigate the potential therapeutic implications of these findings, we used ONX-0914, a specific immunoproteasome inhibitor. We demonstrated that this inhibitor prevents PA-induced insulin-glucose axis imbalance. Given the interplay between mitochondrial dysfunction and metabolic disturbances, we explored the impact of ONX-0914 on mitochondrial function. Notably, ONX-0914 preserved mitochondrial membrane potential and attenuated mitochondrial ROS production in the presence of PA. Moreover, we found that ONX-0914 reduced mitophagy in the presence of PA.</p><p><strong>Conclusions: </strong>Our findings strongly support the pathogenic involvement of the immunoproteasome in hypothalamic neurons in the context of HFD-induced obesity and metabolic disturbances. Targeting the immunoproteasome highlights a promising therapeutic strategy to mitigate the detrimental effects of obesity on the insulin-glucose axis and cellular homeostasis. This study provides valuable insights into the mechanisms driving obesity-related metabolic diseases and offers potential avenues for developing novel therapeutic interventions.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297766/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878903","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
Inhibition of ANGPTL8 protects against diabetes-associated cognitive dysfunction by reducing synaptic loss via the PirB signaling pathway. 抑制 ANGPTL8 可通过 PirB 信号通路减少突触损失,从而预防与糖尿病相关的认知功能障碍。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-02 DOI: 10.1186/s12974-024-03183-8
Xiaoyu Meng, Danpei Li, Ranran Kan, Yuxi Xiang, Limeng Pan, Yaming Guo, Peng Yu, Peiqiong Luo, Huajie Zou, Li Huang, Yurong Zhu, Beibei Mao, Yi He, Lei Xie, Jialu Xu, Xiaoyan Liu, Wenjun Li, Yong Chen, Suiqiang Zhu, Yan Yang, Xuefeng Yu
{"title":"Inhibition of ANGPTL8 protects against diabetes-associated cognitive dysfunction by reducing synaptic loss via the PirB signaling pathway.","authors":"Xiaoyu Meng, Danpei Li, Ranran Kan, Yuxi Xiang, Limeng Pan, Yaming Guo, Peng Yu, Peiqiong Luo, Huajie Zou, Li Huang, Yurong Zhu, Beibei Mao, Yi He, Lei Xie, Jialu Xu, Xiaoyan Liu, Wenjun Li, Yong Chen, Suiqiang Zhu, Yan Yang, Xuefeng Yu","doi":"10.1186/s12974-024-03183-8","DOIUrl":"10.1186/s12974-024-03183-8","url":null,"abstract":"<p><strong>Background: </strong>Type 2 diabetes mellitus (T2D) is associated with an increased risk of cognitive dysfunction. Angiopoietin-like protein 8 (ANGPTL8) is an important regulator in T2D, but the role of ANGPTL8 in diabetes-associated cognitive dysfunction remains unknown. Here, we explored the role of ANGPTL8 in diabetes-associated cognitive dysfunction through its interaction with paired immunoglobulin-like receptor B (PirB) in the central nervous system.</p><p><strong>Methods: </strong>The levels of ANGPTL8 in type 2 diabetic patients with cognitive dysfunction and control individuals were measured. Mouse models of diabetes-associated cognitive dysfunction were constructed to investigate the role of ANGPTL8 in cognitive function. The cognitive function of the mice was assessed by the Barnes Maze test and the novel object recognition test, and levels of ANGPTL8, synaptic and axonal markers, and pro-inflammatory cytokines were measured. Primary neurons and microglia were treated with recombinant ANGPTL8 protein (rA8), and subsequent changes were examined. In addition, the changes induced by ANGPTL8 were validated after blocking PirB and its downstream pathways. Finally, mice with central nervous system-specific knockout of Angptl8 and PirB<sup>-/-</sup> mice were generated, and relevant in vivo experiments were performed.</p><p><strong>Results: </strong>Here, we demonstrated that in the diabetic brain, ANGPTL8 was secreted by neurons into the hippocampus, resulting in neuroinflammation and impairment of synaptic plasticity. Moreover, neuron-specific Angptl8 knockout prevented diabetes-associated cognitive dysfunction and neuroinflammation. Mechanistically, ANGPTL8 acted in parallel to neurons and microglia via its receptor PirB, manifesting as downregulation of synaptic and axonal markers in neurons and upregulation of proinflammatory cytokine expression in microglia. In vivo, PirB<sup>-/-</sup> mice exhibited resistance to ANGPTL8-induced neuroinflammation and synaptic damage.</p><p><strong>Conclusion: </strong>Taken together, our findings reveal the role of ANGPTL8 in the pathogenesis of diabetes-associated cognitive dysfunction and identify the ANGPTL8-PirB signaling pathway as a potential target for the management of this condition.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297729/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878901","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
Neuroimmune modulation in liver pathophysiology. 肝脏病理生理学中的神经免疫调节。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-08-01 DOI: 10.1186/s12974-024-03181-w
Ju Zou, Jie Li, Xiaoxu Wang, Daolin Tang, Ruochan Chen
{"title":"Neuroimmune modulation in liver pathophysiology.","authors":"Ju Zou, Jie Li, Xiaoxu Wang, Daolin Tang, Ruochan Chen","doi":"10.1186/s12974-024-03181-w","DOIUrl":"10.1186/s12974-024-03181-w","url":null,"abstract":"<p><p>The liver, the largest organ in the human body, plays a multifaceted role in digestion, coagulation, synthesis, metabolism, detoxification, and immune defense. Changes in liver function often coincide with disruptions in both the central and peripheral nervous systems. The intricate interplay between the nervous and immune systems is vital for maintaining tissue balance and combating diseases. Signaling molecules and pathways, including cytokines, inflammatory mediators, neuropeptides, neurotransmitters, chemoreceptors, and neural pathways, facilitate this complex communication. They establish feedback loops among diverse immune cell populations and the central, peripheral, sympathetic, parasympathetic, and enteric nervous systems within the liver. In this concise review, we provide an overview of the structural and compositional aspects of the hepatic neural and immune systems. We further explore the molecular mechanisms and pathways that govern neuroimmune communication, highlighting their significance in liver pathology. Finally, we summarize the current clinical implications of therapeutic approaches targeting neuroimmune interactions and present prospects for future research in this area.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11295927/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141875118","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
Tracking neuroinflammatory biomarkers in Alzheimer's disease: a strategy for individualized therapeutic approaches? 追踪阿尔茨海默病的神经炎症生物标志物:个体化治疗方法的一种策略?
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-07-30 DOI: 10.1186/s12974-024-03163-y
Simone Lista, Bruno P Imbimbo, Margherita Grasso, Annamaria Fidilio, Enzo Emanuele, Piercarlo Minoretti, Susana López-Ortiz, Juan Martín-Hernández, Audrey Gabelle, Giuseppe Caruso, Marco Malaguti, Daniela Melchiorri, Alejandro Santos-Lozano, Camillo Imbimbo, Michael T Heneka, Filippo Caraci
{"title":"Tracking neuroinflammatory biomarkers in Alzheimer's disease: a strategy for individualized therapeutic approaches?","authors":"Simone Lista, Bruno P Imbimbo, Margherita Grasso, Annamaria Fidilio, Enzo Emanuele, Piercarlo Minoretti, Susana López-Ortiz, Juan Martín-Hernández, Audrey Gabelle, Giuseppe Caruso, Marco Malaguti, Daniela Melchiorri, Alejandro Santos-Lozano, Camillo Imbimbo, Michael T Heneka, Filippo Caraci","doi":"10.1186/s12974-024-03163-y","DOIUrl":"10.1186/s12974-024-03163-y","url":null,"abstract":"<p><strong>Background: </strong>Recent trials of anti-amyloid-β (Aβ) monoclonal antibodies, including lecanemab and donanemab, in early Alzheimer disease (AD) showed that these drugs have limited clinical benefits and their use comes with a significant risk of serious adverse events. Thus, it seems crucial to explore complementary therapeutic approaches. Genome-wide association studies identified robust associations between AD and several AD risk genes related to immune response, including but not restricted to CD33 and TREM2. Here, we critically reviewed the current knowledge on candidate neuroinflammatory biomarkers and their role in characterizing the pathophysiology of AD.</p><p><strong>Main body: </strong>Neuroinflammation is recognized to be a crucial and contributing component of AD pathogenesis. The fact that neuroinflammation is most likely present from earliest pre-stages of AD and co-occurs with the deposition of Aβ reinforces the need to precisely define the sequence and nature of neuroinflammatory events. Numerous clinical trials involving anti-inflammatory drugs previously yielded unfavorable outcomes in early and mild-to-moderate AD. Although the reasons behind these failures remain unclear, these may include the time and the target selected for intervention. Indeed, in our review, we observed a stage-dependent neuroinflammatory process in the AD brain. While the initial activation of glial cells counteracts early brain Aβ deposition, the downregulation in the functional state of microglia occurs at more advanced disease stages. To address this issue, personalized neuroinflammatory modulation therapy is required. The emergence of reliable blood-based neuroinflammatory biomarkers, particularly glial fibrillary acidic protein, a marker of reactive astrocytes, may facilitate the classification of AD patients based on the ATI(N) biomarker framework. This expands upon the traditional classification of Aβ (\"A\"), tau (\"T\"), and neurodegeneration (\"N\"), by incorporating a novel inflammatory component (\"I\").</p><p><strong>Conclusions: </strong>The present review outlines the current knowledge on potential neuroinflammatory biomarkers and, importantly, emphasizes the role of longitudinal analyses, which are needed to accurately monitor the dynamics of cerebral inflammation. Such a precise information on time and place will be required before anti-inflammatory therapeutic interventions can be considered for clinical evaluation. We propose that an effective anti-neuroinflammatory therapy should specifically target microglia and astrocytes, while considering the individual ATI(N) status of patients.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11289964/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141855786","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
Lactylation of histone by BRD4 regulates astrocyte polarization after experimental subarachnoid hemorrhage. BRD4对组蛋白的乳化作用可调节实验性蛛网膜下腔出血后星形胶质细胞的极化。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-07-30 DOI: 10.1186/s12974-024-03185-6
Fan Zhang, Jian Zhou, Peng Lu, Xianhui Zhang, Lei Yang, Jinpeng Wu, Lihan Zhang, Lifang Zhang, Jinwei Pang, Huangfan Xie, Bingqing Xie, Yong Jiang, Jianhua Peng
{"title":"Lactylation of histone by BRD4 regulates astrocyte polarization after experimental subarachnoid hemorrhage.","authors":"Fan Zhang, Jian Zhou, Peng Lu, Xianhui Zhang, Lei Yang, Jinpeng Wu, Lihan Zhang, Lifang Zhang, Jinwei Pang, Huangfan Xie, Bingqing Xie, Yong Jiang, Jianhua Peng","doi":"10.1186/s12974-024-03185-6","DOIUrl":"10.1186/s12974-024-03185-6","url":null,"abstract":"<p><p>Under subarachnoid hemorrhage (SAH) conditions, astrocytes undergo a marked intensification of glycolytic activity, resulting in the generation of substantial amounts of lactate to maintain the energy demand for neurons and other brain cells. Lactate has garnered increasing attention in recent years because of its emerging role in critical biological processes such as inflammation regulation and neuroprotection, particularly through its histone lactylation. Bromodomain-containing protein 4 (BRD4) plays a crucial role in maintaining neural development and promoting memory formation in the central nervous system. Nonetheless, the function and regulatory mechanism of BRD4 and histone lactylation in astrocytes following SAH remain elusive. Our findings indicate that BRD4, a crucial epigenetic regulator, plays a definitive role in histone lactylation. Both in vitro and in vivo, these results demonstrated that targeted silencing of BRD4 in astrocytes can significantly reduce H4K8la lactylation, thereby aggravating the A1 polarization of astrocytes and ultimately affecting the recovery of neural function and prognosis in mice after SAH. In summary, BRD4 plays a pivotal role in modulating astrocyte polarization following SAH via histone lactylation. Targeting this mechanism might offer an efficient therapeutic strategy for SAH.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11290164/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141855785","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
Differential effect of an evolving amyloid and tau pathology on brain phospholipids and bioactive lipid mediators in rat models of Alzheimer-like pathology. 淀粉样蛋白和 tau 病理学的演变对阿尔茨海默病大鼠模型中脑磷脂和生物活性脂质介质的不同影响。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-07-30 DOI: 10.1186/s12974-024-03184-7
Sonia Do Carmo, Marie-Audrey I Kautzmann, Surjyadipta Bhattacharjee, Bokkyoo Jun, Carolyn Steinberg, Joshua T Emmerson, Janice C Malcolm, Quentin Bonomo, Nicolas G Bazan, A Claudio Cuello
{"title":"Differential effect of an evolving amyloid and tau pathology on brain phospholipids and bioactive lipid mediators in rat models of Alzheimer-like pathology.","authors":"Sonia Do Carmo, Marie-Audrey I Kautzmann, Surjyadipta Bhattacharjee, Bokkyoo Jun, Carolyn Steinberg, Joshua T Emmerson, Janice C Malcolm, Quentin Bonomo, Nicolas G Bazan, A Claudio Cuello","doi":"10.1186/s12974-024-03184-7","DOIUrl":"10.1186/s12974-024-03184-7","url":null,"abstract":"<p><strong>Background: </strong>Brain inflammation contributes significantly to the pathophysiology of Alzheimer's disease, and it is manifested by glial cell activation, increased production of cytokines/chemokines, and a shift in lipid mediators from a pro-homeostatic to a pro-inflammatory profile. However, whether the production of bioactive lipid mediators is affected at earlier stages, prior to the deposition of Aβ plaques and tau hyperphosphorylation, is unknown. The differential contribution of an evolving amyloid and tau pathology on the composition and abundance of membrane phospholipids and bioactive lipid mediators also remains unresolved.</p><p><strong>Methods: </strong>In this study, we examined the cortical levels of DHA- and AA-derived bioactive lipid mediators and of membrane phospholipids by liquid chromatography with tandem mass spectrometry in transgenic rat models of the Alzheimer's-like amyloid and tau pathologies at early and advanced pathological stages.</p><p><strong>Results: </strong>Our findings revealed a complex balance between pro-inflammatory and pro-resolving processes in which tau pathology has a more pronounced effect compared to amyloid pathology. At stages preceding tau misfolding and aggregation, there was an increase in pro-resolving lipid mediators (RVD6 and NPD1), DHA-containing phospholipids and IFN-γ levels. However, in advanced tau pathology displaying NFT-like inclusions, neuronal death, glial activation and cognitive deficits, there was an increase in cytokine and PGD2, PGE2, and PGF2α generation accompanied by a drop in IFN-γ levels. This pathology also resulted in a marked increase in AA-containing phospholipids. In comparison, pre-plaque amyloid pathology already presented high levels of cytokines and AA-containing phospholipids together with elevated RVD6 and NPD1 levels. Finally, Aβ plaque deposition was accompanied by a modest increase in prostaglandins, increased AA-containing phospholipids and reduced DHA-containing phospholipids.</p><p><strong>Conclusions: </strong>Our findings suggest a dynamic trajectory of inflammatory and lipid mediators in the evolving amyloid and tau pathologies and support their differing roles on membrane properties and, consequentially, on signal transduction.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11290283/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141855784","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
New insights into metabolism dysregulation after TBI. 创伤后新陈代谢失调的新见解。
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-07-29 DOI: 10.1186/s12974-024-03177-6
Helena C Oft, Dennis W Simon, Dandan Sun
{"title":"New insights into metabolism dysregulation after TBI.","authors":"Helena C Oft, Dennis W Simon, Dandan Sun","doi":"10.1186/s12974-024-03177-6","DOIUrl":"10.1186/s12974-024-03177-6","url":null,"abstract":"<p><p>Traumatic brain injury (TBI) remains a leading cause of death and disability that places a great physical, social, and financial burden on individuals and the health system. In this review, we summarize new research into the metabolic changes described in clinical TBI trials, some of which have already shown promise for informing injury classification and staging. We focus our discussion on derangements in glucose metabolism, cell respiration/mitochondrial function and changes to ketone and lipid metabolism/oxidation to emphasize potentially novel biomarkers for clinical outcome prediction and intervention and offer new insights into possible underlying mechanisms from preclinical research of TBI pathology. Finally, we discuss nutrition supplementation studies that aim to harness the gut/microbiome-brain connection and manipulate systemic/cellular metabolism to improve post-TBI recovery. Taken together, this narrative review summarizes published TBI-associated changes in glucose and lipid metabolism, highlighting potential metabolite biomarkers for clinical use, the cellular processes linking these markers to TBI pathology as well as the limitations and future considerations for TBI \"omics\" work.</p>","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11288120/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141792662","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
The potential of gene delivery for the treatment of traumatic brain injury 基因递送治疗脑外伤的潜力
IF 9.3 1区 医学
Journal of Neuroinflammation Pub Date : 2024-07-28 DOI: 10.1186/s12974-024-03156-x
James Dooley, Jasmine G. Hughes, Edward J. Needham, Katerina A. Palios, Adrian Liston
{"title":"The potential of gene delivery for the treatment of traumatic brain injury","authors":"James Dooley, Jasmine G. Hughes, Edward J. Needham, Katerina A. Palios, Adrian Liston","doi":"10.1186/s12974-024-03156-x","DOIUrl":"https://doi.org/10.1186/s12974-024-03156-x","url":null,"abstract":"Therapeutics for traumatic brains injuries constitute a global unmet medical need. Despite the advances in neurocritical care, which have dramatically improved the survival rate for the ~ 70 million patients annually, few treatments have been developed to counter the long-term neuroinflammatory processes and accompanying cognitive impairments, frequent among patients. This review looks at gene delivery as a potential therapeutic development avenue for traumatic brain injury. We discuss the capacity of gene delivery to function in traumatic brain injury, by producing beneficial biologics within the brain. Gene delivery modalities, promising vectors and key delivery routes are discussed, along with the pathways that biological cargos could target to improve long-term outcomes for patients. Coupling blood-brain barrier crossing with sustained local production, gene delivery has the potential to convert proteins with useful biological properties, but poor pharmacodynamics, into effective therapeutics. Finally, we review the limitations and health economics of traumatic brain injury, and whether future gene delivery approaches will be viable for patients and health care systems.","PeriodicalId":16577,"journal":{"name":"Journal of Neuroinflammation","volume":null,"pages":null},"PeriodicalIF":9.3,"publicationDate":"2024-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141783843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","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