Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology最新文献

筛选
英文 中文
Role of Dysregulated Autophagy in HIV Tat, Cocaine, and cART Mediated NLRP3 Activation in Microglia. 失调的自噬在小胶质细胞中HIV Tat、可卡因和cART介导的NLRP3激活中的作用。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-05-06 DOI: 10.1007/s11481-023-10063-0
Seema Singh, Annadurai Thangaraj, Ernest T Chivero, Ming-Lei Guo, Palsamy Periyasamy, Shilpa Buch
{"title":"Role of Dysregulated Autophagy in HIV Tat, Cocaine, and cART Mediated NLRP3 Activation in Microglia.","authors":"Seema Singh, Annadurai Thangaraj, Ernest T Chivero, Ming-Lei Guo, Palsamy Periyasamy, Shilpa Buch","doi":"10.1007/s11481-023-10063-0","DOIUrl":"10.1007/s11481-023-10063-0","url":null,"abstract":"<p><p>Despite the ability of combination antiretroviral therapy (cART) to suppress viremia, there is persistence low levels of HIV proteins such as Transactivator of transcription (Tat) in the central nervous system (CNS), contributing to glial activation and neuroinflammation. Accumulating evidence also implicates the role of drugs of abuse in exacerbating neurological complications associated with HIV-1. The combined effects of HIV Tat, drugs of abuse, and cART can thus create a toxic milieu in the CNS. The present study investigated the combinatorial effects of HIV-Tat, cocaine, and cART on autophagy and NLRP3 inflammasome activation. We selected a combination of three commonly used cART regimens: tenofovir, emtricitabine, and dolutegravir. Our results demonstrated that exposure of mouse primary microglia (MPMs) to these agents-HIV Tat (25 ng/ml), cocaine (1 μM), and cART (1 μM each) resulted in upregulation of autophagy markers: Beclin1, LC3B-II, and SQSTM1 with impaired lysosomal functioning involving increased lysosomal pH, decreased LAMP2 and cathepsin D, ultimately leading to dysregulated autophagy. Our findings also demonstrated activation of the NLRP3 signaling in microglia exposed to these agents. We further demonstrated that gene silencing of key autophagy protein BECN1 significantly blocked NLRP3-mediated activation of microglia. Silencing of NLRP3, however, failed to block HIV Tat, cocaine, and cART-mediated dysregulation of the autophagy-lysosomal axis; these in vitro phenomena were also validated in vivo using iTat mice administered cocaine and cART. This study thus underscores the cooperative effects of HIV Tat, cocaine, and cART in exacerbating microglial activation involving dysregulated autophagy and activation of the NLRP3 inflammasome signaling.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"327-347"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10729649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9408253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effect and Mechanism of Sodium Butyrate on Neuronal Recovery and Prognosis in Diabetic Stroke. 丁酸钠对糖尿病脑卒中神经元恢复和预后的影响及其机制。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-06-15 DOI: 10.1007/s11481-023-10071-0
Ting-Ting Li, Deng-Ming Zhao, Yu-Ting Wei, Jing-Bo Li, Xue-Fei Li, Qiang Wan, Xin Zhang, Xiang-Nan Liu, Wan-Chao Yang, Wen-Zhi Li
{"title":"Effect and Mechanism of Sodium Butyrate on Neuronal Recovery and Prognosis in Diabetic Stroke.","authors":"Ting-Ting Li,&nbsp;Deng-Ming Zhao,&nbsp;Yu-Ting Wei,&nbsp;Jing-Bo Li,&nbsp;Xue-Fei Li,&nbsp;Qiang Wan,&nbsp;Xin Zhang,&nbsp;Xiang-Nan Liu,&nbsp;Wan-Chao Yang,&nbsp;Wen-Zhi Li","doi":"10.1007/s11481-023-10071-0","DOIUrl":"10.1007/s11481-023-10071-0","url":null,"abstract":"<p><p>Ischemic stroke is a cerebrovascular lesion caused by local ischemia and hypoxia. Diabetes mellitus (DM) is a chronic inflammatory disease that disturbs immune homeostasis and predisposes patients to ischemic stroke. The mechanism by which DM exacerbates stroke remains unclear, although it may involve disturbances in immune homeostasis. Regulatory T cells (Tregs) play a regulatory role in many diseases, but the mechanism of Tregs in diabetes complicated by stroke remains unclear. Sodium butyrate is a short-chain fatty acid that increases Treg levels. This study examined the role of sodium butyrate in the prognosis of neurological function in diabetic stroke and the mechanism by which Tregs are amplified in the bilateral cerebral hemispheres. We evaluated the brain infarct volume, observed 48-h neuronal injury and 28-day behavioral changes, and calculated the 28-day survival rate in mice. We also measured Treg levels in peripheral blood and brain tissue, recorded changes in the blood‒brain barrier and water channel proteins and neurotrophic changes in mice, measured cytokine levels and peripheral B-cell distribution in bilateral hemispheres and peripheral blood, and examined the polarization of microglia and the distribution of peripheral T-cell subpopulations in bilateral hemispheres. Diabetes significantly exacerbated the poor prognosis and neurological deficits in mice with stroke, and sodium butyrate significantly improved infarct volume, prognosis, and neurological function and showed different mechanisms in brain tissue and peripheral blood. The potential regulatory mechanism in brain tissue involved modulating Tregs/TGF-β/microglia to suppress neuroinflammation, while that in peripheral blood involved improving the systemic inflammatory response through Tregs/TGF-β/T cells.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"366-382"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9624588","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pharmacological Inhibition of PTEN Rescues Dopaminergic Neurons by Attenuating Apoptotic and Neuroinflammatory Signaling Events. PTEN的药理学抑制通过减轻凋亡和神经炎症信号事件来拯救多巴胺能神经元。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-08-17 DOI: 10.1007/s11481-023-10077-8
Aishwarya Mary Johnson, Sara Jose, Abdul Rasheed Palakkott, Farheen Badrealam Khan, Nanthini Jayabalan, Jaleel Kizhakkayil, Shamma Abdulla Ali AlNaqbi, Mark Gh Scott, Mohammed Akli Ayoub, Richard Gordon, Hariharan Saminathan
{"title":"Pharmacological Inhibition of PTEN Rescues Dopaminergic Neurons by Attenuating Apoptotic and Neuroinflammatory Signaling Events.","authors":"Aishwarya Mary Johnson,&nbsp;Sara Jose,&nbsp;Abdul Rasheed Palakkott,&nbsp;Farheen Badrealam Khan,&nbsp;Nanthini Jayabalan,&nbsp;Jaleel Kizhakkayil,&nbsp;Shamma Abdulla Ali AlNaqbi,&nbsp;Mark Gh Scott,&nbsp;Mohammed Akli Ayoub,&nbsp;Richard Gordon,&nbsp;Hariharan Saminathan","doi":"10.1007/s11481-023-10077-8","DOIUrl":"10.1007/s11481-023-10077-8","url":null,"abstract":"<p><p>Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the selective degeneration of dopaminergic neurons in the substantia nigra pars compacta resulting in an irreversible and a debilitating motor dysfunction. Though both genetic and idiopathic factors are implicated in the disease etiology, idiopathic PD comprise the majority of clinical cases and is caused by exposure to environmental toxicants and oxidative stress. Fyn kinase activation has been identified as an early molecular signaling event that primes neuroinflammatory and neurodegenerative events associated with dopaminergic cell death. However, the upstream regulator of Fyn activation remains unidentified. We investigated whether the lipid and tyrosine phosphatase PTEN (Phosphatase and Tensin homolog deleted on chromosome 10) could be the upstream regulator of Fyn activation in PD models as PTEN has been previously reported to contribute to Parkinsonian pathology. Our findings, using bioluminescence resonance energy transfer (BRET) and immunoblotting, indicate for the first time that PTEN is a critical early stress sensor in response to oxidative stress and neurotoxicants in in vitro models of PD. Pharmacological attenuation of PTEN activity rescues dopaminergic neurons from neurotoxicant-induced cytotoxicity by modulating Fyn kinase activation. Our findings also identify PTEN's novel roles in contributing to mitochondrial dysfunction which contribute to neurodegenerative processes. Interestingly, we found that PTEN positively regulates interleukin-1β (IL-1β) and the transcription of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Taken together, we have identified PTEN as a disease course altering pharmacological target that may be further validated for the development of novel therapeutic strategies targeting PD.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"462-475"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10070877","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
SIRT2 Inhibition Rescues Neurodegenerative Pathology but Increases Systemic Inflammation in a Transgenic Mouse Model of Alzheimer's Disease. SIRT2抑制挽救了阿尔茨海默病转基因小鼠模型中的神经退行性病理学,但增加了全身炎症。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-09-12 DOI: 10.1007/s11481-023-10084-9
Noemi Sola-Sevilla, Alberto Mesa-Lombardo, Mikel Aleixo, Sara Expósito, Teresa Diaz-Perdigón, Amaya Azqueta, Farzad Zamani, Takayoshi Suzuki, Silvia Maioli, Francesca Eroli, Anna Matton, Maria J Ramírez, Maite Solas, Rosa M Tordera, Eduardo D Martín, Elena Puerta
{"title":"SIRT2 Inhibition Rescues Neurodegenerative Pathology but Increases Systemic Inflammation in a Transgenic Mouse Model of Alzheimer's Disease.","authors":"Noemi Sola-Sevilla,&nbsp;Alberto Mesa-Lombardo,&nbsp;Mikel Aleixo,&nbsp;Sara Expósito,&nbsp;Teresa Diaz-Perdigón,&nbsp;Amaya Azqueta,&nbsp;Farzad Zamani,&nbsp;Takayoshi Suzuki,&nbsp;Silvia Maioli,&nbsp;Francesca Eroli,&nbsp;Anna Matton,&nbsp;Maria J Ramírez,&nbsp;Maite Solas,&nbsp;Rosa M Tordera,&nbsp;Eduardo D Martín,&nbsp;Elena Puerta","doi":"10.1007/s11481-023-10084-9","DOIUrl":"10.1007/s11481-023-10084-9","url":null,"abstract":"<p><p>Sirtuin 2 (SIRT2) has been proposed to have a central role on aging, inflammation, cancer and neurodegenerative diseases; however, its specific function remains controversial. Recent studies propose SIRT2 pharmacological inhibition as a therapeutic strategy for several neurodegenerative diseases including Alzheimer's disease (AD). Surprisingly, none of these published studies regarding the potential interest of SIRT2 inhibition has assessed the peripheral adverse side consequences of this treatment. In this study, we demonstrate that the specific SIRT2 inhibitor, the compound 33i, does not exhibit genotoxic or mutagenic properties. Moreover, pharmacological treatment with 33i, improved cognitive dysfunction and long-term potentiation, reducing amyloid pathology and neuroinflammation in the APP/PS1 AD mouse model. However, this treatment increased peripheral levels of the inflammatory cytokines IL-1β, TNF, IL-6 and MCP-1. Accordingly, peripheral SIRT2 inhibition with the blood brain barrier impermeable compound AGK-2, worsened the cognitive capacities and increased systemic inflammation. The analysis of human samples revealed that SIRT2 is increased in the brain but not in the serum of AD patients. These results suggest that, although SIRT2 pharmacological inhibition may have beneficial consequences in neurodegenerative diseases, its pharmacological inhibition at the periphery would not be recommended and the systemic adverse side effects should be considered. This information is essential to maximize the therapeutic potential of SIRT2 inhibition not only for AD but also for other neurodegenerative diseases.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"529-550"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10577113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10204791","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
L-carnitine Modulates Cognitive Impairment Induced by Doxorubicin and Cyclophosphamide in Rats; Insights to Oxidative Stress, Inflammation, Synaptic Plasticity, Liver/brain, and Kidney/brain Axes. 左旋肉碱对阿霉素和环磷酰胺诱导的大鼠认知功能障碍的调节作用;对氧化应激、炎症、突触可塑性、肝脏/大脑和肾脏/大脑轴的见解。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-05-04 DOI: 10.1007/s11481-023-10062-1
Olivia Fayez Morid, Esther T Menze, Mariane G Tadros, Mina Y George
{"title":"L-carnitine Modulates Cognitive Impairment Induced by Doxorubicin and Cyclophosphamide in Rats; Insights to Oxidative Stress, Inflammation, Synaptic Plasticity, Liver/brain, and Kidney/brain Axes.","authors":"Olivia Fayez Morid,&nbsp;Esther T Menze,&nbsp;Mariane G Tadros,&nbsp;Mina Y George","doi":"10.1007/s11481-023-10062-1","DOIUrl":"10.1007/s11481-023-10062-1","url":null,"abstract":"<p><p>Chemotherapy-induced cognitive impairment in cancer patients is known as \"chemobrain\". Doxorubicin and Cyclophosphamide are two chemotherapeutic agents used in combination to treat solid tumors. L-carnitine was reported for its anti-oxidant and anti-inflammatory activities. The goal of the present study was to elucidate the neuroprotective effect of L-carnitine against chemobrain induced by Doxorubicin and Cyclophosphamide in rats. Rats were divided into five groups: Control group; Doxorubicin (4mg/kg, IV) and Cyclophosphamide (40mg/kg, IV)-treated group; two L-carnitine-treated groups (150 and 300mg/kg, ip) with Doxorubicin and Cyclophosphamide; and L-carnitine alone-treated group (300mg/kg). Doxorubicin and Cyclophosphamide induced histopathological changes in rats' hippocampi and prefrontal cortices, as well as reduced memory as evidenced by behavioural testing. L-carnitine treatment showed opposite effects. In addition, chemotherapy treatment enhanced oxidative stress via reducing catalase and glutathione levels, and inducing lipid peroxidation. By contrast, L-carnitine treatment showed powerful antioxidant effects reversing chemotherapy-induced oxidative damage. Moreover, chemotherapy combination induced inflammation via their effect on nuclear factor kappa B (p65), interleukin-1β, and tumor necrosis factor-α. However, L-carnitine treatment corrected such inflammatory responses. Furthermore, Doxorubicin and Cyclophosphamide reduced synaptic plasticity via hindering expression of brain-derived neurotrophic factor, phosphorylated cyclase response element binding protein, synaptophysin, and postsynaptic density protein 95 whereas protein expression of such synaptic plasticity biomarkers was enhanced by L-carnitine treatment. Finally, acetylcholinesterase activity was found to be enhanced by chemotherapy treatment affecting rats' memory while L-carnitine treatment reduced acetylcholinesterase activity. L-carnitine also showed hepatoprotective and renal protective effects suggesting liver/brain and kidney/brain axes as possible mechanisms for its neuroprotective effects.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"310-326"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10577097/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9404602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Maternal Immune Activation Induces Cortical Catecholaminergic Hypofunction and Cognitive Impairments in Offspring. 母体免疫激活诱导子代皮质儿茶酚胺能低下和认知障碍。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-05-20 DOI: 10.1007/s11481-023-10070-1
Blanca Perez-Palomar, Amaia M Erdozain, Ines Erkizia-Santamaría, Jorge E Ortega, J Javier Meana
{"title":"Maternal Immune Activation Induces Cortical Catecholaminergic Hypofunction and Cognitive Impairments in Offspring.","authors":"Blanca Perez-Palomar,&nbsp;Amaia M Erdozain,&nbsp;Ines Erkizia-Santamaría,&nbsp;Jorge E Ortega,&nbsp;J Javier Meana","doi":"10.1007/s11481-023-10070-1","DOIUrl":"10.1007/s11481-023-10070-1","url":null,"abstract":"<p><strong>Background: </strong>Impairment of specific cognitive domains in schizophrenia has been associated with prefrontal cortex (PFC) catecholaminergic deficits. Among other factors, prenatal exposure to infections represents an environmental risk factor for schizophrenia development in adulthood. However, it remains largely unknown whether the prenatal infection-induced changes in the brain may be associated with concrete switches in a particular neurochemical circuit, and therefore, if they could alter behavioral functions.</p><p><strong>Methods: </strong>In vitro and in vivo neurochemical evaluation of the PFC catecholaminergic systems was performed in offspring from mice undergoing maternal immune activation (MIA). The cognitive status was also evaluated. Prenatal viral infection was mimicked by polyriboinosinic-polyribocytidylic acid (poly(I:C)) administration to pregnant dams (7.5 mg/kg i.p., gestational day 9.5) and consequences were evaluated in adult offspring.</p><p><strong>Results: </strong>MIA-treated offspring showed disrupted recognition memory in the novel object recognition task (t = 2.30, p = 0.031). This poly(I:C)-based group displayed decreased extracellular dopamine (DA) concentrations compared to controls (t = 3.17, p = 0.0068). Potassium-evoked release of DA and noradrenaline (NA) were impaired in the poly(I:C) group (DA: F<sub>t</sub>[10,90] = 43.33, p < 0.0001; F<sub>tr</sub>[1,90] = 1.224, p = 0.2972; F<sub>i</sub>[10,90] = 5.916, p < 0.0001; n = 11); (NA: F<sub>t</sub>[10,90] = 36.27, p < 0.0001; F<sub>tr</sub>[1,90] = 1.841, p = 0.208; F<sub>i</sub>[10,90] = 8.686, p < 0.0001; n = 11). In the same way, amphetamine-evoked release of DA and NA were also impaired in the poly(I:C) group (DA: F<sub>t</sub>[8,328] = 22.01, p < 0.0001; F<sub>tr</sub>[1,328] = 4.507, p = 0.040; F<sub>i</sub>[8,328] = 2.319, p = 0.020; n = 43); (NA: F<sub>t</sub>[8,328] = 52.07; p < 0.0001; F<sub>tr</sub>[1,328] = 4.322; p = 0.044; F<sub>i</sub>[8,398] = 5.727; p < 0.0001; n = 43). This catecholamine imbalance was accompanied by increased dopamine D<sub>1</sub> and D<sub>2</sub> receptor expression (t = 2.64, p = 0.011 and t = 3.55, p = 0.0009; respectively), whereas tyrosine hydroxylase, DA and NA tissue content, DA and NA transporter (DAT/NET) expression and function were unaltered.</p><p><strong>Conclusions: </strong>MIA induces in offspring a presynaptic catecholaminergic hypofunction in PFC with cognitive impairment. This poly(I:C)-based model reproduces catecholamine phenotypes reported in schizophrenia and represents an opportunity for the study of cognitive impairment associated to this disorder.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"348-365"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10577104/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9494446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
As a Potential Therapeutic Target, C1q Induces Synapse Loss Via Inflammasome-activating Apoptotic and Mitochondria Impairment Mechanisms in Alzheimer's Disease. 作为一种潜在的治疗靶点,C1q通过炎症小体激活阿尔茨海默病的凋亡和线粒体损伤机制诱导突触损失。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-06-29 DOI: 10.1007/s11481-023-10076-9
Pei-Pei Guan, Tong-Qi Ge, Pu Wang
{"title":"As a Potential Therapeutic Target, C1q Induces Synapse Loss Via Inflammasome-activating Apoptotic and Mitochondria Impairment Mechanisms in Alzheimer's Disease.","authors":"Pei-Pei Guan,&nbsp;Tong-Qi Ge,&nbsp;Pu Wang","doi":"10.1007/s11481-023-10076-9","DOIUrl":"10.1007/s11481-023-10076-9","url":null,"abstract":"<p><p>C1q, the initiator of the classical pathway of the complement system, is activated during Alzheimer's disease (AD) development and progression and is especially associated with the production and deposition of β-amyloid protein (Aβ) and phosphorylated tau in β-amyloid plaques (APs) and neurofibrillary tangles (NFTs). Activation of C1q is responsible for induction of synapse loss, leading to neurodegeneration in AD. Mechanistically, C1q could activate glial cells, which results in the loss of synapses via regulation of synapse pruning and phagocytosis in AD. In addition, C1q induces neuroinflammation by inducing proinflammatory cytokine secretion, which is partially mediated by inflammasome activation. Activation of inflammasomes might mediate the effects of C1q on induction of synapse apoptosis. On the other hand, activation of C1q impairs mitochondria, which hinders the renovation and regeneration of synapses. All these actions of C1q contribute to the loss of synapses during neurodegeneration in AD. Therefore, pharmacological, or genetic interventions targeting C1q may provide potential therapeutic strategies for combating AD.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"267-284"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10074332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Impact of Adolescent Nicotine Exposure in Pre- and Post-natal Oxycodone Exposed Offspring. 青春期尼古丁暴露对暴露于羟考酮的产前和产后后代的影响。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-06-23 DOI: 10.1007/s11481-023-10074-x
Adrian Flores, Austin Gowen, Victoria L Schaal, Sneh Koul, Jordan B Hernandez, Sowmya V Yelamanchili, Gurudutt Pendyala
{"title":"Impact of Adolescent Nicotine Exposure in Pre- and Post-natal Oxycodone Exposed Offspring.","authors":"Adrian Flores,&nbsp;Austin Gowen,&nbsp;Victoria L Schaal,&nbsp;Sneh Koul,&nbsp;Jordan B Hernandez,&nbsp;Sowmya V Yelamanchili,&nbsp;Gurudutt Pendyala","doi":"10.1007/s11481-023-10074-x","DOIUrl":"10.1007/s11481-023-10074-x","url":null,"abstract":"<p><p>Perinatal exposure to prescription opioids pose a critical public health risk. Notably, research has found significant neurodevelopmental and behavioral deficits between in utero (IUO) and postnatal (PNO) oxycodone-exposed offspring but there is a notable gap in knowledge regarding the interaction of these groups to other drug exposure, particularly nicotine exposure. Nicotine's widespread use represents a ubiquitous clinical interaction that current research does not address. Children often experiment with drugs and risky behavior; therefore, adolescence is a key timepoint to characterize. This study employed an integrated systems approach to investigate escalating nicotine exposure in adolescence and subsequent nicotine withdrawal in the IUO- and PNO-offspring. Western blot analysis found synaptic protein alterations, especially upregulation of synaptophysin in IUO-withdrawal animals. RT-qPCR further validated immune dysfunction in the central nervous system (CNS). Peripheral nicotine metabolism was consistent with increased catabolism of nicotine concerning IUO animals. Lastly, behavioral assays found subtle deficits to withdrawal in nociception and anxiety-like behavior. This study showed, for the first time, the vulnerabilities of PNO- and IUO-exposed groups concerning nicotine use during early adolescence and withdrawal. Graphical Abstract.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"413-426"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9676491","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Characterization of Depression- and Anxiety-Like Behaviours in a Mouse Model of Relapsing-Remitting Multiple Sclerosis. 复发缓解型多发性硬化症小鼠模型中抑郁和焦虑样行为的特征。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-08-01 DOI: 10.1007/s11481-023-10080-z
Diulle Spat Peres, Fernanda Tibolla Viero, Patrícia Rodrigues, Laura de Barros Bernardes, Náthaly Andriguetto Ruviaro da Silva, Igor Ramos Lima, Gabrielli Martins, Paulo Cesar Lock Silveira, Marcella de Amorim Ferreira, Ana Merian Silva, Juliano Ferreira, Gabriela Trevisan
{"title":"Characterization of Depression- and Anxiety-Like Behaviours in a Mouse Model of Relapsing-Remitting Multiple Sclerosis.","authors":"Diulle Spat Peres,&nbsp;Fernanda Tibolla Viero,&nbsp;Patrícia Rodrigues,&nbsp;Laura de Barros Bernardes,&nbsp;Náthaly Andriguetto Ruviaro da Silva,&nbsp;Igor Ramos Lima,&nbsp;Gabrielli Martins,&nbsp;Paulo Cesar Lock Silveira,&nbsp;Marcella de Amorim Ferreira,&nbsp;Ana Merian Silva,&nbsp;Juliano Ferreira,&nbsp;Gabriela Trevisan","doi":"10.1007/s11481-023-10080-z","DOIUrl":"10.1007/s11481-023-10080-z","url":null,"abstract":"<p><p>Relapsing-remitting multiple sclerosis (RRMS) is an autoimmune neurological disease and is the most common subtype of MS. In addition, it is associated with the development of depression and anxiety. To date, depressive- and anxiety-like behaviours were only studied using models of progressive MS, which causes severe motor alterations. Thus, we sought to standardise the depressive and anxiety-like behaviours in an RRMS model induced by experimental autoimmune encephalomyelitis (RR-EAE) in mice. The RR-EAE model was induced in C57BL/6 female mice using myelin oligodendrocyte glycoprotein (MOG35-55) antigen and Quillaja saponin (Quil A) as an adjuvant. The immunisation of RR-EAE did not induce locomotor alteration but caused relapsing-remitting induction of clinical scores in mice until 35 post-immunization (p.i.). Also, increased levels of tumour necrosis factor alpha (TNF-α), astrocyte marker (GFAP), and microglial markers (IBA-1) were detected in the prefrontal cortex at 35 p.i. of RR-EAE. In the open field test, RR-EAE mice showed decreased time spent at the centre and sniffing behaviour (at days 21 and 34 p.i.). Also, on day 35 p.i. the RR-EAE group spent less time in the open arms and had decreased open-arm entries compared to control mice in the elevated plus maze (EPM) test, confirming the anxiety-like behaviour. At day 36° p.i. in the tail suspension test, mice showed depression-like behaviour with decreased latency time and increased immobility time. Thus, the RR-EAE model mimics the neuroinflammatory and behavioural features of the RRMS, including depression- and anxiety-like symptoms.</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"235-247"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9912115","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Endolysosomal Transporter DMT1 is Required for Morphine Regulation of Neuronal Ferritin Heavy Chain. 内溶酶体转运蛋白DMT1是吗啡调节神经元铁蛋白重链所必需的。
IF 6.2
Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology Pub Date : 2023-09-01 Epub Date: 2023-09-04 DOI: 10.1007/s11481-023-10082-x
Elena Irollo, Bradley Nash, Jared Luchetta, Renato Brandimarti, Olimpia Meucci
{"title":"The Endolysosomal Transporter DMT1 is Required for Morphine Regulation of Neuronal Ferritin Heavy Chain.","authors":"Elena Irollo, Bradley Nash, Jared Luchetta, Renato Brandimarti, Olimpia Meucci","doi":"10.1007/s11481-023-10082-x","DOIUrl":"10.1007/s11481-023-10082-x","url":null,"abstract":"<p><p>NeuroHIV and other neurologic disorders present with altered iron metabolism in central nervous system neurons. Many people with HIV also use opioids, which can worsen neuroHIV symptoms by further dysregulating neuronal iron metabolism. Our previous work demonstrated that the μ-opioid agonist morphine causes neuronal endolysosomes to release their iron stores, and neurons respond by upregulating ferritin heavy chain (FHC), an iron storage protein associated with cognitive impairment in neuroHIV. Here, we investigated if this process required divalent metal transporter 1 (DMT1), a well-known iron transporter expressed on endolysosomes. We first optimized conditions to detect DMT1 isoforms (DMT1 1B ± iron responsive element) using fluorescently labeled rat DMT1 constructs expressed in HEK-293 cells. We also expressed these constructs in primary rat cortical neurons to compare their expression and subcellular distribution with endogenous DMT1 isoforms. We found endogenous DMT1 isoforms in the cytoplasm that colocalized with lysosomal-associated protein 1 (LAMP1), a marker of endolysosomes. Next, we blocked endogenous DMT1 isoforms using ebselen, a potent pharmacological inhibitor of DMT1 iron transport. Ebselen pre-treatment blocked morphine's ability to upregulate FHC protein, suggesting this pathway requires DMT1 iron transport from endolysosomes. This was further validated using viral-mediated genetic silencing of DMT1±IRE in cortical neurons, which also blocked FHC upregulation in the presence of morphine. Overall, our work demonstrates that the μ-opioid agonist morphine utilizes the endolysosomal iron transporter DMT1 to modulate neuronal cellular iron metabolism, upregulate FHC protein, and contribute to cognitive decline in neuroHIV. Morphine requires DMT1 to upregulate neuronal FHC. Cortical neurons treated with morphine release their endolysosomal iron stores to the cytoplasm and upregulate FHC, an iron storage protein associated with dendritic spine deficits and cognitive impairment in neuroHIV. This pathway requires the endolysosomal iron transporter DMT1, as pharmacological and genetic inhibitors of the transporter completely block morphine's ability to upregulate FHC. Created with BioRender.com .</p>","PeriodicalId":73858,"journal":{"name":"Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology","volume":" ","pages":"495-508"},"PeriodicalIF":6.2,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10577102/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10148904","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信