Pub Date : 2024-09-10DOI: 10.1016/j.bbi.2024.09.011
Chronic HIV infection can dysregulate lipid/cholesterol metabolism in the peripheral system, contributing to the higher incidences of diabetes and atherosclerosis in HIV (+) individuals. Recently, accumulating evidence indicate that HIV proteins can also dysregulate lipid/cholesterol metabolism in the brain and such dysregulation could be linked with the pathogenesis of HIV-associated neurological disorders (HAND)/NeuroHIV. To further characterize the association between lipid/cholesterol metabolism and HAND, we employed HIV-inducible transactivator of transcription (iTAT) and control mice to compare their brain lipid profiles. Our results reveal that HIV-iTAT mice possess dysregulated lipid profiles and have increased numbers of lipid droplets (LDs) accumulation microglia (LDAM) in the brains. HIV protein TAT can upregulate LDs formation through enhancing the lipid/cholesterol synthesis in vitro. Mechanistically, HIV-TAT increases the expression of sterol regulatory element-binding protein 2 (SREBP2) through microRNA-124 downregulation. Cholesterol synthesis inhibition can block HIV-TAT-mediated NLRP3 inflammasome activation and microglial activation in vitro as well as mitigate aging-related behavioral impairment and memory deficiency in HIV-iTAT mice. Taken together, our results indicate an inherent role of lipid metabolism and LDAM in the pathogenesis of NeuroHIV (immunometabolism). These findings suggest that LDAM reversal through modulating lipid/cholesterol metabolism could be a novel therapeutic target for ameliorating NeuroHIV symptoms in chronic HIV (+) individuals.
慢性艾滋病病毒感染可使外周系统的脂质/胆固醇代谢失调,导致艾滋病病毒(+)感染者的糖尿病和动脉粥样硬化发病率升高。最近,越来越多的证据表明,HIV 蛋白也会导致大脑中脂质/胆固醇代谢失调,而这种失调可能与 HIV 相关神经系统疾病(HAND)/NeuroHIV 的发病机制有关。为了进一步确定脂质/胆固醇代谢与 HAND 之间的关系,我们采用了 HIV 诱导转录激活因子(iTAT)小鼠和对照小鼠来比较它们的脑脂质状况。我们的研究结果表明,HIV-iTAT 小鼠的脑脂质分布失调,脑内脂滴(LDs)堆积小胶质细胞(LDAM)数量增加。HIV 蛋白 TAT 可通过增强体外脂质/胆固醇合成来上调 LDs 的形成。从机制上讲,HIV-TAT 通过下调 microRNA-124 增加固醇调节元件结合蛋白 2(SREBP2)的表达。胆固醇合成抑制可阻断 HIV-TAT 介导的体外 NLRP3 炎性体激活和小胶质细胞激活,并减轻 HIV-iTAT 小鼠与衰老相关的行为障碍和记忆缺陷。综上所述,我们的研究结果表明,脂质代谢和 LDAM 在神经-艾滋病毒(免疫代谢)的发病机制中起着固有的作用。这些研究结果表明,通过调节脂质/胆固醇代谢来逆转 LDAM 可能是改善慢性 HIV(+)患者神经-HIV 症状的新型治疗靶点。
{"title":"HIV-TAT dysregulates microglial lipid metabolism through SREBP2/miR-124 axis: Implication of lipid droplet accumulation microglia in NeuroHIV","authors":"","doi":"10.1016/j.bbi.2024.09.011","DOIUrl":"10.1016/j.bbi.2024.09.011","url":null,"abstract":"<div><p>Chronic HIV infection can dysregulate lipid/cholesterol metabolism in the peripheral system, contributing to the higher incidences of diabetes and atherosclerosis in HIV (+) individuals. Recently, accumulating evidence indicate that HIV proteins can also dysregulate lipid/cholesterol metabolism in the brain and such dysregulation could be linked with the pathogenesis of HIV-associated neurological disorders (HAND)/NeuroHIV. To further characterize the association between lipid/cholesterol metabolism and HAND, we employed HIV-inducible transactivator of transcription (iTAT) and control mice to compare their brain lipid profiles. Our results reveal that HIV-iTAT mice possess dysregulated lipid profiles and have increased numbers of lipid droplets (LDs) accumulation microglia (LDAM) in the brains. HIV protein TAT can upregulate LDs formation through enhancing the lipid/cholesterol synthesis <em>in vitro</em>. Mechanistically, HIV-TAT increases the expression of sterol regulatory element-binding protein 2 (SREBP2) through microRNA-124 downregulation. Cholesterol synthesis inhibition can block HIV-TAT-mediated NLRP3 inflammasome activation and microglial activation <em>in vitro</em> as well as mitigate aging-related behavioral impairment and memory deficiency in HIV-iTAT mice. Taken together, our results indicate an inherent role of lipid metabolism and LDAM in the pathogenesis of NeuroHIV (immunometabolism). These findings suggest that LDAM reversal through modulating lipid/cholesterol metabolism could be a novel therapeutic target for ameliorating NeuroHIV symptoms in chronic HIV (+) individuals.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142173442","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-05DOI: 10.1016/j.bbi.2024.09.002
Multiple Sclerosis (MS) is a chronic degenerative disease of the central nervous system (CNS) characterized by inflammation, demyelination, and progressive neurodegeneration. These processes, combined with the failure of reparative remyelination initiated by oligodendrocyte precursor cells (OPCs), lead to irreversible neurological impairment. The cytokine tumor necrosis factor (TNF) has been implicated in CNS repair via activation of its cognate receptor TNFR2 in glia. Here, we demonstrate the important role of TNFR2 in regulating OPC function in vivo during demyelinating disease, and that TNFR2 expressed in OPCs modulates OPC-microglia interactions. In PdgfrαCreERT:Tnfrsf1bfl/fl:Eyfp mice with selective TNFR2 ablation in OPCs, we observed an earlier onset and disease peak in experimental autoimmune encephalomyelitis (EAE). This was associated with accelerated immune cell infiltration and increased microglia activation in the spinal cord. Similarly, PdgfrαCreERT:Tnfrsf1bfl/fl:Eyfp mice showed rapid and increased microglia reactivity compared to control mice in the corpus callosum after cuprizone-induced demyelination, followed by chronic reduction in the number of mature myelinating oligodendrocytes (OLs). With EAE and cuprizone models combined, we uncovered that TNFR2 does not have a cell autonomous role in OPC differentiation, but may be important for survival of newly formed mature OLs. Finally, using an in vitro approach, we demonstrated that factors released by Tnfrsf1b ablated OPCs drove microglia to develop an exacerbated “foamy” phenotype when incubated with myelin-rich spinal cord homogenate, aberrantly increasing lysosomal lipid accumulation. Together, our data indicate that TNFR2 signaling in OPCs is protective by dampening their immune-inflammatory activation and by suppressing neurotoxic microglia reactivity. This suggests that boosting TNFR2 activation or its downstream cascades could be an effective strategy to restore OPC reparative capacity in neuroimmune and demyelinating disease.
{"title":"TNFR2 signaling in oligodendrocyte precursor cells suppresses their immune-inflammatory function and detrimental microglia activation in CNS demyelinating disease","authors":"","doi":"10.1016/j.bbi.2024.09.002","DOIUrl":"10.1016/j.bbi.2024.09.002","url":null,"abstract":"<div><p>Multiple Sclerosis (MS) is a chronic degenerative disease of the central nervous system (CNS) characterized by inflammation, demyelination, and progressive neurodegeneration. These processes, combined with the failure of reparative remyelination initiated by oligodendrocyte precursor cells (OPCs), lead to irreversible neurological impairment. The cytokine tumor necrosis factor (TNF) has been implicated in CNS repair via activation of its cognate receptor TNFR2 in glia. Here, we demonstrate the important role of TNFR2 in regulating OPC function <em>in vivo</em> during demyelinating disease, and that TNFR2 expressed in OPCs modulates OPC-microglia interactions. In <em>Pdgfrα</em><sup>CreERT</sup>:<em>Tnfrsf1b</em><sup>fl/fl</sup>:<em>Eyfp</em> mice with selective TNFR2 ablation in OPCs, we observed an earlier onset and disease peak in experimental autoimmune encephalomyelitis (EAE). This was associated with accelerated immune cell infiltration and increased microglia activation in the spinal cord. Similarly, <em>Pdgfrα</em><sup>CreERT</sup>:<em>Tnfrsf1b</em><sup>fl/fl</sup>:<em>Eyfp</em> mice showed rapid and increased microglia reactivity compared to control mice in the corpus callosum after cuprizone-induced demyelination, followed by chronic reduction in the number of mature myelinating oligodendrocytes (OLs). With EAE and cuprizone models combined, we uncovered that TNFR2 does not have a cell autonomous role in OPC differentiation, but may be important for survival of newly formed mature OLs. Finally, using an <em>in vitro</em> approach, we demonstrated that factors released by <em>Tnfrsf1b</em> ablated OPCs drove microglia to develop an exacerbated “foamy” phenotype when incubated with myelin-rich spinal cord homogenate, aberrantly increasing lysosomal lipid accumulation. Together, our data indicate that TNFR2 signaling in OPCs is protective by dampening their immune-inflammatory activation and by suppressing neurotoxic microglia reactivity. This suggests that boosting TNFR2 activation or its downstream cascades could be an effective strategy to restore OPC reparative capacity in neuroimmune and demyelinating disease.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145203","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-05DOI: 10.1016/j.bbi.2024.09.004
Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) mediated by CD4+ T helper (Th) cells, and characterized by immune cell infiltration, demyelination and neurodegeneration, with no definitive cure available. Thus, it is pivotal and imperative to acquire more profound comprehension of the underlying mechanisms implicated in MS. Dysregulated immune responses are widely believed to play a primary role in the pathogenesis of MS. Recently, a plethora of studies have demonstrated the involvement of T follicular helper (Tfh) cells and tertiary lymphoid-like structures (TLSs) in the pathogenesis and progression of MS. Cathepsin C (CatC) is a cysteine exopeptidase which is crucial for the activation of immune-cell-associated serine proteinases in many inflammatory diseases in peripheral system, such as rheumatoid arthritis and septicemia. We have previously demonstrated that CatC is involved in neuroinflammation and exacerbates demyelination in both cuprizone-induced and experimental autoimmune encephalomyelitis (EAE) mouse models. However, the underlying immunopathological mechanism remains elusive. In the present study, we established a recombinant myelin oligodendrocyte glycoprotein 35–55 peptide-induced EAE model using conditional CatC overexpression mice to investigate the effects of CatC on the alteration of CD4+ Th subsets, including Th1, Th2, Th17, Tfh and T regulatory cells. Our findings demonstrated that CatC particularly enhanced the population of Tfh cell in the brain, resulting in the earlier onset and more severe chronic syndrome of EAE. Furthermore, CatC promoted the formation of TLSs in the brain, leading to persistent neuroinflammation and exacerbating the severity of EAE in the chronic phase. Conversely, treatment with AZD7986, a specific inhibitor of CatC, effectively attenuated the syndrome of EAE and its effects caused by CatC both in vivo and in vitro. These findings provide a novel insight into the critical role of CatC in innate and adaptive immunity in EAE, and specific inhibitor of CatC, AZD7986, may contribute to potential therapeutic strategies for MS.
多发性硬化症(MS)是一种由 CD4+ T 辅助细胞(Th)介导的中枢神经系统(CNS)慢性自身免疫性疾病,以免疫细胞浸润、脱髓鞘和神经变性为特征,目前尚无根治方法。因此,更深入地了解多发性硬化症的内在机制至关重要,势在必行。人们普遍认为,失调的免疫反应在多发性硬化症的发病机制中起着主要作用。最近,大量研究表明,T 滤泡辅助细胞(Tfh)和三级淋巴样结构(TLSs)参与了多发性硬化症的发病和进展。Cathepsin C(CatC)是一种半胱氨酸外肽酶,在类风湿性关节炎和败血症等许多外周系统炎症性疾病中,它是激活免疫细胞相关丝氨酸蛋白酶的关键。我们之前已经证明,CatC 参与了铜绿素诱导的神经炎症,并加剧了实验性自身免疫性脑脊髓炎(EAE)小鼠模型的脱髓鞘。然而,其潜在的免疫病理机制仍然难以捉摸。在本研究中,我们利用条件性CatC过表达小鼠建立了重组髓鞘少突胶质细胞糖蛋白35-55肽诱导的EAE模型,研究了CatC对CD4+ Th亚群(包括Th1、Th2、Th17、Tfh和T调节细胞)改变的影响。我们的研究结果表明,CatC特别增强了脑内Tfh细胞的数量,导致EAE慢性综合征发病更早、更严重。此外,CatC还能促进脑内TLS的形成,导致持续性神经炎症,加重慢性期EAE的严重程度。相反,CatC的特异性抑制剂AZD7986能有效减轻EAE综合征及其在体内和体外由CatC引起的影响。这些研究结果提供了一个新的视角,让我们了解到CatC在EAE的先天性免疫和适应性免疫中的关键作用,而CatC的特异性抑制剂AZD7986可能有助于多发性硬化症的潜在治疗策略。
{"title":"Cathepsin C exacerbates EAE by promoting the expansion of Tfh cells and the formation of TLSs in the CNS","authors":"","doi":"10.1016/j.bbi.2024.09.004","DOIUrl":"10.1016/j.bbi.2024.09.004","url":null,"abstract":"<div><p>Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) mediated by CD4<sup>+</sup> T helper (Th) cells, and characterized by immune cell infiltration, demyelination and neurodegeneration, with no definitive cure available. Thus, it is pivotal and imperative to acquire more profound comprehension of the underlying mechanisms implicated in MS. Dysregulated immune responses are widely believed to play a primary role in the pathogenesis of MS. Recently, a plethora of studies have demonstrated the involvement of T follicular helper (Tfh) cells and tertiary lymphoid-like structures (TLSs) in the pathogenesis and progression of MS. Cathepsin C (CatC) is a cysteine exopeptidase which is crucial for the activation of immune-cell-associated serine proteinases in many inflammatory diseases in peripheral system, such as rheumatoid arthritis and septicemia. We have previously demonstrated that CatC is involved in neuroinflammation and exacerbates demyelination in both cuprizone-induced and experimental autoimmune encephalomyelitis (EAE) mouse models. However, the underlying immunopathological mechanism remains elusive. In the present study, we established a recombinant myelin oligodendrocyte glycoprotein 35–55 peptide-induced EAE model using conditional CatC overexpression mice to investigate the effects of CatC on the alteration of CD4<sup>+</sup> Th subsets, including Th1, Th2, Th17, Tfh and T regulatory cells. Our findings demonstrated that CatC particularly enhanced the population of Tfh cell in the brain, resulting in the earlier onset and more severe chronic syndrome of EAE. Furthermore, CatC promoted the formation of TLSs in the brain, leading to persistent neuroinflammation and exacerbating the severity of EAE in the chronic phase. Conversely, treatment with AZD7986, a specific inhibitor of CatC, effectively attenuated the syndrome of EAE and its effects caused by CatC both <em>in vivo</em> and <em>in vitro</em>. These findings provide a novel insight into the critical role of CatC in innate and adaptive immunity in EAE, and specific inhibitor of CatC, AZD7986, may contribute to potential therapeutic strategies for MS.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145199","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-05DOI: 10.1016/j.bbi.2024.09.003
<div><h3>Background</h3><p>Major depressive disorder (MDD) is a highly prevalent condition with a substantial incidence of relapse or treatment resistance. A subset of patients show evidence of low-grade inflammation, with these patients having a higher likelihood of more severe or difficult to treat courses of illness. Anti-inflammatory treatment of MDD has been investigated with mixed results, and no known studies have included assessments beyond cessation of the anti-inflammatory agent, meaning it remains unknown if any benefit from treatment persists. The objective of the present study was to investigate treatment outcomes up to 29 weeks post-cessation of celecoxib or placebo augmentation of an antidepressant, and how concentrations of selected inflammatory markers change over the same period.</p></div><div><h3>Methods</h3><p>The PREDDICT parallel-group, randomised, double-blind, placebo-controlled trial (University of Adelaide, Australia) ran from December 2017 to April 2020. Participants with MDD were stratified into normal range or elevated inflammation strata according to screening concentrations of high sensitivity C-reactive protein (hsCRP). Participants were randomised to treatment with vortioxetine and celecoxib or vortioxetine and placebo for six weeks, and vortioxetine alone for an additional 29 weeks (35 total weeks). Following a previous publication of results from the six-week RCT phase, exploratory analyses were performed on Montgomery–Åsberg Depression Rating Scale (MADRS) scores, response and remission outcomes, and selected peripheral inflammatory markers across the entire study duration up to week 35.</p></div><div><h3>Results</h3><p>Participants retained at each observation were baseline N=119, week 2 N=115, week 4 N=103, week 6 N=104, week 8 N=98, week 22 N=81, and week 35 N=60. Those in the elevated hsCRP celecoxib-augmented group had a statistically significantly greater reduction in MADRS score from baseline to week 35 compared to all other groups, demonstrating the greatest clinical improvement long-term, despite no group or strata differences at preceding time points. Response and remission outcomes did not differ by treatment group or hsCRP strata at any time point. Changes in hsCRP between baseline and week 35 and Tumour Necrosis Factor-α (TNF-α) concentrations between baseline and week 6 and baseline and week 35 were statistically significantly associated with MADRS scores observed at week 6 and week 35 respectively, with reducing TNF-α concentrations associated with reducing MADRS scores and vice versa in each case. A post-hoc stratification of the participant cohort by baseline TNF-α concentrations led to significant prediction by the derived strata on clinical response at weeks 6, 8 and 35, with participants with elevated baseline TNF-α less likely to achieve clinical response.</p></div><div><h3>Interpretation</h3><p>The present analysis suggests for the first time a possible longer-term clinical benefit of cele
{"title":"Long-term characterisation of the relationship between change in depression severity and change in inflammatory markers following inflammation-stratified treatment with vortioxetine augmented with celecoxib or placebo","authors":"","doi":"10.1016/j.bbi.2024.09.003","DOIUrl":"10.1016/j.bbi.2024.09.003","url":null,"abstract":"<div><h3>Background</h3><p>Major depressive disorder (MDD) is a highly prevalent condition with a substantial incidence of relapse or treatment resistance. A subset of patients show evidence of low-grade inflammation, with these patients having a higher likelihood of more severe or difficult to treat courses of illness. Anti-inflammatory treatment of MDD has been investigated with mixed results, and no known studies have included assessments beyond cessation of the anti-inflammatory agent, meaning it remains unknown if any benefit from treatment persists. The objective of the present study was to investigate treatment outcomes up to 29 weeks post-cessation of celecoxib or placebo augmentation of an antidepressant, and how concentrations of selected inflammatory markers change over the same period.</p></div><div><h3>Methods</h3><p>The PREDDICT parallel-group, randomised, double-blind, placebo-controlled trial (University of Adelaide, Australia) ran from December 2017 to April 2020. Participants with MDD were stratified into normal range or elevated inflammation strata according to screening concentrations of high sensitivity C-reactive protein (hsCRP). Participants were randomised to treatment with vortioxetine and celecoxib or vortioxetine and placebo for six weeks, and vortioxetine alone for an additional 29 weeks (35 total weeks). Following a previous publication of results from the six-week RCT phase, exploratory analyses were performed on Montgomery–Åsberg Depression Rating Scale (MADRS) scores, response and remission outcomes, and selected peripheral inflammatory markers across the entire study duration up to week 35.</p></div><div><h3>Results</h3><p>Participants retained at each observation were baseline N=119, week 2 N=115, week 4 N=103, week 6 N=104, week 8 N=98, week 22 N=81, and week 35 N=60. Those in the elevated hsCRP celecoxib-augmented group had a statistically significantly greater reduction in MADRS score from baseline to week 35 compared to all other groups, demonstrating the greatest clinical improvement long-term, despite no group or strata differences at preceding time points. Response and remission outcomes did not differ by treatment group or hsCRP strata at any time point. Changes in hsCRP between baseline and week 35 and Tumour Necrosis Factor-α (TNF-α) concentrations between baseline and week 6 and baseline and week 35 were statistically significantly associated with MADRS scores observed at week 6 and week 35 respectively, with reducing TNF-α concentrations associated with reducing MADRS scores and vice versa in each case. A post-hoc stratification of the participant cohort by baseline TNF-α concentrations led to significant prediction by the derived strata on clinical response at weeks 6, 8 and 35, with participants with elevated baseline TNF-α less likely to achieve clinical response.</p></div><div><h3>Interpretation</h3><p>The present analysis suggests for the first time a possible longer-term clinical benefit of cele","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S088915912400597X/pdfft?md5=8e0dd42e9e6036c6e53c18bda2b2679b&pid=1-s2.0-S088915912400597X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145201","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-05DOI: 10.1016/j.bbi.2024.09.001
Autism spectrum disorder (ASD) is a set of heterogeneous neurodevelopmental conditions, with a highly diverse genetic hereditary component, including altered neuronal circuits, that has an impact on communication skills and behaviours of the affected individuals. Beside the recognised role of neuronal alterations, perturbations of microglia and the associated neuroinflammatory processes have emerged as credible contributors to aetiology and physiopathology of ASD. Mutations in NRXN1, a member of the neurexin family of cell-surface receptors that bind neuroligin, have been associated to ASD. NRXN1 is known to be expressed by neurons where it facilitates synaptic contacts, but it has also been identified in glial cells including microglia. Asserting the impact of ASD-related genes on neuronal versus microglia functions has been challenging. Here, we present an ASD subject-derived induced pluripotent stem cells (iPSC)-based in vitro system to characterise the effects of the ASD-associated NRXN1 gene deletion on neurons and microglia, as well as on the ability of microglia to support neuronal circuit formation and function. Using this approach, we demonstrated that NRXN1 deletion, impacting on the expression of the alpha isoform (NRXN1α), in microglia leads to microglial alterations and release of IL6, a pro-inflammatory interleukin associated with ASD. Moreover, microglia bearing the NRXN1α-deletion, lost the ability to support the formation of functional neuronal networks. The use of recombinant IL6 protein on control microglia-neuron co-cultures or neutralizing antibody to IL6 on their NRXN1α-deficient counterparts, supported a direct contribution of IL6 to the observed neuronal phenotype. Altogether, our data suggest that, in addition to neurons, microglia are also negatively affected by NRXN1α-deletion, and this significantly contributes to the observed neuronal circuit aberrations.
{"title":"Bi-allelic NRXN1α deletion in microglia derived from iPSC of an autistic patient increases interleukin-6 production and impairs supporting function on neuronal networking","authors":"","doi":"10.1016/j.bbi.2024.09.001","DOIUrl":"10.1016/j.bbi.2024.09.001","url":null,"abstract":"<div><p>Autism spectrum disorder (ASD) is a set of heterogeneous neurodevelopmental conditions, with a highly diverse genetic hereditary component, including altered neuronal circuits, that has an impact on communication skills and behaviours of the affected individuals. Beside the recognised role of neuronal alterations, perturbations of microglia and the associated neuroinflammatory processes have emerged as credible contributors to aetiology and physiopathology of ASD. Mutations in <em>NRXN1</em>, a member of the neurexin family of cell-surface receptors that bind neuroligin, have been associated to ASD. <em>NRXN1</em> is known to be expressed by neurons where it facilitates synaptic contacts, but it has also been identified in glial cells including microglia. Asserting the impact of ASD-related genes on neuronal versus microglia functions has been challenging. Here, we present an ASD subject-derived induced pluripotent stem cells (iPSC)-based <em>in vitro</em> system to characterise the effects of the ASD-associated <em>NRXN1</em> gene deletion on neurons and microglia, as well as on the ability of microglia to support neuronal circuit formation and function. Using this approach, we demonstrated that <em>NRXN1</em> deletion, impacting on the expression of the alpha isoform (NRXN1α), in microglia leads to microglial alterations and release of IL6, a pro-inflammatory interleukin associated with ASD. Moreover, microglia bearing the <em>NRXN1α</em>-deletion, lost the ability to support the formation of functional neuronal networks. The use of recombinant IL6 protein on control microglia-neuron co-cultures or neutralizing antibody to IL6 on their <em>NRXN1α</em>-deficient counterparts, supported a direct contribution of IL6 to the observed neuronal phenotype. Altogether, our data suggest that, in addition to neurons, microglia are also negatively affected by <em>NRXN1α</em>-deletion, and this significantly contributes to the observed neuronal circuit aberrations.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0889159124005956/pdfft?md5=7d20d55a84200c0aeaa89eba7b5747a6&pid=1-s2.0-S0889159124005956-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145198","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-04DOI: 10.1016/j.bbi.2024.09.006
Traumatic brain injury (TBI) is a disabling neurotraumatic condition and the leading cause of injury-related deaths and disability in the United States. Attenuation of neuroinflammation early after TBI is considered an important treatment target; however, while these inflammatory responses can induce secondary brain injury, they are also involved in the repair of the nervous system. Pioglitazone, which activates peroxisome proliferator-activated receptor gamma, has been shown to decrease inflammation acutely after TBI, but the long-term consequences of its use remain unknown. For this reason, the impacts of treatment with pioglitazone during the acute/subacute phase (30 min after injury and each subsequent 24 h for 5 days) after TBI were interrogated during the chronic phase (30- and 274-days post-injury (DPI)) in mice using the controlled cortical impact model of experimental TBI. Acute/subacute pioglitazone treatment after TBI results in long-term deleterious consequences, including disruption of tau homeostasis, chronic glial cell activation, neuronal pathology, and worsened injury severity particularly at 274 DPI, with male mice being more susceptible than female mice. Further, male pioglitazone-treated TBI mice exhibited increased dominant and offensive-like behavior while having a decreased non-social exploring behavior at 274 DPI. After TBI, both sexes exhibited glial activation at 30 DPI when treated with pioglitazone; however, while injury severity was increased in females it was not impacted in male mice. This work reveals that although pioglitazone has been shown to lead to attenuated TBI outcomes acutely, sex-based differences, timing and long-term consequences of treatment with glitazones must be considered and further studied prior to their clinical use for TBI therapy.
{"title":"Chronic glial activation and behavioral alterations induced by acute/subacute pioglitazone treatment in a mouse model of traumatic brain injury","authors":"","doi":"10.1016/j.bbi.2024.09.006","DOIUrl":"10.1016/j.bbi.2024.09.006","url":null,"abstract":"<div><p>Traumatic brain injury (TBI) is a disabling neurotraumatic condition and the leading cause of injury-related deaths and disability in the United States. Attenuation of neuroinflammation early after TBI is considered an important treatment target; however, while these inflammatory responses can induce secondary brain injury, they are also involved in the repair of the nervous system. Pioglitazone, which activates peroxisome proliferator-activated receptor gamma, has been shown to decrease inflammation acutely after TBI, but the long-term consequences of its use remain unknown. For this reason, the impacts of treatment with pioglitazone during the acute/subacute phase (30 min after injury and each subsequent 24 h for 5 days) after TBI were interrogated during the chronic phase (30- and 274-days post-injury (DPI)) in mice using the controlled cortical impact model of experimental TBI. Acute/subacute pioglitazone treatment after TBI results in long-term deleterious consequences, including disruption of tau homeostasis, chronic glial cell activation, neuronal pathology, and worsened injury severity particularly at 274 DPI, with male mice being more susceptible than female mice. Further, male pioglitazone-treated TBI mice exhibited increased dominant and offensive-like behavior while having a decreased non-social exploring behavior at 274 DPI. After TBI, both sexes exhibited glial activation at 30 DPI when treated with pioglitazone; however, while injury severity was increased in females it was not impacted in male mice. This work reveals that although pioglitazone has been shown to lead to attenuated TBI outcomes acutely, sex-based differences, timing and long-term consequences of treatment with glitazones must be considered and further studied prior to their clinical use for TBI therapy.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0889159124005981/pdfft?md5=024e88ba831a724fc2e4a998ca770226&pid=1-s2.0-S0889159124005981-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-04DOI: 10.1016/j.bbi.2024.09.007
Background
Individuals with Schizophrenia Spectrum Disorders (SSD) often suffer from obesity and do limited Physical Activity (PA). PA has many beneficial effects on a variety of somatic and mental variables and it should be strengthened among people with mental disorders. The relationship between Body Mass Index (BMI), Waist Circumference (WC), and PA in this population is poorly understood, with a lack of precise PA assessment. This study investigates the association between BMI, WC, weight, and PA in individuals with SSD and controls using accelerometers.
Methods
One hundred twenty-six patients with SSD (residents and outpatients) and 110 sex- and age-matched controls were enrolled. Clinical, sociodemographic, and quality-of-life data were collected. PA was measured with a tri-axial ActiGraph GT9X and quantified by Vector Magnitude (VM). Relationships between PA and BMI, WC, and weight changes were analysed using linear regression models.
Results
Patients were more likely to be unmarried, unemployed, and less educated compared to controls (p < 0.001). Residents had more medical comorbidities (p = 0.001), while outpatients had higher BMI, weight, and WC (p < 0.001). Residents reported more severe psychopathology, lower functioning, and greater use of psychopharmacological medications (p < 0.001). Higher PA levels were not significantly associated with lower BMI, WC, or weight. Although not statistically significant, increased PA showed a trend towards lower obesity risk.
Conclusions
Sociodemographic, medical, and clinical characteristics of individuals with SSD define vulnerability factors that can inform tailored interventions to improve PA.
{"title":"Association of Weight Status and Waist Circumference with Physical Activity in people with Schizophrenia Spectrum Disorders and healthy controls","authors":"","doi":"10.1016/j.bbi.2024.09.007","DOIUrl":"10.1016/j.bbi.2024.09.007","url":null,"abstract":"<div><h3>Background</h3><p>Individuals with Schizophrenia Spectrum Disorders (SSD) often suffer from obesity and do limited Physical Activity (PA). PA has many beneficial effects on a variety of somatic and mental variables and it should be strengthened among people with mental disorders. The relationship between Body Mass Index (BMI), Waist Circumference (WC), and PA in this population is poorly understood, with a lack of precise PA assessment. This study investigates the association between BMI, WC, weight, and PA in individuals with SSD and controls using accelerometers.</p></div><div><h3>Methods</h3><p>One hundred twenty-six patients with SSD (residents and outpatients) and 110 sex- and age-matched controls were enrolled. Clinical, sociodemographic, and quality-of-life data were collected. PA was measured with a tri-axial ActiGraph GT9X and quantified by Vector Magnitude (VM). Relationships between PA and BMI, WC, and weight changes were analysed using linear regression models.</p></div><div><h3>Results</h3><p>Patients were more likely to be unmarried, unemployed, and less educated compared to controls (p < 0.001). Residents had more medical comorbidities (p = 0.001), while outpatients had higher BMI, weight, and WC (p < 0.001). Residents reported more severe psychopathology, lower functioning, and greater use of psychopharmacological medications (p < 0.001). Higher PA levels were not significantly associated with lower BMI, WC, or weight. Although not statistically significant, increased PA showed a trend towards lower obesity risk.</p></div><div><h3>Conclusions</h3><p>Sociodemographic, medical, and clinical characteristics of individuals with SSD define vulnerability factors that can inform tailored interventions to improve PA.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145204","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-04DOI: 10.1016/S0889-1591(24)00586-5
{"title":"PNIRS Society Announcements","authors":"","doi":"10.1016/S0889-1591(24)00586-5","DOIUrl":"10.1016/S0889-1591(24)00586-5","url":null,"abstract":"","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142137185","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-04DOI: 10.1016/j.bbi.2024.09.005
Immune abnormalities including an insufficiency of regulatory T cells (Treg) and increased blood-based inflammatory markers have been observed in bipolar disorders (BD), particularly during depression. As Tregs are pivotal to control inflammation, Treg stimulation by low-dose IL-2 (IL-2LD) could have a therapeutic impact on bipolar depression. We performed a randomized, double-blind, placebo-controlled (2 active: 1 placebo) proof-of-concept trial of add-on IL-2LD in patients with bipolar depression. Patients received a placebo or IL-2LD (1MIU) once a day for 5 days, and then once a week for 4 weeks starting on week 2. The primary objective was to demonstrate a biological Treg response to IL-2LD assessed by fold increase in Treg percentage of CD4 + cells from baseline to day 5. Secondary objectives included safety assessment and mood improvement throughout the study period. This trial is registered with ClinicalTrials.gov, number NCT04133233. Fourteen patients with bipolar depression were included, with 4 receiving placebo and 10 IL-2LD. Baseline clinical and biological characteristics were balanced between groups. The primary evaluation criterion was met, with IL-2LD expanding 1.17 [95 % CI 1.01–1.34] vs 1.01 [95 % CI 0.90–1.12] (p = 0.0421) and activating Tregs. Secondary evaluation criteria were also met with significant improvements of depressive symptoms and global functioning from day-15 onwards in the IL-2LD treated patients. The treatment was well-tolerated, with no serious adverse events related to treatment. This proof-of-concept trial shows that stimulating Tregs in patients with bipolar depression is safe and associated with clinical improvements. This supports a pathophysiological role of inflammation in BD and warrants pursuing the evaluation of IL-2LD as an adjunct treatment of major mood disorders.
在双相情感障碍(BD)中,尤其是在抑郁期间,已观察到包括调节性T细胞(Treg)不足和血液中炎症标志物增加在内的免疫异常。由于Tregs在控制炎症方面起着关键作用,因此通过低剂量IL-2(IL-2LD)刺激Treg可能会对双相抑郁症产生治疗效果。我们在双相抑郁症患者中开展了一项随机、双盲、安慰剂对照(2 项活性:1 项安慰剂)的附加 IL-2LD 概念验证试验。患者接受安慰剂或IL-2LD(1MIU)治疗,每天一次,共5天,然后从第2周开始每周一次,共4周。首要目标是证明IL-2LD的生物Treg反应,评估指标是CD4 +细胞的Treg百分比从基线到第5天的增加倍数。次要目标包括整个研究期间的安全性评估和情绪改善。该试验已在 ClinicalTrials.gov 注册,编号为 NCT04133233。14名双相抑郁症患者参与了该试验,其中4人接受安慰剂治疗,10人接受IL-2LD治疗。各组的基线临床和生物学特征均衡。结果符合主要评估标准,IL-2LD的治疗效果为1.17 [95 % CI 1.01-1.34] vs 1.01 [95 % CI 0.90-1.12] (p = 0.0421),并激活了Tregs。IL-2LD治疗患者的抑郁症状和整体功能自第15天起显著改善,符合二级评估标准。治疗耐受性良好,未出现与治疗相关的严重不良事件。这项概念验证试验表明,刺激双相抑郁症患者体内的Tregs是安全的,并能改善临床症状。这支持了炎症在双相抑郁症中的病理生理作用,值得继续评估IL-2LD作为重性情绪障碍辅助治疗的效果。
{"title":"Low-dose interleukin-2 in patients with bipolar depression: A phase 2 randomised double-blind placebo-controlled trial","authors":"","doi":"10.1016/j.bbi.2024.09.005","DOIUrl":"10.1016/j.bbi.2024.09.005","url":null,"abstract":"<div><p>Immune abnormalities including an insufficiency of regulatory T cells (Treg) and increased blood-based inflammatory markers have been observed in bipolar disorders (BD), particularly during depression. As Tregs are pivotal to control inflammation, Treg stimulation by low-dose IL-2 (IL-2<sub>LD</sub>) could have a therapeutic impact on bipolar depression. We performed a randomized, double-blind, placebo-controlled (2 active: 1 placebo) proof-of-concept trial of add-on IL-2<sub>LD</sub> in patients with bipolar depression. Patients received a placebo or IL-2<sub>LD</sub> (1MIU) once a day for 5 days, and then once a week for 4 weeks starting on week 2. The primary objective was to demonstrate a biological Treg response to IL-2<sub>LD</sub> assessed by fold increase in Treg percentage of CD4 + cells from baseline to day 5. Secondary objectives included safety assessment and mood improvement throughout the study period. This trial is registered with <span><span>ClinicalTrials.gov</span><svg><path></path></svg></span>, number NCT04133233. Fourteen patients with bipolar depression were included, with 4 receiving placebo and 10 IL-2<sub>LD</sub>. Baseline clinical and biological characteristics were balanced between groups. The primary evaluation criterion was met, with IL-2<sub>LD</sub> expanding 1.17 [95 % CI 1.01–1.34] vs 1.01 [95 % CI 0.90–1.12] (p = 0.0421) and activating Tregs. Secondary evaluation criteria were also met with significant improvements of depressive symptoms and global functioning from day-15 onwards in the IL-2<sub>LD</sub> treated patients. The treatment was well-tolerated, with no serious adverse events related to treatment. This proof-of-concept trial shows that stimulating Tregs in patients with bipolar depression is safe and associated with clinical improvements. This supports a pathophysiological role of inflammation in BD and warrants pursuing the evaluation of IL-2<sub>LD</sub> as an adjunct treatment of major mood disorders.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142145202","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-03DOI: 10.1016/j.bbi.2024.08.054
Much evidence on heightened inflammation and social behavior focuses on social withdrawal. Building on recent theory (Muscatell and Inagaki, 2021), we focused instead on the socially affiliative experience of sex. We investigated the interplay between immunology and motivation on sexual well-being among 158 individuals in romantic relationships. Inflammation, indexed by C-reactive protein (CRP), and sexual well-being were measured multiple times over a month. Relational approach motivation (i.e., motivation toward rewards in relationships) was measured at study entry. Results revealed significant associations between CRP and sexual satisfaction and partnered orgasms frequency for those most motivated to approach rewards with their partner. Interaction effects were replicated with relationship-focused psychological correlates of sexual well-being (e.g., touch, shared laughter, social support), but not with individual-focused outcomes (e.g., adapting to change, goal progress). This is one of the first human studies to demonstrate the body and mind coordinate to promote satisfying sexual experiences within romantic relationships.
许多关于炎症加剧和社会行为的证据都集中在社会退缩上。基于最近的理论(Muscatell 和 Inagaki,2021 年),我们转而关注性的社会从属体验。我们调查了 158 名处于恋爱关系中的人的免疫学和性动机之间的相互作用。我们在一个月内多次测量了以 C 反应蛋白(CRP)为指标的炎症和性幸福感。在研究开始时测量了关系接近动机(即在关系中获得回报的动机)。结果显示,对于那些最想与伴侣一起获得奖励的人来说,CRP 与性满意度和伴侣性高潮频率之间存在明显的关联。交互效应与以关系为中心的性健康心理相关因素(如抚摸、共享笑声、社会支持)相同,但与以个人为中心的结果(如适应变化、目标进展)不相同。这是第一批证明身体和心理协调促进浪漫关系中令人满意的性体验的人类研究之一。
{"title":"Do inflammation and relational motivation coordinate having better sex? The interplay between C-reactive protein and relational approach motivation on sexual well-being","authors":"","doi":"10.1016/j.bbi.2024.08.054","DOIUrl":"10.1016/j.bbi.2024.08.054","url":null,"abstract":"<div><p>Much evidence on heightened inflammation and social behavior focuses on social withdrawal. Building on recent theory (Muscatell and Inagaki, 2021), we focused instead on the socially affiliative experience of sex. We investigated the interplay between immunology and motivation on sexual well-being among 158 individuals in romantic relationships. Inflammation, indexed by C-reactive protein (CRP), and sexual well-being were measured multiple times over a month. Relational approach motivation (i.e., motivation toward rewards in relationships) was measured at study entry. Results revealed significant associations between CRP and sexual satisfaction and partnered orgasms frequency for those most motivated to approach rewards with their partner. Interaction effects were replicated with relationship-focused psychological correlates of sexual well-being (e.g., touch, shared laughter, social support), but not with individual-focused outcomes (e.g., adapting to change, goal progress). This is one of the first human studies to demonstrate the body and mind coordinate to promote satisfying sexual experiences within romantic relationships.</p></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142139310","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}