Pub Date : 2024-11-01DOI: 10.1016/j.bbi.2024.10.031
Igor Rafael Correia Rocha, Maggie R Finch, Jayson B Ball, Michael E Harland, Madison Clements, Suzanne Green-Fulgham, Guiyun Song, Yi Liu, Daniel Banov, Linda R Watkins
Psoriasis is a chronic immune-mediated skin disorder characterized by intense local inflammation, epidermal hyperplasia, and leukocyte infiltration. Current treatment approaches for psoriasis aim to alleviate symptoms and prevent disease progression, including systemically administered drugs with whole body side effects. Despite some advances in psoriasis treatment, success has been quite limited. To begin to address this challenge, we undertook an initial investigation of whether transcutaneous delivery of an endogenous anti-inflammatory cytokine could provide an effective, local treatment of psoriatic-like skin conditions. To do this, we utilized a previously documented rodent model of psoriasis, induced via a single topical application of Imiquimod (IMQ) to the shaved back of rats. The therapeutic approach used for this initial investigation was delivery of plasmid DNA encoding rat interleukin-10 (pDNA-rIL10), a non-viral gene therapy approach previously shown to be effective in suppressing neuroinflammatory disorders after localized delivery either intracerebrally or intrathecally. Translation of this CNS therapeutic for use in psoriatic-like skin disorders required reformulation to enable transcutaneous delivery. Toward that end, pDNA-rIL10 was topically applied in Lipoderm HMW, a base explicitly designed to deliver higher molecular weight compounds into skin. Here we show that a single topical application of pDNA-rIL10 in Lipoderm HMW was effective in decreasing mRNA levels of pro-inflammatory cytokines as well as reducing the recruitment of T-cells to IMQ-treated skin. Furthermore, this transcutaneous IL-10 gene therapy decreased signs of skin inflammation, reflected by reduced erythema. Moreover, the results provide an initial indication that IL10 may stimulate hair regrowth in psoriatic-like skin.
银屑病是一种由免疫介导的慢性皮肤病,以局部强烈发炎、表皮增生和白细胞浸润为特征。目前治疗银屑病的方法旨在缓解症状和防止病情恶化,包括全身用药,但副作用较大。尽管银屑病治疗取得了一些进展,但取得的成功仍然十分有限。为了开始应对这一挑战,我们对经皮给药内源性抗炎细胞因子能否有效治疗银屑病样皮肤病进行了初步研究。为此,我们利用了以前记录的啮齿动物银屑病模型,通过在剃光毛发的大鼠背部单次局部应用咪喹莫特(IMQ)来诱导银屑病。这项初步研究采用的治疗方法是递送编码大鼠白细胞介素-10(pDNA-rIL10)的 DNA 质粒,这种非病毒基因治疗方法以前曾被证明在脑内或经皮内局部递送后能有效抑制神经炎性疾病。要将这种中枢神经系统疗法应用于银屑病样皮肤病,需要重新配制,以实现经皮给药。为此,pDNA-rIL10 在 Lipoderm HMW(一种专门用于向皮肤输送高分子量化合物的基质)中进行了局部应用。我们在这里展示了在 Lipoderm HMW 中局部施用 pDNA-rIL10 能有效降低促炎细胞因子的 mRNA 水平,并减少 T 细胞对 IMQ 处理过的皮肤的招募。此外,这种经皮 IL-10 基因疗法还能减少皮肤炎症症状,红斑的减少就反映了这一点。此外,研究结果还初步表明,IL10 可刺激银屑病样皮肤的毛发再生。
{"title":"An initial investigation of transcutaneous delivery of plasmid DNA encoding interleukin-10 for the treatment of psoriatic skin conditions.","authors":"Igor Rafael Correia Rocha, Maggie R Finch, Jayson B Ball, Michael E Harland, Madison Clements, Suzanne Green-Fulgham, Guiyun Song, Yi Liu, Daniel Banov, Linda R Watkins","doi":"10.1016/j.bbi.2024.10.031","DOIUrl":"10.1016/j.bbi.2024.10.031","url":null,"abstract":"<p><p>Psoriasis is a chronic immune-mediated skin disorder characterized by intense local inflammation, epidermal hyperplasia, and leukocyte infiltration. Current treatment approaches for psoriasis aim to alleviate symptoms and prevent disease progression, including systemically administered drugs with whole body side effects. Despite some advances in psoriasis treatment, success has been quite limited. To begin to address this challenge, we undertook an initial investigation of whether transcutaneous delivery of an endogenous anti-inflammatory cytokine could provide an effective, local treatment of psoriatic-like skin conditions. To do this, we utilized a previously documented rodent model of psoriasis, induced via a single topical application of Imiquimod (IMQ) to the shaved back of rats. The therapeutic approach used for this initial investigation was delivery of plasmid DNA encoding rat interleukin-10 (pDNA-rIL10), a non-viral gene therapy approach previously shown to be effective in suppressing neuroinflammatory disorders after localized delivery either intracerebrally or intrathecally. Translation of this CNS therapeutic for use in psoriatic-like skin disorders required reformulation to enable transcutaneous delivery. Toward that end, pDNA-rIL10 was topically applied in Lipoderm HMW, a base explicitly designed to deliver higher molecular weight compounds into skin. Here we show that a single topical application of pDNA-rIL10 in Lipoderm HMW was effective in decreasing mRNA levels of pro-inflammatory cytokines as well as reducing the recruitment of T-cells to IMQ-treated skin. Furthermore, this transcutaneous IL-10 gene therapy decreased signs of skin inflammation, reflected by reduced erythema. Moreover, the results provide an initial indication that IL10 may stimulate hair regrowth in psoriatic-like skin.</p>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142567249","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 : 2024-10-30DOI: 10.1016/j.bbi.2024.10.037
{"title":"Evaluating Methodological validity in the Analysis of Tai Chi and cognitive behavioral therapy for inflammation Reduction in Insomnia: A Letter to the Editor","authors":"","doi":"10.1016/j.bbi.2024.10.037","DOIUrl":"10.1016/j.bbi.2024.10.037","url":null,"abstract":"","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142557204","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 : 2024-10-30DOI: 10.1016/j.bbi.2024.10.038
{"title":"In response to Eto-Kimura et al.","authors":"","doi":"10.1016/j.bbi.2024.10.038","DOIUrl":"https://doi.org/10.1016/j.bbi.2024.10.038","url":null,"abstract":"","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142563804","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 : 2024-10-29DOI: 10.1016/j.bbi.2024.10.035
A L Dawson, A A Willette
{"title":"Serology and Alzheimer's disease: Is infectious disease in the driver's seat?","authors":"A L Dawson, A A Willette","doi":"10.1016/j.bbi.2024.10.035","DOIUrl":"10.1016/j.bbi.2024.10.035","url":null,"abstract":"","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142557206","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 : 2024-10-29DOI: 10.1016/j.bbi.2024.10.032
Urvinder Kaur Sardarni, Anoop T Ambikan, Arpan Acharya, Samuel D Johnson, Sean N Avedissian, Ákos Végvári, Ujjwal Neogi, Siddappa N Byrareddy
Despite significant effort, a clear understanding of host tissue-specific responses and their implications for immunopathogenicity against the severe acute respiratory syndrome coronavirus2 (SARS-CoV-2) variant infection has remained poorly defined. To shed light on the interaction between tissues and SARS-CoV-2 variants, we sought to characterize the complex relationship among acute multisystem manifestations, dysbiosis of the gut microbiota, and the resulting implications for SARS-CoV-2 variant-specific immunopathogenesis in the Golden Syrian Hamster (GSH) model using multi-omics approaches. Our investigation revealed the presence of increased SARS-CoV-2 genomic RNA in diverse tissues of delta-infected GSH compared to the omicron variant. Multi-omics analyses uncovered distinctive metabolic responses between the delta and omicron variants, with the former demonstrating dysregulation in synaptic transmission proteins associated with neurocognitive disorders. Additionally, delta-infected GSH exhibited an altered fecal microbiota composition, marked by increased inflammation-associated taxa and reduced commensal bacteria compared to the omicron variant. These findings underscore the SARS-CoV-2-mediated tissue insult, characterized by modified host metabolites, neurological protein dysregulation, and gut dysbiosis, highlighting the compromised gut-lung-brain axis during acute infection.
{"title":"SARS-CoV-2 variants mediated tissue-specific metabolic reprogramming determines the disease pathophysiology in a hamster model.","authors":"Urvinder Kaur Sardarni, Anoop T Ambikan, Arpan Acharya, Samuel D Johnson, Sean N Avedissian, Ákos Végvári, Ujjwal Neogi, Siddappa N Byrareddy","doi":"10.1016/j.bbi.2024.10.032","DOIUrl":"10.1016/j.bbi.2024.10.032","url":null,"abstract":"<p><p>Despite significant effort, a clear understanding of host tissue-specific responses and their implications for immunopathogenicity against the severe acute respiratory syndrome coronavirus2 (SARS-CoV-2) variant infection has remained poorly defined. To shed light on the interaction between tissues and SARS-CoV-2 variants, we sought to characterize the complex relationship among acute multisystem manifestations, dysbiosis of the gut microbiota, and the resulting implications for SARS-CoV-2 variant-specific immunopathogenesis in the Golden Syrian Hamster (GSH) model using multi-omics approaches. Our investigation revealed the presence of increased SARS-CoV-2 genomic RNA in diverse tissues of delta-infected GSH compared to the omicron variant. Multi-omics analyses uncovered distinctive metabolic responses between the delta and omicron variants, with the former demonstrating dysregulation in synaptic transmission proteins associated with neurocognitive disorders. Additionally, delta-infected GSH exhibited an altered fecal microbiota composition, marked by increased inflammation-associated taxa and reduced commensal bacteria compared to the omicron variant. These findings underscore the SARS-CoV-2-mediated tissue insult, characterized by modified host metabolites, neurological protein dysregulation, and gut dysbiosis, highlighting the compromised gut-lung-brain axis during acute infection.</p>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142557205","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 : 2024-10-29DOI: 10.1016/j.bbi.2024.10.039
Immune dysregulation in the brain and periphery is thought to contribute to the detrimental neurodegeneration that occurs in Parkinson’s disease (PD). Identifying mechanisms to reverse this dysregulation is key to developing disease-altering therapeutics for this currently incurable disease. Here we utilized the longitudinal data from the Parkinson’s Progression Marker Initiative to demonstrate that circulating lymphocytes progressively decline in PD and can be used to predict future motor symptom progression. Deep brain stimulation (DBS), which is used as a symptomatic treatment, could halt this progressive decline. By analyzing specific immune populations from a second cohort of patients, we could show that DBS causes a shift from the pro-inflammatory CD4+ T helper 17 cells driving neurodegeneration to anti-inflammatory CD4+ regulatory T cells. RNA-sequencing and immunohistochemistry in the brain of the A53T alpha-synuclein rat model of PD revealed that DBS also decreases neuroinflammation. These data suggest a potential disease-altering role for DBS by halting inflammatory processes.
大脑和外周的免疫失调被认为是导致帕金森病(PD)发生有害神经变性的原因之一。确定逆转这种失调的机制是为这种目前无法治愈的疾病开发改变疾病疗法的关键。在这里,我们利用帕金森病进展标志物倡议(Parkinson's Progression Marker Initiative)的纵向数据证明,循环淋巴细胞在帕金森病中会逐渐减少,并可用于预测未来运动症状的进展。作为对症治疗的脑深部刺激(DBS)可以阻止这种进行性下降。通过分析第二批患者的特定免疫群体,我们可以证明 DBS 会导致驱动神经退行性病变的促炎性 CD4+ T 辅助 17 细胞向抗炎性 CD4+ 调节性 T 细胞转变。A53Tα-突触核蛋白帕金森病大鼠模型脑部的RNA测序和免疫组化显示,DBS还能减少神经炎症。这些数据表明,通过阻止炎症过程,DBS 有可能起到改变疾病的作用。
{"title":"Deep brain stimulation halts Parkinson’s disease-related immune dysregulation in the brain and peripheral blood","authors":"","doi":"10.1016/j.bbi.2024.10.039","DOIUrl":"10.1016/j.bbi.2024.10.039","url":null,"abstract":"<div><div>Immune dysregulation in the brain and periphery is thought to contribute to the detrimental neurodegeneration that occurs in Parkinson’s disease (PD). Identifying mechanisms to reverse this dysregulation is key to developing disease-altering therapeutics for this currently incurable disease. Here we utilized the longitudinal data from the Parkinson’s Progression Marker Initiative to demonstrate that circulating lymphocytes progressively decline in PD and can be used to predict future motor symptom progression. Deep brain stimulation (DBS), which is used as a symptomatic treatment, could halt this progressive decline. By analyzing specific immune populations from a second cohort of patients, we could show that DBS causes a shift from the pro-inflammatory CD4<sup>+</sup> T helper 17 cells driving neurodegeneration to anti-inflammatory CD4<sup>+</sup> regulatory T cells. RNA-sequencing and immunohistochemistry in the brain of the A53T alpha-synuclein rat model of PD revealed that DBS also decreases neuroinflammation. These data suggest a potential disease-altering role for DBS by halting inflammatory processes.</div></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142557203","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 : 2024-10-28DOI: 10.1016/j.bbi.2024.10.040
{"title":"mAbsolutely FABulous: From a case of mistaken identity to pinpoint precision in the antibodies formerly known as ‘VGKC’","authors":"","doi":"10.1016/j.bbi.2024.10.040","DOIUrl":"10.1016/j.bbi.2024.10.040","url":null,"abstract":"","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142543763","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 : 2024-10-28DOI: 10.1016/j.bbi.2024.10.034
Background
Few studies have integrated the impact of individual and cumulative childhood maltreatment on multiple psychiatric symptoms, with the mechanisms underlying these associations largely unknown. This study aims to comprehensively assess the associations between childhood maltreatment, multiple peripheral immune biomarkers, and various psychiatric symptoms in adulthood and to explore whether peripheral immune inflammation plays a mediator role in the associations between childhood maltreatment and psychiatric symptoms in adulthood.
Methods
Using data from the UK Biobank, we constructed a retrospective cohort study of 138,915 participants who provided self-reported childhood maltreatment and had peripheral immune biomarkers assessed. We examined seven types of psychiatric symptoms in adulthood, including depressive symptoms, anxiety symptoms, mania, post-traumatic stress disorder (PTSD), psychotic experiences, self-harm, and alcohol use disorder. Logistic regression models were performed to explore the associations between childhood maltreatment, immune biomarkers, and psychiatric symptoms, calculating the average marginal effects for each indicator of childhood maltreatment. Mediation analyses were conducted to determine the extent to which the immune biomarkers could explain the association between childhood maltreatment and psychiatric symptoms in adulthood. Subgroup and sensitivity analyses were also performed.
Results
Among the participants, 77,937 (56.10 %) were female, with a mean age of 55.91 (SD: 7.73) years at baseline. There were dose–response relationships existed between the accumulation of childhood maltreatment indicators and all seven assessed psychiatric symptoms and multimorbidity in adulthood (e.g., for depressive symptoms, OR = 1.67 [95 %CI, 1.57 to 1.78] for one childhood maltreatment indicator; OR = 2.77 [95 % CI, 2.58 to 2.97] for two; OR = 4.91 [95 % CI, 4.61 to 5.24] for three or more). Emotional abuse and physical neglect showed the strongest average marginal effects on psychiatric symptoms. Levels of C-reactive protein (CRP) and counts of leukocytes and neutrophils were positively associated with depressive symptoms (e.g., OR = 1.13 [95 % CI, 1.08 to 1.17] for CRP level), anxiety symptoms, PTSD, and psychotic experiences. Moreover, levels of CRP partially mediated the association between childhood maltreatment scores and psychiatric symptoms, albeit with a relatively low mediation proportion (0.65 %-1.77 %).
Conclusions
Our findings underscore the importance of interventions that address multiple forms of childhood maltreatment to mitigate long-term mental health challenges substantially. While peripheral immunity responses may serve as predictors of mental health problems, they might not to be the primary mechanism through which childhood maltreatment influences psychiatric symptoms in adulthood.
背景:很少有研究综合了个体和累积的童年虐待对多种精神症状的影响,这些关联的内在机制在很大程度上也是未知的。本研究旨在全面评估童年虐待、多种外周免疫生物标志物与成年后各种精神症状之间的关联,并探讨外周免疫炎症是否在童年虐待与成年后精神症状之间的关联中起中介作用:我们利用英国生物库的数据构建了一项回顾性队列研究,研究对象为138,915名自我报告童年遭受虐待并接受外周免疫生物标志物评估的参与者。我们研究了成年后的七种精神症状,包括抑郁症状、焦虑症状、躁狂症、创伤后应激障碍(PTSD)、精神病性体验、自残和酒精使用障碍。为探讨童年虐待、免疫生物标志物和精神症状之间的关联,我们建立了逻辑回归模型,计算了每个童年虐待指标的平均边际效应。我们还进行了中介分析,以确定免疫生物标志物在多大程度上可以解释童年虐待与成年后精神症状之间的关联。此外还进行了分组分析和敏感性分析:参与者中有 77,937 人(56.10%)为女性,基线平均年龄为 55.91 岁(标准差:7.73)。童年虐待指标的累积与所有七项评估的精神症状以及成年后的多病症之间存在剂量-反应关系(例如,就抑郁症状而言,一个童年虐待指标的OR = 1.67 [95 % CI, 1.57 to 1.78];两个指标的OR = 2.77 [95 % CI, 2.58 to 2.97];三个或更多指标的OR = 4.91 [95 % CI, 4.61 to 5.24])。精神虐待和身体忽视对精神症状的平均边际效应最强。C反应蛋白(CRP)水平以及白细胞和中性粒细胞计数与抑郁症状(例如,CRP水平的OR = 1.13 [95 % CI, 1.08 to 1.17])、焦虑症状、创伤后应激障碍和精神病经历呈正相关。此外,CRP水平对童年虐待评分与精神症状之间的关联起到了部分中介作用,尽管中介比例相对较低(0.65%-1.77%):我们的研究结果凸显了针对多种形式的儿童虐待进行干预的重要性,以大幅减轻长期的心理健康挑战。虽然外周免疫反应可能是心理健康问题的预测因素,但它们可能并不是童年虐待影响成年后精神症状的主要机制。
{"title":"Associations between childhood maltreatment, peripheral immune biomarkers, and psychiatric symptoms in adults: A cohort study of over 138,000 participants","authors":"","doi":"10.1016/j.bbi.2024.10.034","DOIUrl":"10.1016/j.bbi.2024.10.034","url":null,"abstract":"<div><h3>Background</h3><div>Few studies have integrated the impact of individual and cumulative childhood maltreatment on multiple psychiatric symptoms, with the mechanisms underlying these associations largely unknown. This study aims to comprehensively assess the associations between childhood maltreatment, multiple peripheral immune biomarkers, and various psychiatric symptoms in adulthood and to explore whether peripheral immune inflammation plays a mediator role in the associations between childhood maltreatment and psychiatric symptoms in adulthood.</div></div><div><h3>Methods</h3><div>Using data from the UK Biobank, we constructed a retrospective cohort study of 138,915 participants who provided self-reported childhood maltreatment and had peripheral immune biomarkers assessed. We examined seven types of psychiatric symptoms in adulthood, including depressive symptoms, anxiety symptoms, mania, post-traumatic stress disorder (PTSD), psychotic experiences, self-harm, and alcohol use disorder. Logistic regression models were performed to explore the associations between childhood maltreatment, immune biomarkers, and psychiatric symptoms, calculating the average marginal effects for each indicator of childhood maltreatment. Mediation analyses were conducted to determine the extent to which the immune biomarkers could explain the association between childhood maltreatment and psychiatric symptoms in adulthood. Subgroup and sensitivity analyses were also performed.</div></div><div><h3>Results</h3><div>Among the participants, 77,937 (56.10 %) were female, with a mean age of 55.91 (SD: 7.73) years at baseline. There were dose–response relationships existed between the accumulation of childhood maltreatment indicators and all seven assessed psychiatric symptoms and multimorbidity in adulthood (e.g., for depressive symptoms, OR = 1.67 [95 %CI, 1.57 to 1.78] for one childhood maltreatment indicator; OR = 2.77 [95 % CI, 2.58 to 2.97] for two; OR = 4.91 [95 % CI, 4.61 to 5.24] for three or more). Emotional abuse and physical neglect showed the strongest average marginal effects on psychiatric symptoms. Levels of C-reactive protein (CRP) and counts of leukocytes and neutrophils were positively associated with depressive symptoms (e.g., OR = 1.13 [95 % CI, 1.08 to 1.17] for CRP level), anxiety symptoms, PTSD, and psychotic experiences. Moreover, levels of CRP partially mediated the association between childhood maltreatment scores and psychiatric symptoms, albeit with a relatively low mediation proportion (0.65 %-1.77 %).</div></div><div><h3>Conclusions</h3><div>Our findings underscore the importance of interventions that address multiple forms of childhood maltreatment to mitigate long-term mental health challenges substantially. While peripheral immunity responses may serve as predictors of mental health problems, they might not to be the primary mechanism through which childhood maltreatment influences psychiatric symptoms in adulthood.</div></div>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142543762","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 : 2024-10-28DOI: 10.1016/j.bbi.2024.10.033
Sarah R Vreijling, Brenda W J H Penninx, Josine E Verhoeven, Charlotte E Teunissen, Elena R Blujdea, Aartjan T F Beekman, Femke Lamers, Rick Jansen
Background: Exercise promotes immunometabolic health and is increasingly recognized as an effective depression treatment. Exercise may be beneficial for patients with immunometabolic depression (IMD), who experience inflammatory and metabolic dysregulations and may respond less to antidepressants. This secondary analysis of the MOTAR study compared the effects of running therapy and antidepressants on IMD features among patients with depression and/or anxiety disorder. We additionally assessed whether baseline IMD moderated intervention effects on depression.
Methods: Participants received 16 weeks of group-based running therapy (N = 96) or escitalopram/sertraline (N = 45) in a partially randomized patient preference design. IMD features included atypical, energy-related symptom (AES) severity, inflammation index (CRP, IFN-γ, IL-6, TNF-α), metabolic syndrome index, three metabolite principle components (PC) (derived from 73 metabolites) and a composite IMD index.
Results: Interventions differed in changes in the metabolic syndrome index (d = 0.59, p = 0.026) and IMD index (d = 0.85, p < 0.001). While running therapy decreased both outcomes, the antidepressant group showed an increased IMD index. Although groups did not differ statistically significant in changes in AES severity, inflammation index, and metabolite PC1, results indicated a consistent trend towards greater improvement with running therapy across these outcomes as well (d = 0.38 to 0.52). Baseline IMD did not moderate intervention effects on depression outcomes.
Conclusions: This study suggests that exercise more effectively targets the IMD dimension than antidepressants. Patients with IMD did not benefit more from running therapy than antidepressants in terms of reductions in depression. Exercise should be considered an alternative or complementary treatment to particularly reduce IMD features in depressed patients.
Trial registration: Trialregister.nl Number of identification: NTR3460.
{"title":"Running therapy or antidepressants as treatments for immunometabolic depression in patients with depressive and anxiety disorders: A secondary analysis of the MOTAR study.","authors":"Sarah R Vreijling, Brenda W J H Penninx, Josine E Verhoeven, Charlotte E Teunissen, Elena R Blujdea, Aartjan T F Beekman, Femke Lamers, Rick Jansen","doi":"10.1016/j.bbi.2024.10.033","DOIUrl":"10.1016/j.bbi.2024.10.033","url":null,"abstract":"<p><strong>Background: </strong>Exercise promotes immunometabolic health and is increasingly recognized as an effective depression treatment. Exercise may be beneficial for patients with immunometabolic depression (IMD), who experience inflammatory and metabolic dysregulations and may respond less to antidepressants. This secondary analysis of the MOTAR study compared the effects of running therapy and antidepressants on IMD features among patients with depression and/or anxiety disorder. We additionally assessed whether baseline IMD moderated intervention effects on depression.</p><p><strong>Methods: </strong>Participants received 16 weeks of group-based running therapy (N = 96) or escitalopram/sertraline (N = 45) in a partially randomized patient preference design. IMD features included atypical, energy-related symptom (AES) severity, inflammation index (CRP, IFN-γ, IL-6, TNF-α), metabolic syndrome index, three metabolite principle components (PC) (derived from 73 metabolites) and a composite IMD index.</p><p><strong>Results: </strong>Interventions differed in changes in the metabolic syndrome index (d = 0.59, p = 0.026) and IMD index (d = 0.85, p < 0.001). While running therapy decreased both outcomes, the antidepressant group showed an increased IMD index. Although groups did not differ statistically significant in changes in AES severity, inflammation index, and metabolite PC1, results indicated a consistent trend towards greater improvement with running therapy across these outcomes as well (d = 0.38 to 0.52). Baseline IMD did not moderate intervention effects on depression outcomes.</p><p><strong>Conclusions: </strong>This study suggests that exercise more effectively targets the IMD dimension than antidepressants. Patients with IMD did not benefit more from running therapy than antidepressants in terms of reductions in depression. Exercise should be considered an alternative or complementary treatment to particularly reduce IMD features in depressed patients.</p><p><strong>Trial registration: </strong>Trialregister.nl Number of identification: NTR3460.</p>","PeriodicalId":9199,"journal":{"name":"Brain, Behavior, and Immunity","volume":null,"pages":null},"PeriodicalIF":8.8,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142543764","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}