Gut-brain axis

肠 - 脑轴
  • 文章类型: Journal Article
    功能性胃肠病(FGIDs)是指一组具有慢性症状的疾病,比如腹痛,吞咽困难,消化不良,腹泻,便秘,和腹胀。其中,功能性便秘显着影响生活质量,并与合并症有关,如焦虑和抑郁。尽管广泛发生,但确切的病理生理学仍不清楚。研究表明,肠-脑轴在FGID中起作用。大脑和胃肠道(GI)之间的双向通信中断会导致胃肠道症状和情绪障碍。对FGID病理生理学的不完全理解导致治疗选择有限。传统治疗通常集中在单一症状上,并伴有副作用,提示需要解决GI和心理成分的替代方法。包括草药补充剂在内的替代方法通过促进规律性和肠道健康,为传统医学提供了天然的替代品。AbelmoschusesculentusL.或秋葵有在传统医学中使用的历史。在秋葵中发现的生物活性化合物如多糖和纤维提供胃保护益处。Withaniasomnifera是一种通常被称为ashwagandha的植物。植物根因其促进健康的作用而被使用。研究支持使用W.somnifera来帮助缓解压力和睡眠。Digexin是一种草药补充剂,结合了W.somnifera(ashwagandha)和A.esculentus(秋葵)。通过调节肠-脑轴,它已显示出改善胃肠道规律性和情绪的希望。临床研究支持一种有助于FGID管理的新型草药补充剂的潜力。这篇叙述性评论着眼于FGID,病因,目前的治疗,和可能的治疗补充剂,以帮助症状管理。
    Functional gastrointestinal disorders (FGIDs) refer to a group of disorders with chronic symptoms, such as abdominal pain, dysphagia, dyspepsia, diarrhea, constipation, and bloating. Among these, functional constipation significantly impacts the quality of life and is linked with comorbidities, such as anxiety and depression. The exact pathophysiology remains unclear despite the widespread occurrence. Research suggests that the gut-brain axis plays a role in FGIDs. Disruptions in the bidirectional communication between the brain and gastrointestinal (GI) tract contribute to GI symptoms and mood disturbances. The incomplete understanding of FGID pathophysiology has led to limited treatment options. Traditional treatments often focus on single symptoms and come with side effects, prompting the need for alternative approaches that address both GI and psychological components. Alternative approaches including herbal supplements offer a natural alternative to conventional medicine by promoting regularity and gut health. Abelmoschus esculentus L. or okra has a history of use in traditional medicine. Bioactive compounds such as polysaccharides and fibers found in okra offer gastroprotective benefits. Withania somnifera is a plant commonly referred to as ashwagandha. The plant root has been used for its health-promoting effects. Research supports the use of W. somnifera to help with stress and sleep. Digexin is a herbal supplement combining W. somnifera (ashwagandha) and A. esculentus (okra). It has shown promise in improving both GI regularity and mood by modulating the gut-brain axis. Clinical studies support the potential of a novel herbal supplement that aids in the management of FGIDs. This narrative review looks at FGIDs, etiologies, current treatment, and possible therapeutic supplements to aid in symptom management.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    最近对帕金森病(PD)的见解,进行性神经退行性疾病,提示肠道微生物组对其发病机理和通过肠-脑轴的进展有重大影响。本研究整合了16SrRNA测序,高通量转录组测序,和动物模型实验,以探索支持肠-脑轴在PD中的作用的分子机制,关注由SCFA受体FFAR2和FFAR3介导的短链脂肪酸(SCFA)。我们的发现强调了PD患者和健康个体之间肠道菌群组成的显著差异,特别是在类群中,如大肠杆菌志贺氏菌和拟杆菌,这可能通过次级代谢产物生物合成影响SCFA水平。值得注意的是,从健康的粪便微生物群移植(FMT)到1-甲基-4-苯基-1,2,3,6-四氢吡啶(MPTP)诱导的PD小鼠模型显着改善运动功能,纹状体中多巴胺和5-羟色胺水平增强,并增加黑质中多巴胺能神经元的数量,同时减少神经胶质细胞的活化。这种治疗效果与SCFA水平的增加有关,如乙酸盐,丙酸盐,MPTP损伤小鼠肠道中的丁酸盐。此外,转录组学分析显示MPTP损伤小鼠中FFAR2和FFAR3的表达上调,表明它们在介导FMT对中枢神经系统的益处中的关键作用。这些结果提供了令人信服的证据,表明肠道微生物群和SCFA在调节肠-脑轴中起着关键作用。为PD的病因和治疗干预的潜在目标提供了新的见解。
    Recent insights into Parkinson\'s disease (PD), a progressive neurodegenerative disorder, suggest a significant influence of the gut microbiome on its pathogenesis and progression through the gut-brain axis. This study integrates 16S rRNA sequencing, high-throughput transcriptomic sequencing, and animal model experiments to explore the molecular mechanisms underpinning the role of gut-brain axis in PD, with a focus on short-chain fatty acids (SCFAs) mediated by the SCFA receptors FFAR2 and FFAR3. Our findings highlighted prominent differences in the gut microbiota composition between PD patients and healthy individuals, particularly in taxa such as Escherichia_Shigella and Bacteroidetes, which potentially impact SCFA levels through secondary metabolite biosynthesis. Notably, fecal microbiota transplantation (FMT) from healthy to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse models significantly improved motor function, enhanced dopamine and serotonin levels in the striatum, and increased the number of dopaminergic neurons in the substantia nigra while reducing glial cell activation. This therapeutic effect was associated with increased levels of SCFAs such as acetate, propionate, and butyrate in the gut of MPTP-lesioned mice. Moreover, transcriptomic analyses revealed upregulated expression of FFAR2 and FFAR3 in MPTP-lesioned mice, indicating their crucial role in mediating the benefits of FMT on the central nervous system. These results provide compelling evidence that gut microbiota and SCFAs play a critical role in modulating the gut-brain axis, offering new insights into PD\'s etiology and potential targets for therapeutic intervention.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    抑郁症,是影响近2.8亿人的全球健康问题。它不仅给经济和医疗保健系统带来沉重负担,而且还表现出复杂的生理联系和后果。胍丁胺,一种推定的神经调质,主要来自有益的肠道微生物,特别是乳酸菌,已经成为心理健康的潜在治疗剂。微生物群-肠-脑轴通过周围神经系统参与抑郁症的发展,内分泌系统,和免疫系统,可能是胍丁胺作用的关键因素。因此,本研究旨在探讨胍丁胺在抗生素诱导的大鼠菌群失调和抑郁样行为中的潜在机制,专注于它对肠道-脑轴的调节。通过口服广谱抗生素组合的七天方案诱导与菌群失调相关的抑郁样行为,包含氨苄青霉素和甲硝唑,并通过微生物验证,生物化学,和行为改变。在第8天,抗生素治疗的大鼠表现出松散的粪便稠度,改变了粪便微生物群,强迫游泳测试中类似抑郁的行为。促炎细胞因子增强,而海马和前额叶皮质中的胍基胺和单胺水平降低。服用抗生素破坏了回肠中的紧密连接蛋白,影响肠道结构。单独口服胍丁胺或与益生菌联合使用可显著逆转抗生素诱导的菌群失调,恢复肠道微生物群和减轻抑郁样行为。这种干预还恢复了神经炎症标志物,增加胍基胺和单胺水平,并保持肠道完整性。该研究强调了内源性胍丁胺在广谱抗生素诱导的生态失调和相关的抑郁样行为中肠-脑轴的调节作用。
    Depression, is a global health concern affecting nearly 280 million individuals. It not only imposes a significant burden on economies and healthcare systems but also manifests complex physiological connections and consequences. Agmatine, a putative neuromodulator derived primarily from beneficial gut microbes specially Lactobacillus, has emerged as a potential therapeutic agent for mental health. The microbiota-gut-brain axis is involved in the development of depression through the peripheral nervous system, endocrine system, and immune system and may be a key factor in the effect of agmatine. Therefore, this study aimed to investigate the potential mechanism of agmatine in antibiotic-induced dysbiosis and depression-like behaviour in rats, focusing on its modulation of the gut-brain axis. Depression-like behavior associated with dysbiosis was induced through a seven-day regimen of the oral broad-spectrum antibiotic combination, comprising ampicillin and metronidazole and validated through microbial, biochemical, and behavioral alterations. On day 8, antibiotic-treated rats exhibited loose fecal consistency, altered fecal microbiota, and depression-like behavior in forced swim test. Pro-inflammatory cytokines were enhanced, while agmatine and monoamine levels decreased in the hippocampus and prefrontal cortex. Antibiotic administration disrupted tight junction proteins in the ileum, affecting gut architecture. Oral agmatine alone or combined with probiotics significantly reversed antibiotic-induced dysbiosis, restoring gut microbiota and mitigating depression-like behaviors. This intervention also restored neuro-inflammatory markers, increased agmatine and monoamine levels, and preserved gut integrity. The study highlights the regulatory role of endogenous agmatine in the gut-brain axis in broad-spectrum antibiotic induced dysbiosis and associated depression-like behaviour.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    越来越多的注意力被给予理解口服药物和肠道微生物群之间存在的双向关系。经常被忽视,然而,是药物赋形剂对肠道微生物群的影响。随后,在这项研究中,我们对比了难溶性化合物的两种常用制剂之间的药代动力学性能和肠道微生物群相互作用,即1)由聚(乙烯基吡咯烷酮)K-30稳定的无定形固体分散体(ASD),和2)由中链甘油酯和卵磷脂组成的脂质纳米乳液(LNE)。溶解性差的抗精神病药,Lurasidone,由于其限速溶出而与ASD和LNE一起配制,口服生物利用度差,和显著的食物效果。在模拟胃肠道环境的体外溶出研究中,ASD和LNE均显示出促进鲁拉西酮过饱和。这转化为大鼠口服药物动力学的深刻改善,ASD和LNE在lurasidone生物利用度方面发挥了相当的~2.5倍的改善,与纯药物相比。口服制剂对肠道微生物群具有对比作用,随着LNE耗尽微生物生态系统的丰富性和丰度,这通过α多样性(Chao1指数)和操作分类单位(OTU)的减少得到了证明。相比之下,ASD发挥了“肠道中性”效应,由此观察到α多样性和OTU的轻度富集。重要的是,这表明ASD是有效的溶解度增强制剂,可以在不包含肠道微生物群完整性的情况下使用-这是治疗精神健康障碍的不可或缺的考虑因素。比如精神分裂症,由于肠道菌群在调节情绪和认知方面的作用。
    Increasing attention is being afforded to understanding the bidirectional relationship that exists between oral drugs and the gut microbiota. Often overlooked, however, is the impact that pharmaceutical excipients exert on the gut microbiota. Subsequently, in this study, we contrasted the pharmacokinetic performance and gut microbiota interactions between two commonly employed formulations for poorly soluble compounds, namely 1) an amorphous solid dispersion (ASD) stabilised by poly(vinyl pyrrolidone) K-30, and 2) a lipid nanoemulsion (LNE) comprised of medium chain glycerides and lecithin. The poorly soluble antipsychotic, lurasidone, was formulated with ASD and LNE due to its rate-limiting dissolution, poor oral bioavailability, and significant food effect. Both the ASD and LNE were shown to facilitate lurasidone supersaturation within in vitro dissolution studies simulating the gastrointestinal environment. This translated into profound improvements in oral pharmacokinetics in rats, with the ASD and LNE exerting comparable ∼ 2.5-fold improvements in lurasidone bioavailability, compared to the pure drug. The oral formulations imparted contrasting effects on the gut microbiota, with the LNE depleting the richness and abundance of the microbial ecosystem, as evidenced through reductions in alpha diversity (Chao1 index) and operational taxonomical units (OTUs). In contrast, the ASD exerted a \'gut neutral\' effect, whereby a mild enrichment of alpha diversity and OTUs was observed. Importantly, this suggests that ASDs are effective solubility-enhancing formulations that can be used without comprising the integrity of the gut microbiota - an integral consideration in the treatment of mental health disorders, such as schizophrenia, due to the role of the gut microbiota in regulating mood and cognition.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    精神分裂症是一种严重的,以妄想为特征的慢性精神疾病,幻觉,认知障碍,和情绪失调。这种精神疾病通常对治疗有抵抗力。这篇文献综述旨在分析这种复杂的心理障碍与人体内发现的肠道微生物群之间的关系。大脑和肠道是相互联系的,新兴的研究表明,肠道菌群失调和精神分裂症之间存在联系。肠道菌群失调是指肠道微生物组的组成和功能的失衡或破坏。这项研究将精神分裂症患者的肠道微生物群与那些在门和属水平上没有显着差异的研究进行了比较,提供肠道微生物组改变的证据。精神分裂症患者中微生物群多样性的缺乏可以通过饮食干预来改变和改善为更健康的微生物组。针对肠脑轴的干预措施,如膳食益生菌或益生元,可能有助于缓解精神分裂症的某些症状,并有助于改善患者的健康。了解肠道微生物组健康与精神分裂症之间复杂的相互作用,可能有助于制定有针对性的干预措施,改变精神分裂症患者的肠道微生物组,反过来,减轻他们的症状,提高他们的生活质量。
    Schizophrenia is a severe, chronic psychiatric disorder characterized by delusions, hallucinations, cognitive impairments, and emotional dysregulation. This psychiatric illness is often resistant to treatment. This literature review aims to analyze the relationship between this complex psychological disorder and the gut microbiota found within the human body. The brain and gut are interconnected, and emerging research suggests a link between gut dysbiosis and schizophrenia. Gut dysbiosis refers to an imbalance or disruption in the composition and function of the gut microbiome. The studies comparing the gut microbiota of patients with schizophrenia to those without highlight significant differences at the phylum and genus levels, providing evidence of gut microbiome alteration. The lack of diversity of microbiota in schizophrenia patients can be altered and improved to a healthier microbiome by way of dietary intervention. Interventions that target the gut-brain axis, such as dietary probiotics or prebiotics, may help alleviate certain symptoms of schizophrenia and help improve patients\' well-being. Understanding the complex interplay between gut microbiome health and schizophrenia may allow for the development of targeted interventions that alter the gut microbiome of patients with schizophrenia and, in turn, mitigate their symptoms and improve their quality of life.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:肠-脑轴是通过神经元连接胃肠道和中枢神经系统的双向通信网络,荷尔蒙,和抗体信号通路。这种联系的核心是肠道健康,包括肠道微生物群的平衡和功能,这对精神和认知健康有重大影响。这项研究调查了成年人肠道健康与认知功能之间的关系,强调肠道微生物群影响大脑健康的机制。
    目的:为了研究肠道健康对成人认知能力的影响,重点关注肠道微生物群影响大脑健康的过程。
    方法:于2024年1月至2024年4月在伊斯兰堡进行了一项定量横断面研究,涉及140名成年参与者。使用全面的16项肠道健康问卷和认知自评估评定量表(C-SARS)收集数据。评估了这些量表的心理测量特性,并使用统计产品和服务解决方案(SPSS,v26;IBMSPSSStatisticsforWindows,Armonk,NY).分析和描述性统计,包括回归,卡方,独立样本t检验,以及平均值和标准偏差,被应用了。
    结果:研究发现,肠道健康与认知能力之间存在中度关联,特别是在记忆和处理速度方面(对于一般认知,R²=0.17,β=-1.9,p=0.12;对于记忆,R²=0.01,β=-0.98,p=0.02;对于处理速度,R²=0.03,β=-0.18,p=0.03)。性别和婚姻状况差异显著,男性的肠道健康评分优于女性(M=34.1,SD=3.2与M=31.2,SD=3.2,p=0.00),与已婚人士相比,单身人士表现出更好的认知表现(M=9.4,SD=5.4与M=6.5,SD=3.7,p=0.03)。
    结论:该研究强调了肠道健康和认知功能之间的显著关联,这表明肠道菌群组成可以影响认知表现。性别和婚姻状况差异强调了在肠-脑轴研究中需要考虑个体差异。未来的研究应该在更大的样本中复制这些发现,并探索针对肠道微生物群的干预措施以增强认知健康。
    BACKGROUND: The gut-brain axis is a bidirectional communication network linking the gastrointestinal tract and the central nervous system via neuronal, hormonal, and antibody signaling pathways. Central to this connection is gut health, encompassing the balance and functionality of gut microbiota, which significantly impacts on mental and cognitive health. This study investigates the association between gut health and cognitive functioning in adults, highlighting the mechanisms by which gut microbiota influence brain health.
    OBJECTIVE: To examine the effects of gut health on adult cognitive performance, with a focus on the processes by which gut microbiota impacts brain health.
    METHODS: A quantitative cross-sectional study was conducted in Islamabad from January 2024 to April 2024, involving 140 adult participants. Data were collected using a comprehensive 16-item gut health questionnaire and the cognition self-assessment rating scale (C-SARS). The psychometric properties of these scales were assessed, and the data were analyzed using Statistical Product and Service Solutions (SPSS, v26; IBM SPSS Statistics for Windows, Armonk, NY). Analytical and descriptive statistics, including regression, chi-square, independent sample t-tests, and mean and standard deviation, were applied.
    RESULTS: The study found moderate associations between gut health and cognitive performance, particularly in memory and processing speed (R² = 0.17, β = -1.9, p = 0.12 for general cognition; R² = 0.01, β = -0.98, p = 0.02 for memory; R² = 0.03, β = -0.18, p = 0.03 for processing speed). Gender and marital status differences were significant, with males exhibiting better gut health scores than females (M = 34.1, SD = 3.2 vs. M = 31.2, SD = 3.2, p = 0.00), and singles showing better cognitive performance compared to married individuals (M = 9.4, SD = 5.4 vs. M = 6.5, SD = 3.7, p = 0.03).
    CONCLUSIONS: The study highlights significant associations between gut health and cognitive functions, suggesting that gut microbiota composition can influence cognitive performance. Gender and marital status differences underscore the need to consider individual differences in gut-brain axis research. Future studies should replicate these findings in larger samples and explore gut microbiota-targeted interventions for cognitive health enhancement.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    阿尔茨海默病(AD)是一种以认知功能下降为特征的进行性退行性神经系统疾病,主要影响记忆和逻辑思维,归因于大脑中的淀粉样蛋白-β斑块和tau蛋白缠结,导致神经元丢失和脑萎缩.神经炎症,AD的标志,涉及小胶质细胞和星形胶质细胞的激活,以响应病理变化,可能加剧神经元损伤。肠-脑轴是胃肠道和中枢神经系统之间的双向通讯通路,对维持大脑健康至关重要。植物化学品,在植物中发现的具有抗氧化和抗炎特性的天然化合物,如类黄酮,姜黄素,白藜芦醇,还有槲皮素,已经成为这个轴的潜在调制器,提示对AD预防的影响。植物化学物质的摄入影响肠道微生物组成及其代谢产物,从而影响大脑中的神经炎症和氧化应激。食用富含植物化学物质的食物可以促进健康的肠道微生物群,促进抗炎和神经保护物质的生产。植物化学物质的早期饮食掺入提供了一种非侵入性策略,用于调节肠-脑轴并可能降低AD风险或延迟其发作。探索通过植物化学物质摄入靶向肠-脑轴的干预措施代表了开发针对AD引发和进展的预防或治疗策略的有希望的途径。
    Alzheimer\'s disease (AD) is a progressive degenerative neurological condition characterized by cognitive decline, primarily affecting memory and logical thinking, attributed to amyloid-β plaques and tau protein tangles in the brain, leading to neuronal loss and brain atrophy. Neuroinflammation, a hallmark of AD, involves the activation of microglia and astrocytes in response to pathological changes, potentially exacerbating neuronal damage. The gut-brain axis is a bidirectional communication pathway between the gastrointestinal and central nervous systems, crucial for maintaining brain health. Phytochemicals, natural compounds found in plants with antioxidant and anti-inflammatory properties, such as flavonoids, curcumin, resveratrol, and quercetin, have emerged as potential modulators of this axis, suggesting implications for AD prevention. Intake of phytochemicals influences the gut microbial composition and its metabolites, thereby impacting neuroinflammation and oxidative stress in the brain. Consumption of phytochemical-rich foods may promote a healthy gut microbiota, fostering the production of anti-inflammatory and neuroprotective substances. Early dietary incorporation of phytochemicals offers a non-invasive strategy for modulating the gut-brain axis and potentially reducing AD risk or delaying its onset. The exploration of interventions targeting the gut-brain axis through phytochemical intake represents a promising avenue for the development of preventive or therapeutic strategies against AD initiation and progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    胃肠道(GI),人体中最大的微生物种群,通过各种机制在整体健康中起着至关重要的作用。研究的最新进展揭示了由肠道微生物群及其微生物产物介导的肠-脑通讯在包括2型糖尿病和阿尔茨海默病(AD)在内的各种疾病中的潜在意义。AD是最常见的痴呆类型,大多数病例是散发性的,没有明确的病因。然而,多种因素与散发性AD的进展有关,可以将fi分类为不可修饰的(例如,遗传)和可变(例如2型糖尿病,饮食等。).本综述集中于主要参与者,特别是可改变的因素,如2型糖尿病(T2D)和饮食及其对健康大脑的微生物群-肠道-大脑(MGB)和大脑-肠道(BG)沟通和认知功能的影响,以及它们在阿尔茨海默病中的功能障碍。特别强调了饮食对肠道微生物群的影响的机理方面的阐明以及一些肠道微生物产物在T2D和AD病理学中的意义。例如,机械上,HFD通过驱动代谢物诱导肠道生态失调,进而导致肠道屏障完整性丧失,伴随结肠和系统性慢性低度炎症,与肥胖和T2D相关。HFD诱导的肥胖和T2D平行神经炎症,淀粉样β(Aβ)的沉积,最终导致认知障碍。该综述还提供了饮食对大脑-肠道和微生物群-肠道-大脑沟通的影响的新视角,转录因子作为一种常见的语言,可能促进肥胖糖尿病患者的肠道和大脑之间的相互作用。患认知障碍和AD的风险较高。还讨论了其他共性,例如酪氨酸激酶的表达和一方面维持肠道完整性的功能,以及大脑中迁移的小胶质细胞功能的吞噬作用。最后,还讨论了关键参与者的表征,这些参与者的未来研究可能会阐明阻碍AD进展的新潜在药理靶标。
    The gastrointestinal (GI) tract, home to the largest microbial population in the human body, plays a crucial role in overall health through various mechanisms. Recent advancements in research have revealed the potential implications of gut-brain and vice-versa communication mediated by gut-microbiota and their microbial products in various diseases including type-2 diabetes and Alzheimer\'s disease (AD). AD is the most common type of dementia where most of cases are sporadic with no clearly identified cause. However, multiple factors are implicated in the progression of sporadic AD which can be classified as non-modifiable (e.g., genetic) and modifiable (e.g. Type-2 diabetes, diet etc.). Present review focusses on key players particularly the modifiable factors such as Type-2 diabetes (T2D) and diet and their implications in microbiota-gut-brain (MGB) and brain-gut (BG) communication and cognitive functions of healthy brain and their dysfunction in Alzheimer\'s Disease. Special emphasis has been given on elucidation of the mechanistic aspects of the impact of diet on gut-microbiota and the implications of some of the gut-microbial products in T2D and AD pathology. For example, mechanistically, HFD induces gut dysbiosis with driven metabolites that in turn cause loss of integrity of intestinal barrier with concomitant colonic and systemic chronic low-grade inflammation, associated with obesity and T2D. HFD-induced obesity and T2D parallel neuroinflammation, deposition of Amyloid β (Aβ), and ultimately cognitive impairment. The review also provides a new perspective of the impact of diet on brain-gut and microbiota-gut-brain communication in terms of transcription factors as a commonly spoken language that may facilitates the interaction between gut and brain of obese diabetic patients who are at a higher risk of developing cognitive impairment and AD. Other commonality such as tyrosine kinase expression and functions maintaining intestinal integrity on one hand and the phagocytic clarence by migratory microglial functions in brain are also discussed. Lastly, the characterization of the key players future research that might shed lights on novel potential pharmacological target to impede AD progression are also discussed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    迷幻药已成为几种精神疾病的有希望的疗法。围绕其机制的假设围绕其对5-羟色胺2A受体的部分激动,导致神经可塑性增强和大脑连通性变化,这些变化是积极心态转变的基础。然而,这些说法没有认识到肠道微生物群,通过肠-脑轴作用,也可能在调节迷幻药对行为的积极影响中发挥作用。在这次审查中,我们提供了现有的证据,表明肠道微生物群的组成可能对迷幻药物有反应,反过来,迷幻药的作用可以通过微生物代谢来调节。我们讨论了在未来研究中应考虑微生物组的替代机制模型和方法。意识到微生物对迷幻作用的贡献有可能显着影响临床实践,例如,通过允许基于肠道微生物群异质性的个性化迷幻疗法。ETOCBLURB:利用它们与血清素的结构相似性,我们认为,迷幻药对大脑的影响部分是由肠道微生物群介导的。识别迷幻微生物相互作用可以促进精准医学的实施,通过将患者微生物组的异质性映射到对基于迷幻药的疗法的反应的变异性。
    Psychedelics have emerged as promising therapeutics for several psychiatric disorders. Hypotheses around their mechanisms have revolved around their partial agonism at the serotonin 2 A receptor, leading to enhanced neuroplasticity and brain connectivity changes that underlie positive mindset shifts. However, these accounts fail to recognise that the gut microbiota, acting via the gut-brain axis, may also have a role in mediating the positive effects of psychedelics on behaviour. In this review, we present existing evidence that the composition of the gut microbiota may be responsive to psychedelic drugs, and in turn, that the effect of psychedelics could be modulated by microbial metabolism. We discuss various alternative mechanistic models and emphasize the importance of incorporating hypotheses that address the contributions of the microbiome in future research. Awareness of the microbial contribution to psychedelic action has the potential to significantly shape clinical practice, for example, by allowing personalised psychedelic therapies based on the heterogeneity of the gut microbiota.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:代谢综合征(MetS)在精神分裂症(SZ)患者中非常普遍,导致过早死亡等负面后果。肠道菌群失调,指的是微生物群的不平衡,慢性炎症与SZ和MetS有关。然而,肠道菌群失调之间的关系,宿主免疫功能障碍,SZ与MetS共病(SZ-MetS)尚不清楚。本研究旨在探讨SZ-MetS中肠道菌群的变化及其与免疫功能障碍的相关性。为其发病机制提供了新的见解。
    结果:我们从浙江招募了114名中国SZ-MetS患者和111名年龄匹配的健康对照,中国,使用针对16SrRNA基因V3-V4高变区的IlluminaMiSeq测序研究粪便微生物群。使用Bio-PlexPro人类细胞因子27-Plex测定来评估宿主免疫应答以检查细胞因子谱。在SZ-MetS,我们观察到细菌α-多样性下降和β-多样性显著差异。LEfSe分析确定了富集的产乙酸属(Megamonas和乳杆菌),和减少产生丁酸的细菌(亚杜里颗粒,和粪杆菌)在SZ-MetS中。这些改变的属与体重指数相关,症状的严重程度(通过阳性症状评估量表和阴性症状评估量表衡量),和甘油三酯水平。改变与脂多糖生物合成相关的细菌代谢途径,脂质代谢,和各种氨基酸代谢也被发现。此外,SZ-MetS表现出免疫功能障碍,促炎细胞因子增加,与微分属相关。
    结论:这些发现表明,肠道菌群失调和免疫功能障碍在SZ-MetS的发育中起着至关重要的作用。突出了针对肠道微生物群的潜在治疗方法。虽然这些疗法显示出希望,在临床实施前,需要进一步的机制研究来充分了解其有效性和安全性.
    BACKGROUND: Metabolic syndrome (MetS) is highly prevalent in individuals with schizophrenia (SZ), leading to negative consequences like premature mortality. Gut dysbiosis, which refers to an imbalance of the microbiota, and chronic inflammation are associated with both SZ and MetS. However, the relationship between gut dysbiosis, host immunological dysfunction, and SZ comorbid with MetS (SZ-MetS) remains unclear. This study aims to explore alterations in gut microbiota and their correlation with immune dysfunction in SZ-MetS, offering new insights into its pathogenesis.
    RESULTS: We enrolled 114 Chinese patients with SZ-MetS and 111 age-matched healthy controls from Zhejiang, China, to investigate fecal microbiota using Illumina MiSeq sequencing targeting 16 S rRNA gene V3-V4 hypervariable regions. Host immune responses were assessed using the Bio-Plex Pro Human Cytokine 27-Plex Assay to examine cytokine profiles. In SZ-MetS, we observed decreased bacterial α-diversity and significant differences in β-diversity. LEfSe analysis identified enriched acetate-producing genera (Megamonas and Lactobacillus), and decreased butyrate-producing bacteria (Subdoligranulum, and Faecalibacterium) in SZ-MetS. These altered genera correlated with body mass index, the severity of symptoms (as measured by the Scale for Assessment of Positive Symptoms and Scale for Assessment of Negative Symptoms), and triglyceride levels. Altered bacterial metabolic pathways related to lipopolysaccharide biosynthesis, lipid metabolism, and various amino acid metabolism were also found. Additionally, SZ-MetS exhibited immunological dysfunction with increased pro-inflammatory cytokines, which correlated with the differential genera.
    CONCLUSIONS: These findings suggested that gut microbiota dysbiosis and immune dysfunction play a vital role in SZ-MetS development, highlighting potential therapeutic approaches targeting the gut microbiota. While these therapies show promise, further mechanistic studies are needed to fully understand their efficacy and safety before clinical implementation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号