Gut-brain axis

肠 - 脑轴
  • 文章类型: Journal Article
    失眠在中年人和老年人中作为一种普遍的睡眠障碍持续存在,显著影响生活质量和增加对年龄相关疾病的易感性。它分为客观失眠(O-IN)和矛盾失眠(P-IN),其中主观和客观的睡眠评估分歧。两个患者组的当前治疗方案产生不令人满意的结果。因此,研究P-IN和O-IN之间的神经生理学差异对于设计与主要预测相一致的新型精确干预措施至关重要,有针对性的预防,和个性化医疗(PPPM)原则。工作假设和方法论。鉴于肠道微生物群(GM)通过肠-脑轴对睡眠生理的影响,我们的研究重点是表征96名意大利绝经后妇女的GM概况,包括54例失眠患者(18个O-IN和36个P-IN)和42例对照,通过16SrRNA扩增子测序。与一般病史和临床病史的关系进行了探讨,睡眠模式,压力,血液生化参数,和营养模式。
    在O-IN和P-IN患者之间公布了独特的GM谱。O-IN患者在科氏杆菌科表现突出,包括Collinsella和Adlercreutzia,以及Erypelotricaceae,梭菌属,和片球菌.相反,P-IN患者主要通过拟杆菌区分,葡萄球菌,肉杆菌,假单胞菌,和各自的家庭,还有Odoribacter.
    这些发现为微生物群介导的O-IN与P-IN发病机制提供了有价值的见解。因此,GM概况可以作为定制的分层标准,能够识别有特定失眠亚型风险的女性,并促进基于微生物群的综合预测诊断的发展,有针对性的预防,和个性化治疗,最终提高临床疗效。
    在线版本包含补充材料,可在10.1007/s13167-024-00369-1获得。
    UNASSIGNED: Insomnia persists as a prevalent sleep disorder among middle-aged and older adults, significantly impacting quality of life and increasing susceptibility to age-related diseases. It is classified into objective insomnia (O-IN) and paradoxical insomnia (P-IN), where subjective and objective sleep assessments diverge. Current treatment regimens for both patient groups yield unsatisfactory outcomes. Consequently, investigating the neurophysiological distinctions between P-IN and O-IN is imperative for devising novel precision interventions aligned with primary prediction, targeted prevention, and personalized medicine (PPPM) principles.Working hypothesis and methodology.Given the emerging influence of gut microbiota (GM) on sleep physiology via the gut-brain axis, our study focused on characterizing the GM profiles of a well-characterized cohort of 96 Italian postmenopausal women, comprising 54 insomniac patients (18 O-IN and 36 P-IN) and 42 controls, through 16S rRNA amplicon sequencing. Associations were explored with general and clinical history, sleep patterns, stress, hematobiochemical parameters, and nutritional patterns.
    UNASSIGNED: Distinctive GM profiles were unveiled between O-IN and P-IN patients. O-IN patients exhibited prominence in the Coriobacteriaceae family, including Collinsella and Adlercreutzia, along with Erysipelotrichaceae, Clostridium, and Pediococcus. Conversely, P-IN patients were mainly discriminated by Bacteroides, Staphylococcus, Carnobacterium, Pseudomonas, and respective families, along with Odoribacter.
    UNASSIGNED: These findings provide valuable insights into the microbiota-mediated mechanism of O-IN versus P-IN onset. GM profiling may thus serve as a tailored stratification criterion, enabling the identification of women at risk for specific insomnia subtypes and facilitating the development of integrated microbiota-based predictive diagnostics, targeted prevention, and personalized therapies, ultimately enhancing clinical effectiveness.
    UNASSIGNED: The online version contains supplementary material available at 10.1007/s13167-024-00369-1.
    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
    人类微生物群是一个复杂的微生态系统,包括各种动态微生物种群,主要由细菌组成。其与宿主的相互作用强烈影响几个生理和病理过程。肠道微生物群越来越被认为是维持体内平衡的关键参与者,有助于肠道和大脑等远端器官的主要功能。然而,肠道菌群失调,具有肠屏障功能障碍的微生物群的组成和功能改变与几种病理的发展和进展有关,包括肠道炎性疾病,全身性自身免疫性疾病,比如风湿性关节炎,和神经退行性疾病,如老年痴呆症。此外,由于口腔微生物群研究对整体健康的潜在影响,近年来引起了人们的极大兴趣.关于微生物群-宿主相互作用在健康和疾病中的作用的新证据引发了人们对细菌胞外囊泡(BEV)作为王国间通信介质的功能作用的显着兴趣。越来越多的证据表明,BEV通过转运和递送到调节宿主信号通路和细胞过程的宿主细胞效应分子来介导宿主相互作用。影响健康和疾病。这篇综述讨论了BEV在肠道中的关键作用,肺,皮肤和口腔的上皮,免疫系统,和中枢神经系统相互作用。
    The human microbiota is an intricate micro-ecosystem comprising a diverse range of dynamic microbial populations mainly consisting of bacteria, whose interactions with hosts strongly affect several physiological and pathological processes. The gut microbiota is being increasingly recognized as a critical player in maintaining homeostasis, contributing to the main functions of the intestine and distal organs such as the brain. However, gut dysbiosis, characterized by composition and function alterations of microbiota with intestinal barrier dysfunction has been linked to the development and progression of several pathologies, including intestinal inflammatory diseases, systemic autoimmune diseases, such as rheumatic arthritis, and neurodegenerative diseases, such as Alzheimer\'s disease. Moreover, oral microbiota research has gained significant interest in recent years due to its potential impact on overall health. Emerging evidence on the role of microbiota-host interactions in health and disease has triggered a marked interest on the functional role of bacterial extracellular vesicles (BEVs) as mediators of inter-kingdom communication. Accumulating evidence reveals that BEVs mediate host interactions by transporting and delivering into host cells effector molecules that modulate host signaling pathways and cell processes, influencing health and disease. This review discusses the critical role of BEVs from the gut, lung, skin and oral cavity in the epithelium, immune system, and CNS interactions.
    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
    患有认知疾病的肝硬化患者被认为患有肝性脑病(HE),这导致了不必要的药物,而忽略了导致这些投诉的潜在疾病过程。自从神经心理学测试以来,目前诊断HE的金标准,不是现成的,需要一个有序的测试。我们旨在开发和验证快速肠道微生物群测试,以排除HE并确定利益相关者对这种方法的投入。从两个队列中收集粪便:双中心训练队列(n=305,在/不在HE相关治疗上)和多中心验证队列(n=30,在HE治疗上)。使用纳米孔分析快速分析粪便微生物群。使用半定量问卷评估利益相关者(患者和临床医生)需求评估。在训练组中,与黄金标准神经心理学测试相比,使用神经网络的机器学习识别出20种特征,该特征区分HE和无HE,特异性为84%。无论患者是否接受HE相关治疗,这种高特异性仍然存在。在验证队列中,该配置文件的应用导致对>40%患者的HE诊断和治疗进行重新评估.患者(验证队列中的退伍军人)和临床医生(全国40名)利益相关者都可以接受这种方法。我们得出的结论是,基于在训练集中开发并在单独的多中心前瞻性队列中验证的20种微生物物种的机器学习粪便特征可区分与没有他,被误诊为HE的患者,并且患者和临床医生利益相关者都可以接受。
    Patients with cirrhosis who have cognitive complaints are presumed to have hepatic encephalopathy (HE), which leads to unwarranted medications while ignoring the underlying disease process causing these complaints. Since neuropsychological testing, the current gold standard for HE diagnosis, is not readily available, an orderable test is needed. We aimed to develop and validate a rapid gut microbiota test to exclude HE and determine stakeholder input on this approach. Stool was collected from two cohorts: a two-center training cohort (n = 305, on/not on HE-related therapy) and a multicenter validation cohort (n = 30, on HE treatment). Stool microbiota was analyzed rapidly using nanopore analysis. Stakeholder (patients and clinicians) needs assessment was evaluated using semi-quantitative questionnaires. In the training cohort, machine learning using neural network identified a 20-species signature that differentiated HE vs no-HE with 84% specificity compared to the gold standard neuropsychological testing. This high specificity persisted regardless of whether patients were on HE-related therapy or not. In the validation cohort, application of this profile led to reevaluation of the HE diagnosis and treatment in > 40% of the patients. This approach was acceptable to patients (Veterans in the validation cohort) and clinician (n = 40 nationwide) stakeholders. We conclude that a machine learning stool signature based on 20 microbial species developed in a training set and validated in a separate multicenter prospective cohort differentiated those with vs. without HE, identified patients misdiagnosed with HE, and was acceptable to patients and clinician stakeholders.
    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
    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
    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
    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
    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)

  • 文章类型: Journal Article
    阿尔茨海默病(AD)是痴呆的最普遍和最不可逆的形式,占痴呆病例的一半以上。AD最重要的危险因素是与衰老相关的恶化,解剖通路的退化,环境变量和线粒体功能障碍。找到决定性的治疗解决方案是当前的主要问题。AD患者的主要神经病理学机制与微生物组之间的细微差别的相互作用最近受到越来越多的关注。肠道中细菌淀粉样蛋白的存在会触发免疫系统,导致中枢神经系统内免疫反馈和内源性神经元淀粉样蛋白增加。此外,早期的临床研究表明,用有益细菌或益生菌改变微生物组可能会影响AD患者的脑功能.新方法集中于AD中疾病改善药物的可能的神经保护作用。在本次审查中,我们讨论了肠道微生物群对大脑的影响,并回顾了新兴的研究,这些研究表明微生物群-脑轴的破坏可通过介导神经炎症影响AD.这样的新方法可以帮助开发用于AD的新疗法。
    Alzheimer\'s disease (AD) is the most widespread and irreversible form of dementia and accounts for more than half of dementia cases. The most significant risk factors for AD are aging-related exacerbations, degradation of anatomical pathways, environmental variables and mitochondrial dysfunction. Finding a decisive therapeutic solution is a major current issue. Nuanced interactions between major neuropathological mechanisms in AD in patients and microbiome have recently gained rising attention. The presence of bacterial amyloid in the gut triggers the immune system, resulting in increased immune feedbacks and endogenous neuronal amyloid within the CNS. Also, early clinical research revealed that changing the microbiome with beneficial bacteria or probiotics could affect brain function in AD. New approaches focus on the possible neuroprotective action of disease-modifying medications in AD. In the present review, we discuss the impact of the gut microbiota on the brain and review emerging research that suggests a disruption in the microbiota-brain axis can affect AD by mediating neuroinflammation. Such novel methods could help the development of novel therapeutics for AD.
    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
    维持肠道健康对于成功的家禽生产至关重要,也是家禽生产行业的目标之一。几十年来,家禽业一直依靠包含抗生素生长促进剂(AGP)来实现这一目标并提高生长性能。随着AGP的移除,慢性的出现,低水平的肠道炎症已经成为家禽业关注的焦点,饮食是炎症触发因素的主要来源。我们开发了一种低年级饮食模型,肉鸡的慢性肠道炎症,采用饲喂由30%米糠组成的高非淀粉多糖(NSP)饮食来研究这种炎症对鸟类性能和生理的影响。对于目前的研究,我们假设低度慢性炎症会导致肠道肠神经系统中的神经元分泌神经化学物质,这些神经化学物质激活免疫细胞,从而驱动炎症并对鸟类的表现产生负面影响。为了检验我们的假设,将1天大的肉鸡称重,并分为2种饮食方案:对照玉米大豆饮食和一组高NSP饮食(30%米糠)。在7-,14-,21-,和28-d接种后(PH),鸟被称重,收集的粪便,处死5只鸟,切除十二指肠和盲肠组织,收集十二指肠和盲肠内容物进行超高效液相色谱分析(UHPLC)。UHPLC显示,与对照饲喂的鸟类相比,饲喂高NSP饮食的鸟类中去甲肾上腺素(NOR)的浓度增加了1000倍。Further,在所有时间点,接受NSP饮食的禽类的粪便NOR浓度也显著升高.从1到14天PH,增重和饲料转化率没有差异,但是饲喂高NSP饮食的鸟类的增重和饲料转化率从14到28dPH显着降低。结果表明,饮食诱导的低度慢性炎症反应增加了肠道中的NOR产生,这对鸟类的表现产生了负面影响。这项研究表明,神经免疫途径可以作为开发新的干预措施的机制目标,以降低慢性炎症的发生率,从而有益于表现。
    Maintenance of intestinal health is critical to successful poultry production and one of the goals of the poultry production industry. For decades the poultry industry has relied upon the inclusion of antibiotic growth promoters (AGP) to achieve this goal and improve growth performance. With the removal of AGPs, the emergence of chronic, low-level gut inflammation has come to the forefront of concern in the poultry industry with the diet being the primary source of inflammatory triggers. We have developed a dietary model of low-grade, chronic intestinal inflammation in broilers that employs feeding a high nonstarch polysaccharides (NSP) diet composed of 30% rice bran to study the effects of this inflammation on bird performance and physiology. For the present studies, we hypothesize that the low-grade chronic inflammation causes neurons in the intestinal enteric nervous system to secrete neurochemicals that activate immune cells that drive the inflammation and negatively affect bird performance. To test our hypothesis, 1-day-old broiler chickens were weighed and divided into 2 dietary regimes: a control corn-soybean diet and a group fed a high NSP diet (30% rice bran). At 7-, 14-, 21-, and 28-d posthatch (PH), birds were weighed, fecal material collected, and 5 birds were sacrificed and sections of duodenal and cecal tissues excised, and duodenal and cecal contents collected for ultra-high performance liquid chromatography analyses (UHPLC). UHPLC revealed 1000s-fold increase in the concentration of norepinephrine (NOR) in birds fed the high NSP diet compared to the control fed birds. Further, the fecal concentrations of NOR were also found to be significantly elevated in the birds on the NSP diet throughout all time points. There were no differences in weight gain nor feed conversion from 1 to 14 d PH, but birds fed the high NSP diet had significantly reduced weight gain and feed conversion from 14 to 28 d PH. The results revealed that a dietary-induced low-grade chronic inflammatory response increased NOR production in the gut which negatively affected bird performance. This study suggests that neuroimmune pathways may serve as a mechanistic target for the development of new interventions to decrease the incidence of chronic inflammation and thereby benefit performance.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号