Brain-gut axis

大脑 - 肠道轴
  • 文章类型: Journal Article
    背景:肺炎克雷伯菌因医院感染和败血症而臭名昭著,这也与阿尔茨海默病(AD)相关的神经炎症和神经退行性损害有关。然而,其在AD病理中的病因和机制作用仍未研究。
    方法:在AD模型(3xTg-AD小鼠)中开发了肺炎克雷伯菌肠道感染和定植的临床前模型,以研究肺炎克雷伯菌的发病机制是否以及如何通过肠-血-脑轴加剧神经发病机制。
    结果:K.肺炎,特别是在抗生素诱导的生态失调下,能够通过穿透肠道上皮屏障从肠道转移到血液。随后,肺炎克雷伯菌通过破坏血脑屏障浸润大脑。在肺炎克雷伯氏菌脑感染的小鼠中观察到显著的神经炎症表型。肺炎克雷伯菌感染的小鼠还表现出神经行为功能受损和脑中总tau水平升高。宏基因组分析显示肺炎克雷伯菌与肠道生物群落多样性和共生细菌呈负相关。强调抗生素诱导的菌群失调如何引发肠脓毒症“病理组”特征与肠-脑扰动有关。
    结论:研究结果表明,医院感染后的感染因子和随后的抗生素治疗方案可能会引起肠道菌群失调和病理组,并增加脓毒症的风险,从而通过破坏肠-血脑屏障增加神经炎症和神经行为障碍的易感性。
    BACKGROUND: Klebsiella pneumoniae is infamous for hospital-acquired infections and sepsis, which have also been linked to Alzheimer disease (AD)-related neuroinflammatory and neurodegenerative impairment. However, its causative and mechanistic role in AD pathology remains unstudied.
    METHODS: A preclinical model of K. pneumoniae enteric infection and colonization is developed in an AD model (3xTg-AD mice) to investigate whether and how K. pneumoniae pathogenesis exacerbates neuropathogenesis via the gut-blood-brain axis.
    RESULTS: K. pneumoniae, particularly under antibiotic-induced dysbiosis, was able to translocate from the gut to the bloodstream by penetrating the gut epithelial barrier. Subsequently, K. pneumoniae infiltrated the brain by breaching the blood-brain barrier. Significant neuroinflammatory phenotype was observed in mice with K. pneumoniae brain infection. K. pneumoniae-infected mice also exhibited impaired neurobehavioral function and elevated total tau levels in the brain. Metagenomic analyses revealed an inverse correlation of K. pneumoniae with gut biome diversity and commensal bacteria, highlighting how antibiotic-induced dysbiosis triggers an enteroseptic \"pathobiome\" signature implicated in gut-brain perturbations.
    CONCLUSIONS: The findings demonstrate how infectious agents following hospital-acquired infections and consequent antibiotic regimen may induce gut dysbiosis and pathobiome and increase the risk of sepsis, thereby increasing the predisposition to neuroinflammatory and neurobehavioral impairments via breaching the gut-blood-brain barrier.
    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
    益生菌,postbiotics,n-3多不饱和脂肪酸(PUFA)具有抗抑郁作用。然而,多巴胺能通路的潜在机制尚不清楚.本研究调查了以下假设:益生菌和postbiotics结合n-3PUFA通过调节大脑-肠轴的多巴胺能途径协同改善抑郁症。大鼠随机分为7组:非慢性轻度应激(CMS)与n-6PUFA,和带有n-6PUFA的CMS,n-3PUFA,益生菌,postbiotics,益生菌联合n-3PUFA,和与n-3PUFA联用的postbiotics。益生菌,postbiotics,n-3PUFA改善了抑郁行为,干扰素-γ的血液浓度降低,白细胞介素-1β,并增加大脑和肠道中短链脂肪酸和多巴胺的浓度。此外,益生菌,postbiotics,n-3PUFA增加了糖皮质激素受体和酪氨酸羟化酶的脑和肠道表达;l型氨基酸转运蛋白1和多巴胺受体(DR)D1的脑表达;以及DRD2的肠道表达。脑中磷酸化蛋白激酶A/蛋白激酶A和磷酸化cAMP反应元件结合蛋白/cAMP反应元件结合蛋白的表达增加,然而,通过补充益生菌在肠道中减少,postbiotics,和n-3PUFA。益生菌和益生菌联合n-3PUFA对血液中的抑郁行为和多巴胺能通路有协同作用,大脑,和直觉。此外,益生菌和益生菌之间的多巴胺能途径没有显着差异。总之,益生菌和益生菌,在暴露于CMS的大鼠中,与n-3PUFA结合对通过脑-肠轴的多巴胺能途径具有协同抗抑郁样作用。
    Probiotics, postbiotics, and n-3 polyunsaturated fatty acids (PUFA) have antidepressant-like effects. However, the underlying mechanisms of the dopaminergic pathway are unclear. The present study investigated the hypothesis that probiotics and postbiotics combined with n-3 PUFA synergistically improve depression by modulating the dopaminergic pathway through the brain-gut axis. Rats were randomly divided into seven groups: non-chronic mild stress (CMS) with n-6 PUFA, and CMS with n-6 PUFA, n-3 PUFA, probiotics, postbiotics, probiotics combined with n-3 PUFA, and postbiotics combined with n-3 PUFA. Probiotics, postbiotics, and n-3 PUFA improved depressive behaviors, decreased blood concentrations of interferon-γ, and interleukin-1β, and increased the brain and gut concentrations of short chain fatty acids and dopamine. Moreover, probiotics, postbiotics, and n-3 PUFA increased the brain and gut expression of glucocorticoid receptor and tyrosine hydroxylase; brain expression of l-type amino acid transporter 1 and dopamine receptor (DR) D1; and gut expression of DRD2. The expression of phosphorylated protein kinase A/protein kinase A and phosphorylated cAMP response element-binding protein/cAMP response element-binding protein increased in the brain, however, decreased in the gut by the supplementation of probiotics, postbiotics, and n-3 PUFA. There was synergistic effect of probiotics and postbiotics combined with n-3 PUFA on the depressive behaviors and dopaminergic pathway in blood, brain, and gut. Moreover, no significant difference in the dopaminergic pathways between the probiotics and postbiotics was observed. In conclusion, probiotics and postbiotics, combined with n-3 PUFA have synergistic antidepressant-like effects on the dopaminergic pathway through the brain-gut axis in rats exposed to CMS.
    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
    Tau与α-突触核蛋白(α-Syn)相互作用并与它共同定位在路易体中,影响帕金森病(PD)的α-Syn病理学。然而,这些生化事件是否调节从肠道到大脑的α-Syn病理扩散仍未完全了解。这里,我们显示,α-Syn和Tau共病在肠诱导型SYN103+/-和/或TAU368+/-转基因小鼠模型中扩散到大脑中,引发行为缺陷。最初观察到肠道病理,和α-Syn或Tau病理随后传播到DMV或NTS,然后传播到其他大脑区域。值得注意的是,在双转基因小鼠(Both)中发现比在单转基因小鼠中更广泛的传播和广泛的神经元丢失。截短迷走神经切断术和α-Syn缺乏症可显着抑制突触核蛋白病或tau蛋白病的扩散。α-SynPET示踪剂[18F]-F0502B在肠和脑中检测到α-Syn聚集体。因此,α-Syn和Tau共病可以从肠道传播到大脑,引发行为障碍。
    Tau interacts with α-Synuclein (α-Syn) and co-localizes with it in the Lewy bodies, influencing α-Syn pathology in Parkinson\'s disease (PD). However, whether these biochemical events regulate α-Syn pathology spreading from the gut into the brain remains incompletely understood. Here, we show that α-Syn and Tau co-pathology is spread into the brain in gut-inducible SYN103+/- and/or TAU368+/- transgenic mouse models, eliciting behavioral defects. Gut pathology was initially observed, and α-Syn or Tau pathology was subsequently propagated into the DMV or NTS and then to other brain regions. Remarkably, more extensive spreading and widespread neuronal loss were found in double transgenic mice (Both) than in single transgenic mice. Truncal vagotomy and α-Syn deficiency significantly inhibited synucleinopathy or tauopathy spreading. The α-Syn PET tracer [18F]-F0502B detected α-Syn aggregates in the gut and brain. Thus, α-Syn and Tau co-pathology can propagate from the gut to the brain, triggering behavioral disorders.
    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
    哺乳动物的肠道含有称为肠道微生物组的微生物群落。肠道微生物组被整合到哺乳动物生理学中,有助于新陈代谢,代谢产物的产生,并促进免疫调节作用。小胶质细胞,大脑的固有免疫细胞,在稳态神经发生中起着至关重要的作用,突触重塑,和神经胶质成熟。小胶质细胞功能障碍与几种神经精神疾病的发病机理有关。最近的发现表明,小胶质细胞在整个生命过程中受到肠道微生物组及其衍生代谢物的影响。微生物群调节小胶质细胞的途径才刚刚开始被理解,但是这一发现有可能为与微生物组改变相关的脑部疾病的发病机制提供有价值的见解。这里,我们讨论了有关肠道微生物组在发育和成年期调节小胶质细胞中的作用的最新文献,并总结了在神经精神和神经退行性疾病的某些例子中这种双向串扰的关键发现。我们还强调了该领域的一些当前警告和观点。
    The mammalian gut contains a community of microorganisms called gut microbiome. The gut microbiome is integrated into mammalian physiology, contributing to metabolism, production of metabolites, and promoting immunomodulatory actions. Microglia, the brain\'s resident innate immune cells, play an essential role in homeostatic neurogenesis, synaptic remodeling, and glial maturation. Microglial dysfunction has been implicated in the pathogenesis of several neuropsychiatric disorders. Recent findings indicate that microglia are influenced by the gut microbiome and their derived metabolites throughout life. The pathways by which microbiota regulate microglia have only started to be understood, but this discovery has the potential to provide valuable insights into the pathogenesis of brain disorders associated with an altered microbiome. Here, we discuss the recent literature on the role of the gut microbiome in modulating microglia during development and adulthood and summarize the key findings on this bidirectional crosstalk in selected examples of neuropsychiatric and neurodegenerative disorders. We also highlight some current caveats and perspectives for the field.
    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
    神经性贪食症(BN)是一种典型的周期性行为活动,以限制为特征,暴饮暴食和呕吐,以及对食物情感价值的干扰。食物刺激获得过度的相关性,引起一系列兴奋和焦虑的状态。抑郁状况经常伴随着BN。大多数BN患者也会经历一种或多种焦虑症。审查的目的是确定中枢和外周水平的联系,将进食障碍与情绪状态联系起来。BN涉及改变的神经机制。在大脑区域中,BN中的脑岛功能受损。脑岛也与抑郁状态有关。脑岛是主要的味觉皮层,味觉感官信息,如味觉辨别和更高的认知功能,如食物预期和奖励被处理。脑岛在解剖学上与广泛的皮质相连,边缘和旁缘结构,在功能上涉及高阶认知,情绪反应,和移情过程。脑岛在移情中起着至关重要的作用,或者分享他人情绪状态的能力,尤其是负面情绪。事实上,在焦虑和抑郁的情况下,岛叶皮层也被激活。影响贪食症和抑郁症的病理生理因素之一是肠道菌群的组成,因为微生物特征和大脑功能之间有很强的关联。肠道菌群失调状况可能导致饮食失调的发展,包括BN。菌群失调可能会促进肠道炎症,改变肠道通透性,并在饥饿/饱腹感调节中心引发免疫反应,促进饮食失调的病理生理发展。由此,在这些病症中,充分的益生菌整合作为预防和/或治疗工具的重要性得以显现。
    Bulimia nervosa (BN) is a condition marked by a typical cyclical behavioural activity, characterized by restrictions, binges and vomiting, as well as a disturbance of the emotional value of food. Food stimuli acquire excessive relevance, giving rise to a succession of states of excitement and anxiety. The depressive condition accompanies very often BN. Most people with BN also experience one or more anxiety disorders. The aim of the review is to identify a link at a central and peripheral level that connects an eating disorder with a mood state. Altered nervous mechanisms are involved in BN. Among the cerebral areas, the insula is functionally compromised in BN. The insula is also implicated in depressive states. The insula is the primary gustatory cortex, where gustatory sensory information such as taste discrimination and higher cognitive functions such as food anticipation and reward are processed. The insula is anatomically connected to a wide range of cortical, limbic and paralimbic structures, and functionally implicated in high-order cognition, emotional responses, and empathic processes. The insula plays a crucial role in empathy, or in the ability to share the emotional states of others, and in particular negative emotions. In fact, the insular cortex is also activated in conditions of anxiety and depression. One of the pathophysiological factors that influences bulimia and depression is the composition of gut microbiota, as there is a strong association between the microbial signature and the brain function. Gut dysbiosis condition may contribute to the development of eating disorders, including BN. Dysbiosis may promote intestinal inflammation, alter gut permeability, and trigger immune reactions in the hunger/satiety regulation center contributing to the pathophysiological development of eating disorders. From this emerges the importance of adequate probiotic integration as a preventive and/or therapeutic tool in these pathologies.
    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
    肠易激综合征(IBS)是一种以腹痛为特征的常见功能性胃肠病,腹胀,腹泻,还有便秘.最近的研究强调了肠道微生物群在IBS发病机理中的重要作用。体育锻炼,作为一种非药物干预措施,已提出通过调节肠道微生物群来缓解IBS症状。有氧运动,比如跑步,游泳,骑自行车,已被证明可以增强有益肠道细菌的多样性和丰度,包括乳酸菌和双歧杆菌。这些细菌产生具有抗炎特性并支持肠屏障完整性的短链脂肪酸。IBS患者参与有组织的有氧运动项目的研究报告了他们的肠道微生物群的组成和多样性的显著改善,伴随着腹痛和腹胀等症状的缓解。此外,运动通过减轻压力和改善情绪来积极影响心理健康,这可以通过肠-脑轴进一步缓解IBS症状。长期运动干预提供了持续的好处,保持肠道微生物群的多样性和稳定性,支持免疫功能,减少全身炎症。然而,锻炼计划必须根据个人需要进行调整,以避免加重IBS症状.从低强度到中等强度开始并逐渐增加强度的个性化运动计划可以最大程度地提高收益并最大程度地降低风险。这篇综述探讨了不同类型和强度的体育锻炼对IBS患者肠道菌群的影响。强调需要进一步研究以探索最佳运动方案。未来的研究应该包括更大的样本量,更长的随访期,并检查运动和其他生活方式改变的协同作用。将体育锻炼纳入综合IBS管理计划可以增强症状控制并改善患者的生活质量。
    Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder characterized by abdominal pain, bloating, diarrhea, and constipation. Recent studies have underscored the significant role of the gut microbiota in the pathogenesis of IBS. Physical exercise, as a non-pharmacological intervention, has been proposed to alleviate IBS symptoms by modulating the gut microbiota. Aerobic exercise, such as running, swimming, and cycling, has been shown to enhance the diversity and abundance of beneficial gut bacteria, including Lactobacillus and Bifidobacterium. These bacteria produce short-chain fatty acids that possess anti-inflammatory properties and support gut barrier integrity. Studies involving IBS patients participating in structured aerobic exercise programs have reported significant improvements in their gut microbiota\'s composition and diversity, alongside an alleviation of symptoms like abdominal pain and bloating. Additionally, exercise positively influences mental health by reducing stress and improving mood, which can further relieve IBS symptoms via the gut-brain axis. Long-term exercise interventions provide sustained benefits, maintaining the gut microbiota\'s diversity and stability, supporting immune functions, and reducing systemic inflammation. However, exercise programs must be tailored to individual needs to avoid exacerbating IBS symptoms. Personalized exercise plans starting with low-to-moderate intensity and gradually increasing in intensity can maximize the benefits and minimize risks. This review examines the impact of various types and intensities of physical exercise on the gut microbiota in IBS patients, highlighting the need for further studies to explore optimal exercise protocols. Future research should include larger sample sizes, longer follow-up periods, and examine the synergistic effects of exercise and other lifestyle modifications. Integrating physical exercise into comprehensive IBS management plans can enhance symptom control and improve patients\' 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
    这篇叙述性综述综合了目前有关抗炎饮食模式及其对精神障碍和神经退行性疾病患者的潜在益处的证据。慢性低度炎症越来越被认为是这些病症的病因和进展的关键因素。该评论检查了饮食成分和食物组的抗炎和神经保护特性的证据,专注于整个食物,而不是特定的营养素或补充剂。显示潜在益处的关键饮食成分包括水果和蔬菜(尤其是浆果和绿叶蔬菜),全谷物,豆类,富含omega-3的脂肪鱼,坚果(特别是核桃),橄榄油,和发酵食品。这些食物通常富含抗氧化剂,膳食纤维,和可能有助于调节炎症的生物活性化合物,支持肠道健康,促进神经保护。相反,超加工食品,红肉,含糖饮料可能有害。根据这些证据,我们设计了大脑抗炎营养(Brain)饮食。这种饮食的机制包括肠道微生物群和肠-脑轴的调节,炎症途径的调节,减少氧化应激,和促进神经可塑性。Brain饮食显示出有望帮助管理精神和神经退行性疾病。
    This narrative review synthesizes current evidence regarding anti-inflammatory dietary patterns and their potential benefits for individuals with mental disorders and neurodegenerative diseases. Chronic low-grade inflammation is increasingly recognized as a key factor in the etiology and progression of these conditions. The review examines the evidence for the anti-inflammatory and neuroprotective properties of dietary components and food groups, focusing on whole foods rather than specific nutrients or supplements. Key dietary components showing potential benefits include fruits and vegetables (especially berries and leafy greens), whole grains, legumes, fatty fish rich in omega-3, nuts (particularly walnuts), olive oil, and fermented foods. These foods are generally rich in antioxidants, dietary fiber, and bioactive compounds that may help modulate inflammation, support gut health, and promote neuroprotection. Conversely, ultra-processed foods, red meat, and sugary beverages may be harmful. Based on this evidence, we designed the Brain Anti-Inflammatory Nutrition (BrAIN) diet. The mechanisms of this diet include the modulation of the gut microbiota and the gut-brain axis, the regulation of inflammatory pathways, a reduction in oxidative stress, and the promotion of neuroplasticity. The BrAIN diet shows promise as an aid to manage mental and neurodegenerative disorders.
    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
    大量研究表明,神经精神疾病(精神疾病和情绪障碍)伴随侵略(或暴力)对公共卫生构成重大挑战,并在全球范围内造成巨大的经济负担。尤其是,与童年逆境相关的社会混乱(或社会不平等)对心理健康有长期影响,增加患神经精神疾病的风险。肠道细菌,作为一个内分泌器官和第二个大脑,释放各种免疫调节剂和生物活性化合物直接或间接调节宿主的生理和行为稳态。在各种社会挑战下,应激诱导的生态失调增加肠道通透性引起一系列反应:释放神经毒性化合物,导致神经炎症和神经元损伤,最终与攻击性相关的神经精神疾病,暴力,或通过微生物群-肠脑(MGB)轴的复杂双向通信在人类和各种动物中的冲动行为。MGB轴的失调也被认为是社会压力引起的有害行为(羽毛啄食,侵略,和吃人的啄食)。然而,在人类和鸡中预防和治疗这些疾病的现有知识还没有得到很好的理解。在以往的研究中,我们在一项异常行为调查中建立了一种非哺乳动物模型,通过合理化肠道微生物群对鸡的伤害行为的影响。基于我们先前的成功,透视文章概述了通过盲肠微生物群移植改变肠道微生物群来减少鸡应激诱导的伤害行为的可能性,通过恢复肠道微生物群的多样性和功能,有可能为预防精神障碍患者的有害行为提供生物治疗原理。
    Numerous studies have evidenced that neuropsychiatric disorders (mental illness and emotional disturbances) with aggression (or violence) pose a significant challenge to public health and contribute to a substantial economic burden worldwide. Especially, social disorganization (or social inequality) associated with childhood adversity has long-lasting effects on mental health, increasing the risk of developing neuropsychiatric disorders. Intestinal bacteria, functionally as an endocrine organ and a second brain, release various immunomodulators and bioactive compounds directly or indirectly regulating a host\'s physiological and behavioral homeostasis. Under various social challenges, stress-induced dysbiosis increases gut permeability causes serial reactions: releasing neurotoxic compounds, leading to neuroinflammation and neuronal injury, and eventually neuropsychiatric disorders associated with aggressive, violent, or impulsive behavior in humans and various animals via a complex bidirectional communication of the microbiota-gut-brain (MGB) axis. The dysregulation of the MGB axis has also been recognized as one of the reasons for the prevalence of social stress-induced injurious behaviors (feather pecking, aggression, and cannibalistic pecking) in chickens. However, existing knowledge of preventing and treating these disorders in both humans and chickens is not well understood. In previous studies, we developed a non-mammal model in an abnormal behavioral investigation by rationalizing the effects of gut microbiota on injurious behaviors in chickens. Based on our earlier success, the perspective article outlines the possibility of reducing stress-induced injurious behaviors in chickens through modifying gut microbiota via cecal microbiota transplantation, with the potential for providing a biotherapeutic rationale for preventing injurious behaviors among individuals with mental disorders via restoring gut microbiota diversity and function.
    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
    生物人类学的方方面面,包括发展,老化,疾病,甚至健康维护,受肠道微生物群显著的遗传和代谢能力的影响。随着当前测序技术的进步和新的培养无关的方法,研究人员可以超越较早的相关研究,并开发基于机制的微生物组-宿主相互作用研究。微生物群-肠-脑轴(MGBA)调节神经胶质功能,使其成为改善神经退行性疾病(NDDs)治疗的发展和进步的可能目标。肠脑轴(GBA)负责胃肠道和中枢神经系统之间的相互通信,在控制饥饿等生理过程的调节中起着至关重要的作用,新陈代谢,和各种胃肠功能。最近,研究发现肠道微生物群对大脑健康的功能-不同的微生物群通过不同的途径,如免疫学,神经和代谢途径。此外,我们回顾了与肠道微生物群相关的神经递质和肠道激素的参与。我们还通过关注代谢物来探索神经退行性疾病中的MGBA。Further,针对血脑屏障(BBB),肠屏障,脑膜,并对外周免疫系统进行了研究。最后,我们讨论了治疗方法,并评估了有关使用益生元的临床前和临床试验数据,益生菌,副益生菌,粪便微生物移植,个性化医疗,和具有生物活性的天然食物。对GBA的全面研究将有助于创造有效的治疗方法来治疗不同的NDD。
    Every facet of biological anthropology, including development, ageing, diseases, and even health maintenance, is influenced by gut microbiota\'s significant genetic and metabolic capabilities. With current advancements in sequencing technology and with new culture-independent approaches, researchers can surpass older correlative studies and develop mechanism-based studies on microbiome-host interactions. The microbiota-gut-brain axis (MGBA) regulates glial functioning, making it a possible target for the improvement of development and advancement of treatments for neurodegenerative diseases (NDDs). The gut-brain axis (GBA) is accountable for the reciprocal communication between the gastrointestinal and central nervous system, which plays an essential role in the regulation of physiological processes like controlling hunger, metabolism, and various gastrointestinal functions. Lately, studies have discovered the function of the gut microbiome for brain health-different microbiota through different pathways such as immunological, neurological and metabolic pathways. Additionally, we review the involvement of the neurotransmitters and the gut hormones related to gut microbiota. We also explore the MGBA in neurodegenerative disorders by focusing on metabolites. Further, targeting the blood-brain barrier (BBB), intestinal barrier, meninges, and peripheral immune system is investigated. Lastly, we discuss the therapeutics approach and evaluate the pre-clinical and clinical trial data regarding using prebiotics, probiotics, paraprobiotics, fecal microbiota transplantation, personalised medicine, and natural food bioactive in NDDs. A comprehensive study of the GBA will felicitate the creation of efficient therapeutic approaches for treating different NDDs.
    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
    肠道菌群是宿主生理过程和发病机制的重要因素。中枢神经系统(CNS)和微生物群之间的交流通过不同的途径发生,即化学,神经,免疫,和内分泌。肠道微生物群的改变,即肠道菌群失调导致中枢神经系统和肠道微生物群之间的双向交流发生变化,并与神经和神经发育障碍的发病机理有关。因此,如今,微生物区系-肠脑轴(MGBA)已成为治疗代谢紊乱的治疗靶点.但是,从基础研究中获得的MGBA实验数据限制了其在临床研究中的应用。在本研究中,我们首先通过收集不同来源的数据总结了微生物群与大脑生理和发病机理相互作用的分子机制,即PubMed,Scopus,Web的科学。此外,证据表明,脂肪组织(AT)在代谢活动期间是活跃的,也可能与MGBA相互作用。因此,在本研究中,我们关注的是MGBA之间的关系,棕色脂肪组织,和白色脂肪组织。伴随着这个,我们还研究了AT的功能特异性,并了解MGBA和不同类型AT之间的异质性。因此,它们之间的分子相互作用可能为神经系统疾病的治疗提供治疗靶点。
    The gut microbiota is an important factor responsible for the physiological processes as well as pathogenesis of host. The communication between central nervous system (CNS) and microbiota occurs by different pathways i.e., chemical, neural, immune, and endocrine. Alteration in gut microbiota i.e., gut dysbiosis causes alteration in the bidirectional communication between CNS and gut microbiota and linked to the pathogenesis of neurological and neurodevelopmental disorder. Therefore, now-a-days microbiota-gut-brain-axis (MGBA) has emerged as therapeutic target for the treatment of metabolic disorder. But, experimental data available on MGBA from basic research has limited application in clinical study. In present study we first summarized molecular mechanism of microbiota interaction with brain physiology and pathogenesis via collecting data from different sources i.e., PubMed, Scopus, Web of Science. Furthermore, evidence shows that adipose tissue (AT) is active during metabolic activities and may also interact with MGBA. Hence, in present study we have focused on the relationship among MGBA, brown adipose tissue, and white adipose tissue. Along with this, we have also studied functional specificity of AT, and understanding heterogeneity among MGBA and different types of AT. Therefore, molecular interaction among them may provide therapeutic target for the treatment of neurological disorder.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号