肠道菌群(GutMircrobiota≈CNS!)
- 格式:docx
- 大小:2.65 MB
- 文档页数:15
一片看懂肠道菌群在人体中的作用日文名:肠内フローラ解明!惊异の细菌パワー中文名:肠道细菌 / 肠道花园类型:医疗健康时长:49min官网:播出时间:2015年2月22日午後9时00分~9时49分腾讯视频链接英语中字版,非会员只能看5分钟,后台回复“肠道细菌”下载1080p英语中字版收藏观看日语中字版,在线可看全片,后台回复“肠道细菌”下载720p日语中字mp4版收藏观看由于面向读者略有不同,两个版本画面也有15分钟许多不同。
如日语还有几段综艺风格的段落。
影片简介在我们的身体中,存在着各种各样的细菌。
尤其在肠道内,存在着一个肠道细菌生态系统,就是所谓的肠道floral(肠道菌群、肠道微生物组)。
其中的细菌种类超过数百种,细菌总量超过了100兆,这是一个什么样的世界呢随着现代科技和医疗技术的发展,科学家发现这个生态系统中的细菌居然跟我们的健康、美容、甚至性格都有着深不可测的关联。
特别是在医疗方面,癌症、糖尿病、抑郁症等疾病都与之有关。
现在在欧美国家正在掀起一场医疗革命,使用一种称为粪便微生物移植的特殊的治疗方法,就可以彻底治愈很多之前无法治愈的顽疾。
随着研究的进一步深入,将来会有更多的疑难杂症会被攻克。
闲言少叙,让我们来了解一下隐藏在我们的身体中的这些不可思议的小伙伴们吧。
图文解读在我们的身体中隐藏着不为人知的秘密,即美容和保持健康的机制。
这里是吸收营养的肠道内部,实际上存在着肉眼无法看到的微小生命。
这是在我们的肠道中居住着的细菌们,它们的数量超过了100兆。
它们被称为肠道生态系统。
floral是花圃的意思。
肠道中的花圃是各种细菌的家园。
现在肠内生态系统的研究使医疗产生了巨大的变化。
全世界的国家接二连三地启动了国家级的项目,使用最先进的基因解析技术,陆续发现了新的细菌。
研究发现这些肠内细菌会影响到全身的健康。
癌症、糖尿病、肥胖症、过敏,之前从未考虑过这些疾病跟肠内细菌有关。
我们已经发现了30多种疾病与肠内生态系统的关系。
肠道菌群名词解释肠道菌群是一个复杂的生态系统,由多种细菌、病毒、真菌等微生物组成,主要居住在人类的肠道中。
这些微生物通过与宿主(即人体)的相互作用,对宿主的健康产生重要影响。
肠道菌群在维持人体健康方面发挥着许多作用,包括帮助消化食物、合成维生素、调节免疫系统等。
肠道菌群失衡可能会导致各种健康问题,包括肠道疾病、代谢性疾病、免疫系统疾病等。
近年来,随着人们生活方式的改变,如饮食习惯、卫生习惯和抗生素的滥用等,肠道菌群的平衡逐渐受到破坏,引发了一系列健康问题。
因此,了解肠道菌群的结构和功能,以及如何维护肠道菌群的平衡,对于预防和治疗相关疾病具有重要的意义。
肠道菌群的结构和组成因人而异,受到许多因素的影响,如饮食习惯、生活方式、卫生条件、遗传因素等。
通过对肠道菌群的研究,人们发现了一些与健康和疾病相关的菌群特征,如肥胖人群的肠道菌群与瘦弱人群的肠道菌群存在显著差异;糖尿病患者的肠道菌群中某些益生菌的数量减少,而有害菌的数量增加。
这些发现为人们提供了新的思路和方法来预防和治疗相关疾病。
为了维护肠道菌群的平衡,人们需要采取一系列措施,包括合理饮食、适当运动、保持良好的卫生习惯、避免滥用抗生素等。
同时,针对肠道菌群的研究和应用也在不断深入,如益生菌和益生元的开发和利用、肠道微生物组学的研究等。
这些研究和实践将有助于人们更好地了解肠道菌群,从而更好地维护自己的健康。
综上所述,肠道菌群是一个重要的生态系统,对人类健康产生着深远的影响。
通过深入研究和了解肠道菌群的结构和功能,以及如何维护肠道菌群的平衡,人们可以更好地预防和治疗相关疾病,提高自己的健康水平和生活质量。
同时,随着科技的不断发展,人们对肠道菌群的认识和应用也将不断深入和完善。
在未来的研究中,人们需要进一步探索肠道菌群与人体健康的相互作用机制,以及如何通过调节肠道菌群来改善和治疗各种疾病。
此外,还需要加强肠道微生物组学的研究,以便更深入地了解肠道菌群的组成和功能。
一、研究概述肠道微生物(gut microbiota),也称肠道菌群,指肠道中存在的数量庞大的微生物,这群微生物依靠宿主的肠道生活,同时帮助寄主完成多种生理生化功能。
人体肠道内寄生着10万亿个细菌,它们的基因总数约为人自身基因数目的150倍,肠道菌群也因此称为人体的“第二基因组”。
可以说人体与人体共生微生物构成了超级生物体(superorganism)。
肠道微生物与宿主之间进行密切的信息交流,在代谢、免疫、神经系统调控中起到了重要作用,能影响体重和消化能力、抵御感染和自体免疫疾病的患病风险,还能控制人体对癌症治疗药物的反应。
针对肠道菌群的研究主要分为两个方向,一是菌群结构研究,二是菌群代谢物研究。
二、菌群检测肠道菌群结构检测的方法主要为扩增子检测和宏基因组检测两种方式。
扩增子检测扩增子测序是对特定长度的PCR产物或者捕获片段进行测序,可研究样本中属水平以上的微生物群落组成及其丰度差异。
一般研究肠道菌群使用16S rDNA检测。
宏基因组检测宏基因组,又被称为元基因组,它通过直接从环境样品中提取全部微生物的DNA,构建宏基因组文库,利用基因组学的研究策略研究环境样品所包含的全部微生物的遗传组成及其群落功能。
它是在微生物基因组学的基础上发展起来的一种研究微生物多样性、开发新的生理活性物质(或获得新基因)的新理念和新方法。
宏基因组测序研究摆脱了微生物分离纯培养的限制,扩展了微生物资源的利用空间,为环境微生物群落的研究提供了有效工具。
检测方式对比迈维代谢可提供专业的菌群检测技术服务。
三、菌群代谢研究近年来基于高通量测序的微生物组学研究极大加深了人们对微生物与健康和疾病关系的认识。
然而基因测序方法不能直接测定微生物的功能活性,难以鉴定微生物中的关键功能分子,单独使用无法回答肠道微生物何种成员通过何种方式影响宿主等关键问题。
单一组学研究弊端显现出来,多组学联用的优势逐渐突出。
肠道微生物的代谢组学是以微生物群落所有小分子代谢物为研究对象,可发现肠道微生物随宿主病理生理变化的关键代谢物,为微生物组-宿主互作机制研究提供线索,成为微生物组学研究的重要补充。
肠道微生物的英语单词The Complex World of Gut Microbiota.The gut microbiota, often referred to as the "microbiome" or the "intestinal flora," refers to the vast community of microorganisms that reside within the human gastrointestinal tract. This intricate ecosystem plays a crucial role in maintaining our overall health and well-being. The gut microbiota is composed of a diverse range of bacteria, fungi, viruses, and other microorganisms that coexist in a delicate balance.The human body is estimated to contain trillions of microbial cells, outnumbering the human cells by a ratio of 10 to 1. The majority of these microbial cells reside in the gastrointestinal tract, particularly in the colon. The gut microbiota performs various vital functions, including digesting food, synthesizing vitamins, and regulating the immune system.Functions of the Gut Microbiota.Digestion and Nutrition: The gut microbiota aids in the breakdown of dietary fiber and other complex carbohydrates, releasing short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate. These SCFAs serve as a source of energy for the host and have been linked to various health benefits, including improved insulin sensitivity and reduced inflammation.Immune System Regulation: The gut microbiota plays a crucial role in shaping and regulating the immune system. It stimulates the development of immune cells and helps maintain a balanced immune response, protecting against both infectious diseases and autoimmune conditions.Barrier Function: The gut microbiota contributes to maintaining the integrity of the gut barrier, which prevents harmful bacteria and toxins from leaking into the bloodstream. A healthy gut microbiota supports tight junctions between gut cells, ensuring a strong barrier against pathogens.Brain-Gut Axis: The gut microbiota also interacts with the brain through the gut-brain axis, influencing mood, cognition, and behavior. This axis involves a complex communication network between the gastrointestinal tract and the central nervous system, which is believed to play a role in conditions like depression, anxiety, and autism.Importance of Gut Microbiota Balance.Disruptions to the gut microbiota, known as "dysbiosis," can lead to various health issues. Changes in the composition of the microbiota can be triggered by various factors, including diet, antibiotics, stress, and chronic illnesses.Diet: The composition of the gut microbiota is significantly influenced by the diet. A diet rich in fiber and diverse in plant-based foods promotes the growth of beneficial bacteria, while a diet high in processed foods and low in fiber can lead to a decrease in microbial diversity and an increase in harmful bacteria.Antibiotics: The use of antibiotics can have a profound impact on the gut microbiota, killing off both harmful and beneficial bacteria. This can lead to a temporary imbalance in the microbiota, allowing opportunistic pathogens to proliferate.Stress: Chronic stress has been shown to alter the gut microbiota composition, leading to an increase in inflammatory markers and a decrease in beneficial bacteria.Chronic Illnesses: Conditions like inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and obesity have been linked to alterations in the gut microbiota. These changes can contribute to the development and progression of these diseases.Modulating the Gut Microbiota.Given the crucial role of the gut microbiota in maintaining health, there has been increasing interest in modulating its composition through various strategies.Probiotics: Probiotics are live microorganisms that, when administered in adequate amounts, confer a health benefit on the host. They are commonly found in yogurt, fermented foods, and dietary supplements. Probiotics can help restore balance to the gut microbiota, improving digestive health and immune function.Prebiotics: Prebiotics are dietary fibers that promote the growth and activity of beneficial bacteria in the gut. By providing food for the probiotic bacteria, prebiotics can help support a healthy gut microbiota.Dietary Changes: Incorporating a diet rich in fiber, fruits, vegetables, and whole grains can promote the growth of beneficial bacteria and maintain gut microbiota diversity.Conclusion.The gut microbiota plays a pivotal role in maintaining human health and well-being. Its intricate balance ofmicroorganisms is essential for digestion, immune system regulation, and overall physiological functions. Disruptions to this balance can lead to various health issues, emphasizing the importance of maintaining a healthy gut microbiota through diet, lifestyle choices, and probiotic supplementation. As research in this field continues to evolve, so does our understanding of the crucial role the gut microbiota plays in our lives.。
REVIEW ARTICLESThe human gut microbiome:current knowledge, challenges,and future directionsMANEESH DAVE,PETER D.HIGGINS,SUMIT MIDDHA,and KEVIN P.RIOUXROCHESTER,MINN;ANN ARBOR,MICH;AND CALGARY,ALBERTA,CANADAThe Human Genome Project was completed a decade ago,leaving a legacy of pro-cess,tools,and infrastructure now being turned to the study of the microbes that re-side in and on the human body as determinants of health and disease,and has beenbranded‘‘The Human Microbiome Project.’’Of the various niches under investiga-tion,the human gut houses the most complex and abundant microbial communityand is an arena for important host–microbial interactions that have both local andsystemic impact.Initial studies of the human microbiome have been largely descrip-tive,a testing ground for innovative molecular techniques and new hypotheses.Methods for studying the microbiome have quickly evolved from low-resolution sur-veys of microbial community structure to high-definition description of composition,function,and ecology.Next-generation sequencing technologies combined withadvanced bioinformatics place us at the doorstep of revolutionary insight into thecomposition,capability,and activity of the human intestinal microbiome.Renewedefforts to cultivate previously‘‘uncultivable’’microbes will be important to the overallunderstanding of gut ecology.There remain numerous methodological challengesto the effective study and understanding of the gut microbiome,largely relating tostudy design,sample collection,and the number of predictor variables.Strategiccollaboration of clinicians,microbiologists,molecular biologists,computational sci-entists,and bioinformaticians is the ideal paradigm for success in thisfield.Meaning-ful interpretation of the gut microbiome requires that host genetic and environmentalinfluences be controlled or accounted for.Understanding the gut microbiome inhealthy humans is a foundation for discovering its influence in various important gas-trointestinal and nutritional diseases(eg,inflammatory bowel disease,diabetes,andobesity),and for rational translation to human health gains.(Translational Research2012;160:246–257)Abbreviations:GI¼gastrointestinal;IBD¼inflammatory bowel disease;IBS¼irritable bowelsyndrome;TRFLP¼terminal restriction fragment length polymorphism;UC¼ulcerative colitisFrom the Division of Gastroenterology and Hepatology,Mayo Clinic, Rochester,Minn;Division of Gastroenterology and Hepatology, Department of Internal Medicine,University of Michigan Medical Center,Ann Arbor,Mich;Division of Biomedical Statistics and Informatics,Department of Health Sciences Research,Mayo Clinic, Rochester,Minn;Department of Medicine,Division of Gastroenterology and Hepatology,Department of Microbiology and Infectious Diseases,Faculty of Medicine,University of Calgary, Calgary,Alberta,Canada.The authors have nofinancial disclosures relevant to this article.Submitted for publication December6,2011;revision submitted May 8,2012;accepted for publication May8,2012.Reprint requests:Kevin P.Rioux,University of Calgary,1863Health Sciences Centre,3330Hospital Drive NW,Calgary,Alberta,Canada T2N4N1;e-mail:kprioux@ucalgary.ca.1931-5244/$-see front matterÓ2012Mosby,Inc.All rights reserved.doi:10.1016/j.trsl.2012.05.003246Humans live in a biosphere where microbes are ubiqui-tous and have existed and evolved over3.8billion years.1 The intestinal tract of humans harbors a complex micro-bial community estimated to contain approximately100 trillion cells,exceeding the number of human cells by a factor of10.2-4This complex community of microbes in the alimentary tract(bacteria,archaea,eukarya,and viruses)is also known as the‘‘gastrointestinal(GI) microbiota.’’5,6Akin to the human genome,which is the sum of all human genes,the term‘‘microbiome’’refersto all of the microbiota in a defined microbial community,which are usually differentiated by their genetic elements.7Metagenomics refers to the functional and compositional analysis of an assemblage of microbes based on molecular study of their collective genomes.8,9 The gut microbiota performs a variety of beneficial functions(Table I),and given their importance in human health and disease,the Human Microbiome Project was launched by the National Institutes of Health to(1) characterize the microbial communities of various niches of the human body(eg,the nasal passages,oral cavity,skin,urogenital system,and GI tract),(2)to de-termine whether individuals share a core human micro-biome,and(3)to explore whether changes in the human microbiome cause or correlate with human disease.10,11 The distal GI tract contains the most abundant and diverse communities of microbes,in continuous interplay with the human host resulting in both local (mucosal and luminal)and systemic(metabolic and nutritional)effects.Therefore,of all microniches being explored by human microbiome researchers,the GI tract holds the most promise for discovery of important new concepts and understanding of the human‘‘superorganism’’and translation to clinical biomarkers and therapies.12There are excellent reviews that have focused on the temporal and spatial development of the human gut mi-crobiota13and their role in health and disease.14The goal of this article is to highlight the current state of knowledge of the human gut microbiome,discuss tech-nical and practical limitations of scientific devices and tools,and map out some of the knowledge gaps in this challengingfield that will be solved by thoughtful scien-tific approaches,advanced sequencing and computa-tional technology,and persistence.TECHNIQUES FOR ANALYZING MICROBIOTA Studies in the1970s using anaerobic culture–based techniques identified more than400to500distinct bac-terial species in the human gut.15Studying the human GI microbiota by cultivation methods has many draw-backs:It produces selective growth of some organisms and thus distorts composition of the natural community.Moreover,approximately60%to80%of gut microbes simply cannot be grown by conventional in vitro tech-niques.16,17In1977,Woese and Fox18described a tech-nique for molecular characterization of bacterial phylogeny based on ribosomal RNA sequence analysis. In particular,the16S rRNA is a molecule that is univer-sally present in bacteria and has highly conserved do-mainsflanking hypervariable sequences that can be used to distinguish bacterial groups.In the early 1980s,various culture-independent molecular tech-niques based on the16S rRNA became available,and during the last3decades they have been used extensively to assess the structure of complex or fastid-ious prokaryotic communities.1Examples of these techniques are terminal restriction fragment length polymorphism(TRFLP),denaturing gradient gel elec-trophoresis,andfluorescent in situ hybridization.19 These bacterial community profiling or‘‘fingerprint-ing’’techniquesfirst involve isolating bacterial DNA from environmental or biological samples.The DNA is amplified by polymerase chain reaction using univer-sal primers that target conserved regions of the16S rRNA gene,and the resulting amplicons contain vari-able regions that discern the constituent members of bacterial communities by electrophoretic or hybridiza-tion techniques.Cloning and then sequencing of the16S rRNA gene in an automated capillary sequencer is a higher-resolution method of studying bacterial phylogeny.20This tech-nique uses Sanger sequencing to produce a long read ( 800base pairs)of the16S rRNA gene,which enables identification of bacteria at a higher-level phylogenetic resolution(ie,genera and species).This relies on robust bioinformatics tools,such as the Ribosomal Database Project.21In the wake of the Human Genome Project,next-generation sequencing technologies have emerged that have increased the depth and speed of phylogenetic cov-erage and decreased the cost through massively parallel sequencing methods.There are various commercial platforms for next-generation sequencing.One of these newer techniques is pyrosequencing that identifies nu-cleotides by the amplitude of light signals generated when luciferin is converted to oxyluciferin during Table I.Biological effects of the gut microbiota on human hostDevelopment of innate and adaptive immunityIntestinal epithelial integrityEnergy sourceVitamin biosynthesis,bile salt transformation,catabolism of dietary glycans(eg,cellulose and pectins)Barrier to colonization by microbial pathogensXenobiotic metabolismTranslational ResearchVolume160,Number4Dave et al247DNA synthesis.22A limitation of this technique in its first iteration was short sequence reads of100to250 base pairs,which have been quickly surpassed by newer methods.There are several companies that manufacture these machines(eg,Roche454[Roche Diagnostics Corp,Indianapolis,Ind],Illumina/Solexa[Illumina Inc,San Diego,Calif],Applied Biosystems,Foster City,Calif).Although the16S rRNA gene sequences obtained by Sanger or next-generation sequencing en-able greater depth and resolution to identify and discern taxa,the identity and thus functional importance of community members may be missed because of a sub-stantial proportion gut microbes match ribosomal se-quences from uncultured bacteria.Also,there is currently no clear consensus on what constitutes a bacte-rial species at a molecular level,23so alternative methods of describing‘‘phylotypes’’are based on oper-ational taxonomical units,groups of sequences that are highly similar to a single reference sequence.24 Despite the enormous progress made in determining microbial community structure using the16S rRNA gene,the basic technique provides no information about bacterial physiology and ecological significance.1An-other novel approach to assess the diversity of microbes is the whole genome shotgun sequencing of community DNA.25This differs from16S RNA sequencing tech-niques in that entire genomes of microbes are sequenced and compared with previously characterized genes to build a picture of functional capability of the micro-biota.This approach helps not only with identification of rare species in a complex community but also with identification of microbial genes that code for metabolic or biologic functions.12The drawback with this tech-nique is that a large amount of DNA is needed(although it can be overcome by whole genome amplification), contamination with host DNA is problematic,and many genes are identified that do not yet have a known function.Hamady and Knight26have published a com-prehensive review of various sequencing techniques used in microbiome investigation.SEQUENCE DATA ANALYSIS AND CHALLENGESAs mentioned earlier,there are2main approaches to surveying the microbiome,16S rRNA sequencing and whole genome sequencing(Fig1).The former uses a small representative region as a marker or proxy and involves evaluating the informative sites of any of the 16S rRNA variable regions.27Similar organisms are represented by clustered reads and used to infer the phy-logenetic and taxonomic identity.The proportion of var-ious types of organisms is used to infer the structure of microbial communities using statistical analysis.The latter approach of assembly involves splitting the DNA into smaller pieces and sequencing them to later assemble into longer contigs.Those are then used to in-fer functional and metabolic pathway information.28 There are huge challenges in analyses of these data. All the next-generation sequencing technologies have improved vastly but still have higher error rates than tra-ditional Sanger sequencing.There are also systematic sources of error depending on the sequencing instru-ment used that can yield noisy data.29,30Qiime31and Mothur32are popular analysis tools for microbial se-quencing data,but there are many pre-and post analysis steps that have a bearing on the eventual results.It is thus vital to have quality checks and data-cleaning steps along the analysisflow to ensure the validity of the end results.The alignment of these short reads,especially to similar sequences,is error prone.A one-off read sup-porting evidence for a particular rare organism or group should thus be treated with caution.Some studies have come up with a leave-one-out analysis to recommend a minimum number of sequence-reads mapping to a par-ticular feature to be confident of the result.33These anal-yses are continually improving as the availability of complete microbial genomes increases.This would mean a larger number of reference genomes that can be used for accurate and efficient bioinformatics analy-sis and better functional interpretation of data.The current tools use traditional statistical techniques with some refinements that do not take into account the inherent complexity in biological systems and are lim-ited by the assumption that predictor variables(ie,gut microbes)are independent of each other.This is clearly not the case,because the human gut microbiota is a com-plex community whose constituents are linked together through complex homeostatic interactions.The solution to deciphering this complexity could be through the use of machine-learning algorithms that include neural net-works,support vector machines,and decision trees.Ma-chine learning is inherently more suited to the study of complex microbial communities because it uses pattern recognition,does not need identification of predictor variables in advance,and becomes increasingly better at prediction with larger volumes of data.34Another problem facing investigators is the huge volume of data that sequencing generates,which can overwhelm available computing resources of an institution.Cloud computing uses the storage and processing capabilities of a network of remote servers hosted on the internet, providing individual investigators paid access to power-ful computing resources suited to the massive data gen-erated by current sequencing methods.The Cloud BioLinux and Amazon Elastic Compute Cloud(http:// /ec2/)are examples of2currently available commercial resources for metagenomic anal-yses.35In addition,the Data Intensive Academic GridTranslational Research248Dave et al October2012is a free computational cloud sponsored by the National Science Foundation and available to researchers in aca-demic and nonprofit institutions.SAMPLING THE GUT MICROBIOTAAlthough the same bacterial phyla predominate in the stomach,small intestine,and colon,their relative com-position and abundance varies considerably.36Meth-odologic factors likely account for some of this variation,including patient selection,sample handling,and choice of molecular and bioinformatics techniques.Next,we provide data on some of the issues that may be confounding human gut microbiota studies.These con-cepts are summarized in Table II .1.What constitutes a normal,healthy human micro-biota?It has long been apparent that the intestinal micro-biota varies significantly from one individual to another,making it impossible to define the ‘‘normal’’or ‘‘healthy’’human gut microbiota.Indeed,it is difficult to define what constitutes a healthy human,and as-sumed absence of overt disease may not be the most re-liable indicator of a healthy microbiota for study purposes.Although it will be a difficult task to accom-plish,future studies could use a set of phenotypic char-acteristics,for example,the Adult Fitness Test launched as a part of President’s Challenge (http://www.adultfi/).This test incorporates aerobic fit-ness,muscular strength,endurance,flexibility,and body composition to determine overall health-related fitness.A ‘‘core microbiota’’has been proposed,37but it re-mains unclear what the essential constituents are,and,instead,a set of core functions of the microbiota may well emerge as the correct unifying concept.38Age has well-described influences on the fecal microbiota,for example,with substantial differences being de-scribed among healthy infants,adults,and the el-derly.39-42Gender differences have also been described.40On a collective scale,it is also apparent that the human gut microbiota differs between various ethnic groups,which likely reflects both genetic and en-vironmental influences.40Environmental influences that likely affect the GI microbiota are wide-reaching,dy-namic,and difficult to control for in the context of human studies.These include diet,medications (espe-cially antibiotic exposure,even if remote),stress,smok-ing,and GI infections.Early studies suggested that the intestinal microbiomes of healthy adults were stable over time,43,44but such studies generally used fingerprinting techniques with low sensitivity and relatively few points of observation over a short period of time.More recent studies using deep sequencing techniques have shown a surprising degree of species-level variability within an individual’s fecalFig rmatics analysis.Translational Research Volume 160,Number 4Dave et al 249microbiota over a short time span of days to weeks,with an individual core microbiota of perhaps only 10%re-maining unchanged in the long term.45This has obvious implications for basic and clinical studies in humans,because our new understanding of the human gut micro-biome as a moving target makes it difficult to ascribe significance to shifts in the microbiome in the presence of significant background temporal variation.2.Is fecal microbiota representative of the resident microbiota of the distal GI tract?A majority of the studies of the human gut micro-biome have analyzed the microbiota of human fecal specimens,given the ease of acquisition,minimal cost,and avoidance of invasive procedures such as colo-noscopy with requirement for fasting and laxative prep-aration.It is widely believed,however,that the highly evolved biofilm communities that are closely associated with the intestinal epithelium are biologically more rel-evant than planktonic microbes that exist in the lumen of the gut or associated with food residue,and,moreover,fecal specimens may not accurately portray the mucosal microbiota.Even within stool,there appears to be sig-nificant differences between microbiota of the liquid fraction compared with that associated with the solid phase (insoluble plant residue and mucin).46Lepage et al 47used a bacterial small subunit RNA fingerprinting technique to demonstrate that the dominant bacterial groups differ between fecal and mucosa-associated mi-crobiota in patients with inflammatory bowel disease (IBD)and healthy controls.In a deep sequencing study of 3healthy individuals,mucosal tissue samples from the cecum,ascending colon,transverse colon,descend-ing colon,sigmoid colon,and rectum were compared with fecal samples taken 1month after colonoscopy.48This study showed that the mucosa-associated micro-biota was different from stool microbiota within an in-dividual,and the authors hypothesized that the fecal microbiota represents a combination of shed mucosal bacteria and the nonadherent luminal population.In ad-dition,the investigators showed that mucosa-associated microbiota had a patchy and heterogeneous distribution within a subject.Rather than a subset of the fecal micro-biota,the mucosa-associated microbiota is composi-tionally distinct from that of stool.49Marteau et al 50compared cecal luminal contents (obtained by fluoro-scopic placement of an orocecal tube)with fecal sam-ples using both culture-based and culture-independent techniques,showing that the fecal flora was different from the cecal flora both qualitatively and quantita-tively.3.What is the optimal method of sampling mucosa–associated microbiota?In view of the limitations of studying stool specimens,investigators have turned to mucosal biopsies of the GI tract collected by endoscopic means as the source spec-imens for microbiome studies.However,endoscopic bi-opsies also have important limitations because fasting and colon cleansing are common prerequisites of endo-scopic biopsy collection,and tissue specimen them-selves are contaminated by luminal microbes in the process of endoscopic collection.During colonoscopy,for example,biopsy forceps are advanced and then with-drawn via the same channel used to suction stool resi-due.To determine the degree of contamination ofTable II.Technical and methodological challenges to studying the human gut microbiotaTechnicalContamination during sample collectionBiases due to varying efficiency of bacterial nucleic acid extractionInherent polymerase chain reaction biases leading to under-representation or failed detection of minor phylotypes 16S rRNA gene does not accurately reflect bacterial abundance (varying copy numbers)Sequencing errorsMolecular techniques cannot discern microbes as dead or alive,autochthonous or allochthonousMethodologicalBroad definitions of healthy human subject Lack of correlative host genotype dataConfounding influences of diet and drugs largely unaccounted forEffect of fasting and pre-colonoscopy bowel cleansing on relevant microbiotaValidity of mucosal biopsies as representation of ‘‘mucosa-associated’’microbiota Deep sequencing vs fingerprinting techniques How is operational taxonomic unit defined Moving target:temporal instabilityMajority of metagenomic studies to date have not yet assessed functional aspects of microbiome Statistical tools for devising adequately powered clinical studies are limitedTranslational Research250Dave et alOctober 2012biopsies during endoscopic collection,we deviseda study comparing standard and sterile sheathed biopsyforceps.51Sheathed forceps were constructed by cover-ing standard endoscopic biopsy forceps with polyethyl-ene tubing.Paired biopsies using standard and sheathedforceps were obtained from the terminal ileum of sub-jects,and mucosa-associated bacteria were character-ized by16S-rDNA polymerase chain reaction andTRFLP.Our study showed no difference in the micro-biota between specimens collected with standard or ster-ile sheathed biopsy forceps within individual subjects.In a unique study by Mai et al,52the fecal microbiotawas studied before and up to8weeks after colonoscopyin a small series of healthy individuals undergoing coloncancer screening.In some patients,there were persistentalterations in the fecal microbiota after colonoscopy,presumably due to the effects of fasting and the unspec-ified colon-cleansing agent.No similar studies havebeen published to replicate these preliminary data orto extend thefindings to a variety of common-use bowelpreparations or to the mucosa-associated microbiota.4.Do sample storage conditions and method of DNAextraction matter?As part of ongoing microbiome studies,samples arebeing collected from subjects and archived in biobanks.A relevant concern is whether storage conditions affectthe diversity of gut microbiota.A16S rRNA pyrose-quencing study showed that short-term storage of upto14days at20 C,4 C,220 C,or280 C did not sig-nificantly affect the microbial composition.53Likewise,Wu et al54showed that there was no significant differ-ence in bacterial composition determined by pyrose-quencing in fecal samples immediately frozen at 280 C or those stored on ice for24or48hours.54Other investigators have revealed concern about the stabilityof stool specimens stored at room temperature,particu-larly in excess of12hours.55The exact method of DNAextraction also seems to be important,with an initialbead-beating step improving the performance of bothphenol-based protocols54and proprietary DNA extrac-tion kits.56Bead beating refers to mechanical disruptionof bacterial cell walls by vigorous mixing with tinyglass beads and improves extraction of cytosolic com-ponents from difficult to lyse bacteria including Firmi-cutes,which are a major component of the human gutmicrobiota.COMPOSITION OF THE GUT MICROBIOTAIt is estimated that up to80%of the bacteria in the hu-man gut cannot be cultivated by conventional tech-niques,largely because of their fastidious nutrient andanaerobic requirements and their complex dependence on one another.17Molecular techniques have signifi-cantly advanced our understanding of the constituentmembers of the human microbiome and have largelysupplanted cultivation-based techniques for study ofthe gut microbiome.The major members of bacterialcommunities in various segments of the human GI tractare illustrated in Figure2.The most diverse and abun-dant microbial communities generally occur in theoral cavity and distal GI tract and have been the focusof most studies,because they are most amenable to sam-pling.However,the relatively simple indigenous micro-biota of the human esophagus,stomach,and smallbowel are relatively unstudied,and these niches presentthe added challenge of trying to discern allochthonousbacteria(‘‘passersby’’from upstream niches that arelikely irrelevant)from autochthonous microbes(repre-senting stable,functionally relevant community‘‘resi-dents’’).The oral cavity contains members of thephyla Firmicutes,Proteobacteria,Bacteroidetes,Acti-nobacteria,and Fusobacteria,which account for99%of all phyla present.The rare phyla belong to SR1,TM7,Cyanobacteria,Spirochaetes,Tenericutes,andSynergistetes.57A pyrosequencing study of samplesfrom distal esophagus showed the presence of membersof6phyla,Firmicutes,Bacteroides,Actinobacteria,Proteobacteria,Fusobacteria,and TM7.The bacteriaStreptococcus,Prevotella,and Veillonella were foundto be the most prevalent,and the community of the distalesophagus was similar to that of the oral cavity58;how-ever,the majority of esophageal organisms could be cul-tivated,unlike those of the oral microbiome.Given theextremely low pH in the stomach,it was originally be-lieved that,with few exceptions(eg,Helicobacter py-lori),the stomach harbored a few transient microbial species and did not have a complex community likethe distal GI tract.3However,in a study of gastric biopsyspecimens from23human subjects using the16S rRNAgene clone library construction,Bik et al12showed thatthere is a diverse community of gastric microbes domi-nated by members of phyla Proteobacteria,Firmicutes,Actinobacteria,Bacteroidetes,and Fusobacteria.Thismicrobial community was significantly different thanthe oral and esophageal community.Another interestingfeature of this study was that the composition of the bac-terial community was not apparently altered by the pres-ence of H.pylori.The microbiota of the human small bowel is relativelyunstudied.By using TRFLP to study samples obtainedfrom3individuals during autopsy,one group showedthat the microbiota of the jejunum and ileum was lesscomplex than that of the colon.The jejunal and ilealsamples contained more facultative anaerobes and noClostridium coccoides and leptum subgroups,in con-trast to cecum and rectosigmoid samples.59Translational ResearchVolume160,Number4Dave et al251Microbial abundance is greatest in the colon with ap-proximately 1011and 1012microbial cells per gram of stool,and the number of microbial genes is 2orders of magnitude greater than that contained by the human host genome itself.2Recent studies of human fecal and colonic biopsy specimens have revealed members of 9distinct phyla (Firmicutes,Bacteroidetes,Actinobacte-ria,Fusobacteria,Proteobacteria,Verrucomicrobia,Cy-anobacteria,Spirochaetes,and VadinBE97).5,48The phyla Firmicutes and Bacteroides are the dominant phyla in the colon.10,38,48In contrast,in the total biosphere there are up to 70bacterial phyla,which highlights the fact that the human GI microbiota is restricted to a small subset of phyla,but with a high degree of species richness and abundance within these core constituent phyla.By using the Illumina Genome analyzer platform,the Metagenome of Human Intestinal Tract Consortium as-sessed the metagenome of fecal specimens obtained from 124healthy patients,overweight patients,obese patients,and patients with IBD from Denmark and Spain.38This is the most comprehensive study of human microbiome from a large group of subjects to date.The investigators were able to estimate that human gut mi-crobiota harbors approximately 1150bacterial phylo-types or species.In addition,they noted 536,112unique genes in each sample,of which 99%were bacte-rial.Approximately 40%of the bacterial genes from each individual were shared with at least half of the in-dividuals of the cohort.These highly conserved genes are involved in degradation and digestion of complex sugars,production of short chain fatty acids,and bio-synthesis of vitamins.Although significant interindivid-ual differences exist in the composition of the gut microbiome of humans,there is an essential functional-ity of gut microbes that support human health and nutri-tion.Thus,the concept of the ‘‘human superorganism’’has emerged,a physiology and homeostasis that are the complex sum of human and microbial gene expres-sion.10One of the goals of the human microbiome project is to ascertain whether we have a core group of microor-ganisms that are shared between individuals.Evidence from recent studies suggests that,rather than acoreFig 2.The dominant phyla in various locations in the human GI tract.By permission of Mayo Foundation for Medical Education and Research.All rights reserved.(Color version of figure is available online.)Translational Research252Dave et alOctober 2012。
与肠道菌群息息相关的七类病症作者:安利来源:《百科知识》2019年第12期人的肠胃中寄居着多种多样的微生物,这些微生物被称为肠道菌群。
人一出生就继承了从母体带来的一些肠道菌群,周围环境的细菌也会成为肠道菌群的一部分。
每个人的肠道中都有500~1000个不同种类的肠道菌群,总数大约有10万亿。
近年来,肠道菌群成为研究的热点,人们惊奇地发现,这些存在于肠道的微生物与睡眠、肥胖、炎症、癌症、心血管疾病、神经退行性疾病等生理病理过程息息相关,甚至连大脑也逃脱不了肠道微生物的“控制”。
一般认为,高血压与遗传、基因、生活习惯有关,但现在有研究发现,高血压的发生与肠道细菌密切相关,这也能解释为什么高血压会呈现出家族性发病现象。
人们已經知道高盐饮食会增加高血压风险。
动物模型显示,两周的高盐饮食就会令小鼠某些肠道菌群急剧减少,其中尤以乳酸杆菌属细菌关联性最强。
乳酸杆菌属细菌会抑制一种与炎性反应相关的细胞—TH17细胞。
这说明,高盐饮食是通过减少肠道乳酸杆菌属细菌的数量来破坏机体的免疫平衡,导致高血压患病风险提高。
肠道菌群可以影响人体糖类及能量的吸收,而肠道菌群紊乱导致的肠道屏障功能缺失会为细菌内毒素的入侵提供“通道”,诱发慢性炎症反应及胰岛素抵抗—2型糖尿病的标志。
1型糖尿病是一种自身免疫疾病,同样受到肠道菌群紊乱的影响。
研究发现,将患1型糖尿病的人与健康的人进行对比,其肠道菌群很早就表现出了明显的差异,最早可体现在4个月左右的婴儿期。
肠道细菌或对肠癌负有责任。
研究发现,肠癌患者的肠道中通常拥有较高水平的特定大肠杆菌菌株。
大肠杆菌可分为几个亚型,其中一个亚型的大肠杆菌能产生一种被称为colibactin 的基因毒性物质,能够损伤肠道细胞中的DNA,而DNA损伤被认为具有致癌性。
从另一方面看,可以利用肠道菌群来对抗肿瘤,一种潜在的疗法是粪便移植,即用健康人群的肠道微生物占据癌症患者的肠道。
一种名为TET2的基因突变是白血病的诱因。
Mystery奥秘38肠道菌群你所不知的那部分“自己”人体肠道内的细菌数量,是人体细胞总数的10倍!从数量论,肠道菌群才是主要的那部分“自己”。
你,了解这部分的“自己”吗?你是你自己么?拿这个问题去问别人,估计被问到的人都会一愣,然后答道“我当然是我自己啦!”的确,在大多数时候,人总是被视作一个独立、统一、完全的个体。
然而,生物学和医学研究却发现,其实人体并非是一个统一的整体,在人体内部,还有一些与人体在生物学上截然不同而又有紧密联系的生物群体。
这个生物群体,是由大量微生物共同组成的。
其中最重要也是最有名的,就是肠道菌群。
身体内的另一个自己在微生物学诞生后不久,人们就发现,在动物的消化道中存在有不少微生物。
例如在牛、羊、兔等食草动物的胃或盲肠中,就存在大量以细菌为主的微生物群体。
由于在食草动物摄入的植食性饲料中,纤维素、半纤维素等多糖难以依靠动物体自身分泌的酶液消化,而微生物群体中包含的纤维素消化菌、半纤维素消化菌等可以较好地将多糖转化为低聚糖和寡糖,从而促进对这些营养物质的吸收。
随着医学的发展,人们也注意到,在人类的肠道,尤其是结肠(也就是平常所说的大肠)中,也存在着大量微生物。
这些以细菌为主的微生物种类极多、数量极大。
据推测,一个正常成人体内,肠道内的细菌总重量可达1~1.5kg,包含的细菌数量则可以达到1.0×1014个。
而一个成年人自身的细胞数量为1.0×1013个,也就是说,居住在我们肠道内的细菌数量,是人体细胞总数的10倍!在我们每天排出的粪便中,干重量的50%以上是由这些细菌及其“尸体”构成的。
因此曾有人风趣的说,从数量上来看,我们人类并不应该被称为人类,而应被称作细菌。
如此庞大的细菌群体驻扎在肠道内,构成了一个极为复杂的集体。
这个集体,就被称作肠道菌群。
肠道菌群并非是生来就有的,它们实际上是“外来户”。
在母体子宫内,胎儿所处的是一个几乎无菌的环境,因此胎儿肠道内也是无菌的。
肠道菌群分类学-概述说明以及解释1.引言1.1 概述肠道菌群是指人体肠道内存在的大量微生物的群落,包括细菌、真菌、病毒等。
这些微生物与人体共同生活并相互作用,对人体健康和疾病发展起到重要影响。
近年来,肠道菌群的研究成为微生物学和生物医学领域的热点之一。
科学家们发现,肠道菌群与人体健康密切相关,它们不仅参与了人体的消化、吸收和代谢等重要生理功能,还与免疫调节、心理健康、肥胖、炎症性肠病等多种疾病的发生和发展密切相关。
随着DNA测序技术的快速发展,科学家们能够更好地理解肠道菌群的复杂性。
肠道菌群的分类方法也在不断完善和更新。
根据16S rRNA序列或者宏基因组测序分析肠道微生物可以将其分为不同的菌群,并研究它们的功能和相互关系。
本文将深入探讨肠道菌群的分类学,包括肠道菌群的定义和意义、分类方法、常见的菌群分类以及最新的研究进展。
通过对肠道菌群分类的深入研究,我们可以更好地理解和解释肠道菌群在人体健康与疾病中的作用机制,为未来的微生物相关疾病的预防和治疗提供理论依据。
然而,肠道菌群分类研究还存在一些局限性和不足之处,例如技术手段的限制、样本数量的有限以及肠道菌群与人体宿主相互作用的复杂性等。
对这些问题的深入研究和解决将是未来肠道菌群分类学的发展方向。
综上所述,本文旨在全面介绍肠道菌群分类学的相关内容,通过对已有研究的总结和对未来研究的展望,希望能够推动肠道菌群分类学的发展,为人类健康提供更有效的策略和方法。
1.2 文章结构文章结构部分的内容应包括对整篇文章的组织和分段的介绍。
可以使用以下内容作为参考:文章结构部分:本文主要分为引言、正文和结论三个部分。
引言部分包括概述、文章结构、目的和总结四个小节。
在概述中,将介绍肠道菌群分类学的重要性和研究现状。
在文章结构中,将简要说明本文的组织结构,明确每个部分的内容。
在目的中,将说明本文的写作目的和研究问题。
在总结中,将给出引言部分的总结和本文的意义。
正文部分主要包括肠道菌群的定义和意义、肠道菌群的分类方法、常见的肠道菌群分类和肠道菌群分类的研究进展四个小节。
肠道菌群与代谢疾病关系的研究我们的肠道内有着数量众多的细菌、病毒和真菌等微生物,这些被称为肠道微生物群(gut microbiota)。
在这个微生物群体中,细菌是占主要成分的,它们能够进行堆积物质的转化、代谢和腐烂,进而为肠道环境提供能量和维持病原体抑制的作用。
早期的研究表明,肠道微生物群可以对人体的代谢过程和整体健康状况产生重大影响。
不断发展的技术使科学家能够对肠道微生物群进行深入的研究。
其中,代表性的方法包括16S RNA测序、代谢组学(metabolomics)以及功能性分析。
这些方法允许科学家从微生物水平探索宿主和微生物之间的相互作用,并推动了肠道微生物群如何影响宿主健康的研究。
越来越多的研究表明,肠道微生物群可以影响体内多种主要代谢路径,包括甾体(steroid)、氨基酸、磷脂、胆汁酸、一氧化氮等代谢通路。
这些与微生物群相关的代谢变化可能导致冠心病、高血压、糖尿病、脂肪肝等代谢疾病发生。
虽然我们还了解不足,但科学家们致力于探究在这些代谢疾病和微生物群之间建立更加明确和完整的关系。
一方面,肠道微生物群可以产生干扰素厌恶素(interferon-gamma),这是一种与免疫反应相关的蛋白质,对于改善脂肪肝和降低糖尿病等疾病的风险有着显著的疗效。
异常的肠道细菌组成或干扰素厌恶素水平可能会导致慢性代谢疾病的发生。
另一方面,对比不同地域的人群,我们可以观察到寿山茶(Lapsang Souchong tea)和普洱茶(Pu-erh tea)的食用和持续性运动能够影响肠道微生物组成。
依据相关的研究发现,寿山茶和普洱茶的单味茶能够降低肝脏脂质沉积、降低胰岛素抵抗、增加大肠燃烧能力等作用。
除此以外,运用类维生素、医药代谢组学等方法,科学家们正在进行一系列研究,以鉴定和量化肠道微生物群对此类慢性代谢疾病的影响和作用机制,进而帮助人们掌握如何通过微生物组群的管理来进行健康改善。
应该指出,尽管存在许多诱发慢性代谢疾病和肠道菌群变化的因素,尤其是不健康的饮食和生活方式。
人体肠道菌群组成
人体肠道菌群是指寄生在人体肠道内的微生物群落。
它由许多种不同的微生物组成,主要包括细菌、真菌、病毒和原生动物。
其中,细菌是菌群中最主要的成员,对人体健康发挥着重要作用。
人体肠道菌群主要包含以下几个主要类群:
1. 双歧杆菌属(Bifidobacterium):主要参与碳水化合物的分
解和吸收,促进益生菌的生长,调节肠道免疫和维持肠道屏障功能。
2. 乳酸菌属(Lactobacillus):产生乳酸和其他有益物质,帮
助抑制有害菌的生长,维护肠道酸碱平衡。
3. 肠球菌属(Bacteroides):参与蛋白质、脂类、类脂和多糖
的分解,产生抗菌物质,对维持肠道内环境稳定具有重要作用。
4. 菌群中的其他细菌:包括厚壁菌属(Clostridium)、变形菌
属(Proteus)、肠杆菌属(Escherichia)等。
此外,人体肠道菌群中还存在一些真菌和病毒,如毕氏酵母菌(Candida albicans)、肠炎病毒、腺病毒等。
它们在一定程度
上参与肠道生态平衡的维持和调节。
人体肠道菌群的组成在每个人身上是不完全相同的,受到多种因素的影响,包括遗传、生活环境、饮食习惯、使用抗生素、感染疾病等。
人体肠道菌群的平衡与人体健康息息相关,不平衡的肠道菌群组成可能会引发多种疾病,如肠道炎症、肠易激综合征、自身免疫疾病等。
因此,维护肠道菌群的平衡对于维持人体健康非常重要。
微生物与人类健康:肠道菌群的奥秘
微生物与人类健康的关系日益受到科学界的关注,其中肠道菌群的作用尤为突出。
肠道菌群是指生活在人体肠道内的微生物群落,包括细菌、古菌、真菌和病毒等。
这些微生物不仅数量庞大,种类繁多,而且与人体的健康状况息息相关。
肠道菌群对人体健康的影响是多方面的。
首先,它们参与食物的消化吸收过程,帮助人体分解复杂的碳水化合物、蛋白质和脂肪,从而提供能量和营养物质。
其次,肠道菌群能够合成多种维生素和必需氨基酸,这些物质对人体的新陈代谢至关重要。
此外,肠道菌群还参与免疫系统的调节,帮助人体抵御病原体的侵袭,维持肠道屏障的完整性。
肠道菌群的失衡,即肠道菌群失调,可能导致多种健康问题。
例如,肥胖、糖尿病、心血管疾病、炎症性肠病、甚至神经系统疾病等都与肠道菌群的失衡有关。
研究表明,通过调整饮食结构、使用益生菌和益生元等方法,可以改善肠道菌群的组成,从而预防和治疗这些疾病。
近年来,随着宏基因组学、代谢组学等技术的发展,我们对肠道菌群的认识越来越深入。
科学家们正在探索肠道菌群与人体健康之间的复杂关系,并试图开发新的治疗方法。
例如,通过粪便微生物移植(FMT)来治疗某些肠道疾病,已经成为一种新兴的治疗手段。
总之,肠道菌群是人体健康的重要组成部分,它们的奥秘正逐渐被揭开。
未来,随着研究的深入,我们有望更好地利用肠道菌群来维护和促进人类健康。
Impact of Targeted Specific Antibiotic Deliveryfor Gut Microbiota Modulation on High-Fructose-FedRatsPrasant Kumar Jena &Shilpa Singh &Bhumika Prajapati &G.Nareshkumar &Tejal Mehta &Sriram SeshadriReceived:26August 2013/Accepted:3February 2014/Published online:27February 2014#Springer Science+Business Media New York 2014Abstract The objective of present investigation was to study the effect of gut microbiota alteration by oral administration of targeted delivery of pH sensitive cefdinir microspheres to high-fructose-fed (HFD)rats.Rats were fed with a high-fructose diet with or without cefdinir microsphere administration for 30days.The fecal microbiota community,oral glucose tolerance,the markers of liver injury,plasma and hepatic lipids profile,and histological evaluation were investigated.The levels of blood glucose,liver injury markers,lipid profile in plasma and liver,and fat tissue were significantly increased in high-fructose-fed rats.However,after pH-sensitive cefdinir microsphere administration,the elevation of these pa-rameters was significantly suppressed.Cef EL significantly lowered the increased AST (p <0.05)and ALT (p <0.001)levels in HFD group.There is a significant lower (p <0.01)AUC glucose level in Cef EL group than HFD group The histological changes in the liver and the small and large intestines were more profound in HFD group as compared to cefdinir-treated HFD and control groups.Feeding of cefdinir microsphere sustained lactobacilli and bifidobacteria and significantly decreased (p <0.05)the number of Enterobacteriaceae induced by HFD.Experimental evidences demonstrated that the effectiveness of pH-specific cefdinir microsphere on reducing insulin resistance and development of metabolic changes in high-Appl Biochem Biotechnol (2014)172:3810–3826DOI 10.1007/s12010-014-0772-yElectronic supplementary material The online version of this article (doi:10.1007/s12010-014-0772-y)contains supplementary material,which is available to authorized users.P.K.Jena :S.Singh :B.Prajapati :S.Seshadri (*)Institute of Science,Nirma University,Sarkhej-Gandhinagar Highway,Chandlodia,Ahmedabad 382481Gujarat,Indiae-mail:sriram.seshadri@nirmauni.ac.inSriram.Seshadrie-mail:sriramsjpr@G.NareshkumarMolecular Microbiology and Biochemistry Laboratory,Department of Biochemistry,M.S.University of Baroda,Vadodara,Gujarat,IndiaT.MehtaDepartment of Pharmaceutics,Institute of Pharmacy,Nirma University,Ahmedabad,Gujarat,Indiafructose-fed rats and suggested that it may be a promising therapeutic agent in treating type2 diabetes.Intestinal-targeted antibiotic delivery needs to be further explored for its therapeutic applications.Keywords Cefdinir.Microspheres.Fructose.Gut microbiota.Diabetes.Inflammation IntroductionDiabetes is the most common endocrine disorder,and the number of people with diabetes in2011has reached a surprising371million.Around4.8million deaths are because of diabetes,and300million will be later added by2025[1].Obesity,type2 diabetes,and hyperlipidemia often coexist and associate with significantly increased morbidity and mortality[2].A significant increase in total refined carbohydrate intake, fructose has paralleled recent increase in incidence of obesity and diabetes.Metabo-lisms of sugars,particularly fructose,occurs mainly in the liver,and high-fructose flux leads to enhanced hepatic triglyceride accumulation resulting in impaired glucose and lipid metabolism and increased proinflammatory cytokine expression[3].Persistent fructose consumption shows to decrease insulin sensitivity,increases inflammation,oxidative stress,and pancreatic islet dysfunction,and promotes dyslip-idemia,which may increase the risk for development of type2diabetes[4].Type2 diabetes(T2D)processes by impaired glucose tolerance(IGT),where the metabolic and endocrine changes take place,which can effectively prevented or even delayed through lifestyle changes or drug treatment[5].Because of the increasing incidence of T2D,there is an extensive need to find new effective strategies for type2diabetes prevention and/or treatment,including the potential use of nutritional supplements.The mammalian host colonizes by trillions of microbes,which inhabit the gastro-intestinal(GI)tract,predominantly in symbiotic relationship to their host[6].Several lines of evidence suggest that dietary factors might profoundly influence gut micro-biota composition.Switching to a high-fat diet resulted in a reduction of Bacteroidetes,while the figures of Firmicutes and Proteobacteria had been increased [7].Importantly,this was observed both in the presence and absence of obesity,which clearly suggests that diet must be considered as a confounding factor affecting microbial composition.A change in the diet(i.e.,from a low-calorie fat plant polysaccharide to a high-fat sugar-rich diet)shifted the gut microbiota structure within a single day,along with the changes in major metabolic pathways in the microbiome. Community population and function of the microbiota can change due to various ways,including antibiotic treatment,inflammation,or changes in diet pattern[8]. Antibiotic therapy not only target pathogen but also to commensal inhabitants of the human host.The degree of the impact on non-target microbial populations depends on the specific antibiotic drug used,its mode of action,and the degree of resistance within the community[9].Extended loss of the distinctive composition of gut microbiota links with several disorders, including inflammatory bowel diseases[10].Changes in microbial composition have been associated with obesity and weight reduction;however,factors associated with these changes are not well defined[11].The alterations in community population,whether chronic or acute, are accompanied by alteration in the microbiota’s collective genome,or microbiome,and the patterns and specific metabolic capabilities[12].Recently,it was observed that feeding mice a high-fat diet also caused an increased Firmicutes and reduced Bacteroidetes-type microbiota,as described before[13].Therefore,the relevant variables,such as changes in the diet to changes in the microbiome,are important in understanding how environmental factors and behavior influence physiology of individual people.Besides immune and inflammatory mechanisms,other pathways may involve,which are the link between gut microbiota and metabolic syndrome.The microbiota produces enzymes that degrade ingested polysaccharides,which promotes the absorption of various nutrients (especially carbohydrates)that result in increased liver lipogenesis,hyperinsulinemia,and hepatic insulin resistance.It has been demonstrated that high intake of cereal fiber associates with reduced risk for T2D[14].Many pharmaceutical dosage forms irritate the stomach due to their chemical properties.Others undergo biochemical changes in the gastric acid through the action of various enzymes.Specific Eudragit acrylic polymers have been developed for peroral dosage forms with step-wise release of active ingredients in the digestive tract.The pharmaceutical principle of Eudragit coating is to solubilize in a specific environmental pH value[15].Eudragit L100-55,composed of methacrylic acid and methyl methacrylate(1:2, Mw=approx.135,000),was chosen as a pH-sensitive polymer owing to its unique dissolution behavior above pH5.5.Eudragit have been used as pH-sensitive polymers in various applications including enteric coating materials and drug delivery vehicles and exhibited plastic deformation and significant speed sensitivity[16].Eudragit in combinations with other polymers,such as hydroxypropyl methyl cellulose and talc, stabilized loaded drugs and provided a controlled release of them[17].Microparticles made from Eudragit polymers utilize for protein drug delivery to the lower intestine after oral administrations are based on the change of pH during the gastrointestinal passage[18].Cefdinir[8-(2-(2-amino-1,3-thiazol-4-yl)-1-hydroxy-2-nitroso-ethenyl]amino-4-ethenyl-7-oxo-2-thia-6-azabicyclo[4.2.0]oct-4-ene-5-carboxylicacid is a semisynthetic third-generation broad-spectrum oral cephalosporin active against both gram-positive and gram-negative bacteria and is widely used to treat acute chronic bronchitis, rhinosinusitis,and pharyngitis.It has only21–25%of oral bioavailability[19],which is probably due to low aqueous solubility.In trying to answer these problems,this investigation studied the effect of an excess fructose leading pathological changes and activity levels within the liver defense system as well as gut microbiota.The alteration of gut flora by using Eudragit L100-55-coated cefdinir microspheres (Cef EL)which used to disrupt the microbiota in small intestine of male Wistar rat.This would help to understand better the role of gut microbiota in fructose-induced metabolic changes as well as to develop appropriate strategies in the prevention and treatment of obesity and T2D triggered by unhealthy diets.Materials and MethodsStarting Materials and ReagentsEudragit L100-55(average molecular weight approximately320,000g/mol)was provided by Rohm GmbH and Co.KG(Germany).Cefdinir(cefdinir is an extended spectrum oral third-generation antimicrobial agent with a broad-spectrum activity against enteric gram-negative rods and has low permeability)was received as a gift from Macleods Pharmaceuticals Limited (Mumbai,India).All other reagents and solvents were of analytical grade.Preparation of MicrospheresThe preparation of microspheres was either based on an oil/water emulsification—solvent evaporation or solvent extraction method.The usually employed oil/water emulsification process is given as a standard in the preparation of Cef EL.For all different techniques,a fixed amount of polymer(100mg)and drug(10mg)were used.Accurately weighed antibiotic cefdinir was taken in different drug polymer ratio(1:2)and was added in an5-ml acetone organic phase(internal aqueous phase)having10%w/v Eudragit L100-55(EL),and sonicated in an ultrasonicator(JY92-11DN,Syclon,Japan)for10min.This solution was slowly injected (0.33ml/min)into an external aqueous phase containing paraffin light liquid(100ml)and was emulsified containing the emulsifier,span80(2%v/v).The system was stirred continuously using a mechanical stirrer at1,000rpm and37±0.5°C for5h to form a uniform emulsion and allowed complete evaporation of the solvent to form microspheres.The paraffin was decanted off;the microspheres were washed3–4times with petroleum ether(40–60°C),collected by filtration and finally dried at room temperature for3h[20].In Vitro Release Profile StudyThe transit time of a drug through the absorptive area of the gastrointestinal(GI)tract is between9and12h,whereasγscintigraphy studies confirmed that short GI transit time from mouth to cecum is of4to6h Thus,assuming a maximum GI tract transit time of12h,a formulation in the cecum is expected to release its drug load within6h.Considering the same, in vitro drug release from all the batches of microspheres was studied for duration of6h.For the first2h,the drug release profile was performed in simulated gastric fluid(pH1.2with 0.1N HCl).Remaining4h of the release profile of cefdinir from Eudragit(L100-55) microspheres was evaluated in phosphate buffer(pH5.5),which represents the pH of small intestine.Microspheres equivalent to1.5-mg drug/ml were transferred to the dissolution media (20ml)which maintained at37ºC±0.5ºC under stirring at75rpm.A0.5ml of samples was withdrawn every regular time interval up to6h,and the withdrawn volume was replenished immediately by same volume of fresh phosphate buffer.Amount of drug released in the withdrawn sample was estimated by measuring absorbance in a UV spectrophotometer at 287nm for cefdinir against a phosphate buffer(pH5.5)as control blank.Enteric Nature of MicrospheresThis test was performed to determine whether the drug would be released in the acidic environment of the stomach(i.e.,pH between1and3).Cef EL equivalent to1.5-mg drug was transferred to20mL of0.1N HCl that was maintained at37±0.5°C under stirring at 75rpm.A0.5ml of samples was withdrawn every regular time interval up to3h,and the withdrawn volume was replenished immediately by same volume of0.1N HCl.Amount of drug released in the withdrawn sample was estimated by measuring absorbance in a UV spectrophotometer at287nm against a0.1N HCl as blank.Physicochemical Characterization of Cef ELScanning Electron MicroscopyThe external and internal morphology of Cef EL was analyzed by scanning electron micros-copy(SEM).The microspheres were fixed on supports with carbon-glue and coated with goldusing a gold sputter module in a high vacuum evaporator.Samples were then observed with the SEM(LEO-1430VP,UK.)at10kV.Particle Size AnalysisThe particles was analyzed basis on the dynamic light scattering technique(DLS)by using a Mastersizer(Malvern Instrument,Malvern,UK),Particle sizes are expressed as the weighed mean of the volume distribution.Each value resulted from a triplicate determination.FT-IR AnalysisFourier transform infrared(FT-IR)spectra of Cef EL were obtained on a PerkinElmer,GX-FT-IR spectrometer.Eudragit L100-55-containing microspheres(with or without cefdinir)were prepared in KBr disks(2mg sample in200mg KBr).The scanning range was400–4,000cm−1 and the resolution was1cm−1.In Vitro Antimicrobial Profile StudyIn vitro antimicrobial studies of Cef EL were performed as described earlier[21]with modification.Cefdinir was dissolved in small amount of dimethylformamide and suitably diluted with distilled water.Similarly,Cef EL and Eudragit polymer was dissolved separately in distilled water and suitably diluted with the same to obtain the concentration of5μg/mL.One milliliter overnight grown culture of Escherichia coli MTCC443and Lactobacillus casei MTCC1423was inoculated in freshly prepared Luria-Bertani and MRS broths,respectively.Both the broth was adjusted to pH5.5 before addition of inoculum.After2h of incubation at37°C at shaking150rpm, prepared microsphere Cef EL(cefdinir concentration5μg/mL(MIC0.1–0.5μg/ml) was added to both the cultures and incubated at37°C with shaking at150rpm. Changes in optical density(600nm)were recorded every hour for7h.Activities of Cef EL were compared with control cells treated with pure Eudragit L100-55and cefdinir.Animal ExperimentsEight to10week-old healthy male Wistar rats weighing150–200g were procured from the Animal Research Facility,Torrent Research Center,Ahmedabad(India)under the approval Institutional Animal Ethics Committee,protocol no.IS/BT/PhD11-12/ 1004and were maintained at the animal house of Institute of Pharmacy(Nirma University,India).The animals were acclimatized at temperature of25±2°C and relative humidity of50–60%under12/12h light/dark conditions for1week before experiments.Animals were assigned to a normal control group(CD;n=6)that consumed a standard diet and normal drinking water,a high-fructose-fed control group(HFD;n=6)that consumed a standard diet having65%fructose with normal drinking water,and cefdinir microsphere-treated group(HFD Cef EL,n=6)that consumed a high-fructose diet for30days except for the days before oral glucose tolerance tests(OGTTs)and blood collection.The composition of experimental diets is presented in Table1.Animals had free access to food and water with or without fructose.The food and water intakes were recorded daily by correction of spillage and body weight was measured twice a week.Oral Glucose Tolerance TestOral glucose tolerance tests were performed between 8.0and 10.0h at weekly intervals.The diets were removed from animal cages for 12h before the administra-tion of an oral glucose load (2g/kg of body weight)by orogastric gavage.Blood samples were collected from the tail vein at 0,15,30,60,90,and 120min after glucose administration.Glucose concentration was determined with an Accu-Check Advantage Blood Glucose Monitor (Roche Group,Indianapolis,IN,USA).Area under the curve for glucose (AUC glucose )was determined using the trapezoidal rule.Blood and Tissue Sample CollectionAt the end of the experiments,the animals were sacrificed by a deep dose of anesthesia.The blood samples were collected,processed for plasma and serum,and then stored at −80°C.Blood parameters including blood glucose,plasma triglycer-ides,total cholesterol and high-density lipoprotein cholesterol (HDL-C),liver patho-logical marker such as SGPT and SGOT from serum,hepatic triglycerides,and cholesterol were analyzed by enzymatic kit (Accucare diagnostics,India).The liver glycogen was determined as described earlier [22].Body fats were measured while the liver,distal ileum,and proximal colon were washed with ice-cold saline and then were stored at −80°C until used.A small portion of the liver,distal ileum,and proximal colon were excised from animals of each group,fixed with 10%v /v formalin saline,and processed for standard histopathological procedures.Paraffin-embedded specimens were cut into 5μm sections (Yorco Sales Pvt.Ltd.,New Delhi)and stained with hematoxylin and eosin (H&E).The histopathological tissue sections were viewed and digitally photographed using a Cat-Cam 3.0MP Trinocular micro-scope with an attached digital 3XM picture camera (Catalyst Biotech,Mumbai,India).Quantification of Fecal BacteriaFor quantitative determination of Enterobacteriaceae,lactobacilli,and bifidobacteria,1-g cecal content mixed 0.85%of NaCl solution and homogenized by vortexing for 10min.Serial 10-fold dilutions were prepared and volumes of 100μl of three appropriate dilutions Table 1Formulation of the diets a From Central Drug House Pvt Limited,India b From Ankur Foods,India c From M.P.Biosciences,UK d From Amrut Agrofoods,Mumbai Ingredients (%)Control d High fructose High sucrose Starch6500Fructosea 0650Sucrose a0065Casein a202020Corn oil b 555Wheat bran 555Mineral mix c 3.5 3.53.5Vitamin mix c 111D -Methionine 0.30.30.3Choline chloride0.20.20.2were loaded onto the surface of plates in triplicate.EMB,MRS,and BS agars were used for growing Enterobacteriaceae,lactobacilli and bifidobacteria,respectively.All media were obtained from Himedia(Mumbai,India).All these plates were incubated anaerobically at 37°C for72h.The number of colony counting for each microbiota species was expressed as log CFU/g fecal content.Microscopic characteristics of all of the colonies were investigated by Gram staining methods[23].Statistical AnalysisAll the values are expressed as mean±SD.Statistics was applied using GraphPad Prism software version5.One-way ANOV A followed by Tukey’s multiple comparison tests was used to determine the statistical significance of the test groups with the control and within the test groups.Differences were considered to be statistically significant when p<0.05. ResultsPreparation of MicrospheresThree different formulations containing cefdinir with Eudragit L100-55of various drugs to polymer ratio were prepared by using emulsion solvent evaporation techniques.The properties of these formulations,process yield,and entrapment efficiency are listed(Table S2,supple-mentary data).From the results,it was observed that increased polymer ratio with respect to drug leads to enhance drug entrapment efficiency,so as the amount of drug remaining and available for encapsulation increased as the theoretical drug loading increased.However,the polymer with lower concentration lowers the entrapment efficiency because loss of product during the successive decantation,washing,and drying process.The entrapment efficiency of cefdinir in microsphere formulation was found to be enhanced by increasing the drug to polymer ratio from lower to higher side,and maximum entrapment efficiency was observed at drug to polymer ratio1:1(Table S2,supplementary data).In Vitro Drug Release StudyTo evaluate the pH-dependent release profiles of cefdinir microspheres,in vitro release tests were performed up to6h in simulated gastric fluid(Fig.1).In the dissolution medium at pH1.2,nearly5–8%of the drug was released in2h.It was shown that slow drug release behavior was observed for microspheres at pH 1.2.Since the polymer is insoluble in the release media with pH1.2,the microparticles were only slightly swollen and remained intact in this case.At a pH of 5.5,the polymer dissolved rapidly and the microspheres’disintegration resulted in a faster drug release rate compared with pH1.2.As the concentration of polymer increases,release of drug was found to be much slower because more amount of drug entrapped inside the polymer matrix.Whereas more drug concentration,rapid release was observed for the absorption on the surface of microsphere.Scanning Electron MicroscopyThe SEM images of Cef EL and Eudragit L100-55are presented(Fig.2).The unprocessed cefdinir particles(Fig.2a)have needle-like crystals,and drastic change in the morphology andshape of particles was observed for both processed particles (Fig.2c )and Eudragit polymer (Fig.3b ).Under SEM examination,Cef EL exhibited spherical morphology and smooth surfaces as well as a monodispersed size distribution are shown (Fig.2c ).The microsphere particle size distribution of Cef EL (cefdinir and Eudragit 100-55drug polymer ratio 1:1)was presented (Fig.S1supplementary data).FT-IR StudyThe FT-IR spectra of intact cefdinir,Eudragit L100-55,and Eudragit L100-55-coated cefdinir (Cef EL)was shown (Fig.3).IR spectrum of cefdinir (Fig.3a )is character-ized by principal absorption peaks at 2,928cm −1(O –H stretch COOH),2,849cm −1(C –H stretch cyclic),1,761cm −1(C ═O),1,678cm −1(C ═C alkene),1,620cm −1(C ═C aromatic),1,516cm −1(N –H bending),1,391cm −1(C –N stretch),and 656cm −1(C –S).The spectrum of Eudragit L100-55has a broad band characteristic of hydroxyl groups (O –H stretch vibration)in the range of 3,476–2,358cm −1,characteristicbands Fig.1Drug release profiles of CefELFig.2SEM image of a unprocessed crystalline cefdinir particles (×1,000),b Eudragit L100-55and c cefdinir coated with Eudragit L00-55polymerof methyl and methylene (C –H stretch vibration)at 2,976cm −1and 2,895cm −1,and a strong band due to carbonyl groups (C –O stretch vibration)at 1,733cm −1and two bands due to ester linkages (C –O stretch vibration)at 1,368and 1,266cm −1(Fig.3b ).FT-IR spectra of Cef EL polymer mixture shows prominent peaks at 3,427cm −1(O –H),2,922cm −1and 2,852cm −1(C –H),and 1,730cm −1(H –O –H bending).All the peaks of Cefdinir (CEF)completely disappeared with a shift of 1,781–1,767cm −1.The FT-IR spectra of Cef EL (Fig.3c )complex shows complete disappearance of the CEF peaks at 3,300,2,976,2,895,and at 652cm −1with strong decrease in peak intensity.This suggested that,CEF could form inclusion complex with Eudragit L100-55in solid state.The microspheres of Cef EL did not show any new peaks,indicating no chemical bond formation in the complexes.The significant differences in the observed vibrational transitions and the bands in the spectrum of the crystalline form were clearer and sharper than the bands of the amorphous forms.In addition,in IR-spectra,it is already known that significant differences between hydrate and anhydrate form were observed around 2,800–3,800cm −1[24].Due to the O –H stretching vibration of water molecules,the unprocessed cefdinir,which is in monohydrate form,had characteristic peaks observed at 1,164,1,118,1,620,1,7613,257,and 3,357cm −1,but not in processed particles.In Vitro Antimicrobial Profile StudyThe antimicrobial activity of Cef EL,cefdinir,and Eudragit L100-55against gram-positive (L.casei MTCC1423)and gram-negative (E.coli MTCC443)species was checked and results are summarized (Fig.4).These studies revealed that Cef EL have shown similar antimicrobial activity like CEF alone against L.casei while Eudragit L100-55alone have no properties of microbial inhibition as it shows similar growth pattern like L.casei (Fig.4a )However,Cef EL inclusion polymer complex has shown significant microbial inhibition against both the microorganisms as like pure CEF in pH 5.5.However cefdinir known for its gram-negative antimicrobial agents,the effects after Eudragit polymer coating also not reduced much as shown (Fig.4b ).The antimicrobial activity of Cef EL polymer and pure CEF alone have identical inhibition properties against E.coli in pH 5.5where as Eudragit alone had no inhibitory properties as grown with E.coli.Fig.3FT-IR spectra of cefdinirand Eudragit L100-55polymer(A )Eudragit L100-55;(B )cefdinircoated with Eudragit L100-55and(C )cefdinirIn Vivo ExperimentsBody Weight,Food,and Water IntakeAs shown (Table 2),the rats fed with a high-fructose diet (HFD)showed a significant increase in body weight (p <0.001);as compared to the normal control diet (CD)group at week 4.It was found that body weight was significantly reduced (p <0.001)when administered with cefdinir microsphere (Cef EL)as compared HFD-fed group.There are no significant changes observed in food consumption among the groups while water intake was significantly higher in HFD group (p <0.01)than CD group but no significant changes observed in Cef EL group in comparison with CD and/or HFD group.Body weights of HFD groups were recorded significantly higher (p<0.001)than CD group while Cef EL group has no significant changes than CD group animals after the 4-week coadministration period.But the Cef EL group animals had a significantly reduced (p<0.001)body weight than HFD animals.The weight of abdominal fat content was significantly higher (p<0.001)in the HFD group animals (12.8±0.96g)than CD group (6.32±0.43g)while Cef EL (5.17±0.26g)group were significantly different from CD group (p<0.05)and HFD group (p<0.001)animals.Fig.4In vitro antimicrobial assay of Cef EL against a L.casei MTCC 1423and b E.coli MTCC443.Values are presented as mean±SEM (n =3).Values with superscript letters are significantly compared with control (*p <0.05)Table 2Food intake and body weight of experimental rats fed with different experimental diets during 1-month periodParametersCD HFD HFD +Cef EL Food intake (gm/day per rat)19.93±3.216.58±2.617.84±2.3Water intake (ml)38±4.548±5.2b 42±4.2Initial body weight (g)170±12.30165±11.50165±10.6Final body weight (g)195±8.5225±9.5198±8.3Body weight gain (g)25±7.865±9.4c 33±4.6c Body fat (g) 6.32±0.4312.8±0.96c5.17±0.26a,c V alues are presented as mean±SEM drawn from pooled spillage of three cages of the same group and calculated as six animals per group.Values with different superscript letters are significantly different (a p >0.05;b p >0.01;c p >0.001)Oral Glucose Tolerance TestThe OGTTs value after the second week in the HFD group and the AUC glucose values were significantly higher(p<0.01)(46%)than that of the CD group while Cef EL group were significantly14%lower(p<0.05)than that of HFD group(Fig.5b).However,OGTTs were impaired after4weeks in the Cef EL animals,and the AUC glucose values were significantly (p<0.01)lower(34%)than those of the HFD animals.After the fourth-week experimental period,the AUC glucose was150%higher(p<0.001)in the HFD than the CD group,whereas in Cef EL animals,it was34%lower,which is significant(p<0.01)than that of the HFD animals yet significantly(p<0.001)65%higher than that of the CD animals(Fig.5d).Blood and Tissue Biochemical AnalysisEarlier studies indicated that higher triglyceride(TG)and cholesterol levels were the main predictors and causative factors for inducing insulin resistance in type2diabetes.In our current results,the fasting blood glucose levels in the HFD group were significantly higher(p<0.001)Fig.5Effect of Cef EL on oral glucose tolerance test of HFD rats.a–d represents the OGTT and AUC glucose values of animals after15and30days of intervals,respectively.Rats(6–8-week old,male,n=6/group)were administered with2g of glucose per kilogram of body weight after fasting for12h.The blood glucose levels were measured from0to120min.V alues are presented as mean±SD(n=3).Values with different letters are significantly different(p<0.05).AUC glucose area under the curve for glucose,HFD Cef EL,Cef EL and high-fructose-treated group,HFD high-fructose-fed group,CD control group。
肠道菌群定义
肠道菌群,也称为肠道微生物群或肠道微生物组,是指人体肠道内所寄居的非常丰富和多样化的微生物群落,包括细菌、真菌、病毒和其他微生物。
肠道菌群是一种共生关系,与人体相互作用,并对人体的健康发挥着重要作用。
它参与许多生理过程,包括食物消化、免疫调节、维持肠道屏障功能、合成维生素等。
它还具有抗菌和抗炎作用,有助于对抗有害菌的入侵,维护肠道健康。
肠道菌群的组成是高度个体化的,受多种因素影响,包括遗传、生活方式、饮食习惯、生活环境、使用抗生素等。
不良的饮食和生活方式,以及某些药物的使用,如滥用抗生素,可能会导致肠道菌群失衡或紊乱,进而对人体健康产生负面影响。
近年来,越来越多的研究表明,肠道菌群与许多疾病的发生和发展密切相关,包括肠道疾病(如炎症性肠病)、肥胖、2型糖尿病、自身免疫性疾病和心血管疾病等。
因此,理解和维护良好的肠道菌群健康对于维持整体健康非常重要。
值得注意的是,肠道菌群的研究仍在不断发展,我们还需要
更多的科学研究来深入了解它与健康的关系,以及如何通过调整饮食和生活方式来维护良好的肠道菌群健康。
肠道菌群应用的探究进展[摘要]人类的肠道中约含有十万亿多种共生体(50个细菌门和大约100-1000个细菌物种)。
肠道菌群(gut microbiota)是人体不可分割的一部分,在人体的免疫、代谢、结构和神经系统方面发挥着重要的作用。
因此,随着对微生物肠道菌研究的深入,会在人类的健康领域取得重大突破。
本文综述了微生物肠道菌群的应用,方便读者在这一领域的理解。
[关键词]微生物肠道菌群;肠-脑轴;肠-肺轴[Abstract] The human gut contains about ten trillion symbiotes (50 phyla and about 100-1000 bacterial species). Gut microbiota plays a crucial role on the human body and is essential to the immunity, metabolism, structure and nervous system of the human body. Therefore, with the in-depth study of gut microbiota, there will be a major breakthrough in human health. The essay reviews the application of gut microbiota to help readers have a better understanding in this field.近年来,对于肠道菌群的研究逐渐深入,越来越多的科研工作者已经达成了一种共识——微生物肠道菌群是人体最大的“免疫器官”。
然而,随着研究的进一步深入,科学家们发现了越来越多的功能,本文通过文献调研总结出以下几点关于微生物肠道菌群的应用,从而来探究肠道菌群成为药物治疗疾病新靶点的可能性。
肠道菌群-阿尔茨海默病防治的新靶标
王锋;蒋与刚
【期刊名称】《生理科学进展》
【年(卷),期】2022(53)1
【摘要】阿尔茨海默病(Alzheimer's disease,AD)是一种渐进性中枢神经系统退行性疾病,其发病机制仍未完全阐明。
近年来,随着菌群-肠-脑轴研究的兴起,肠道菌群在AD发生和发展中的作用日益得到重视。
本文综述了AD患者的肠道菌群特征,肠道菌群影响AD的主要途径及益生菌、抗生素和饮食等对AD患者的干预作用,为制定靶向肠道菌群的防治策略提供参考。
【总页数】4页(P73-76)
【关键词】阿尔茨海默病;肠道菌群;益生菌;抗生素;饮食
【作者】王锋;蒋与刚
【作者单位】军事科学院军事医学研究院环境医学与作业医学研究所
【正文语种】中文
【中图分类】R153
【相关文献】
1.阿尔茨海默病与肠道菌群的关系及菌群调节对其防治的展望
2.上海交大等发现肠道菌群为糖尿病干预新靶标
3.中医药调节肠道菌群防治阿尔茨海默病研究进展
4.绝经后骨质疏松免疫防治新靶点:肠道菌群
5.中医药防治阿尔茨海默病的新视角——靶向肠道菌群
因版权原因,仅展示原文概要,查看原文内容请购买。
肠道菌群(GutMircrobiota≈CNS!)小白讲实验,带你走进科研殿堂,助你从实验小白变身科研达人实验小白人的胃肠道表面积大约有400平方米(相当于2个网球场的大小),而体内95%的细菌都位于胃肠道中,总量大概有1-2千克,几乎占粪便干重的1/3。
尽管细菌的体积仅仅是细胞的1/10到1/50大小,但是细菌的数量是机体细胞数量的10倍,首尾相连能绕地球两圈半。
从胃到结肠,细菌的数量和代谢强度都是逐渐增加的,因此,结肠是人体中细菌浓度最高,代谢最旺盛的部位。
影响肠道菌群的因素Brain-Gut axis当大脑有应激反应时,可通过肾上腺素能神经途径改变肠道菌群稳态,引起肠道内环境紊乱[1]。
最常见的是应激性肠炎(IBS),即在你紧张的时候可引起急性腹泻的一种疾病。
实验小白将有专刊为大家介绍这种神秘的疾病(“懒驴上磨屎尿多”中的科学知识)。
1.Collins SM, Bercik P: The relationship between intestinal microbiota and the central nervous system in normal gastrointestinal function and disease. Gastroenterology 2009, 136:2003-2014.酒精 & 肠道菌群酒精和乙醛等可增加肠道的通透性,引起肠道菌群紊乱,导致菌群分泌LPS、PGN、flagellin、CpG以及DNA增加,最终改变肝内免疫反应,促进酒精性肝病等肝脏疾病的发生发展。
1. Szabo G: Gut-liver axis in alcoholic liver disease. Gastroenterology 2015, 148:30-36.宿主粪便中的miRNA & 肠道菌群宿主粪便中的miRNA可进入像F.nucleatum和E.coli等肠内细菌,调控其基因的转录,最终影响这些肠内细菌的生长。
粪便中缺少miRNA可导致肠内菌群紊乱,加重大肠炎发展;而移植正常小鼠粪便中的miRNA则可减轻大肠炎症状[1]。
1. Liu S, da Cunha AP, Rezende RM, Cialic R, Wei Z, Bry L, Comstock LE, Gandhi R, Weiner HL: The Host Shapes the Gut Microbiota via Fecal MicroRNA. Cell Host Microbe 2016, 19:32-43.宿主遗传背景 & 肠道菌群2014年cell杂志发文称,同卵双生比异卵双生具有更相似的肠道菌群组成,而且鉴定出多种和遗传有关的肠道细菌种属。
另外,该文章还发现减肥相关肠道细菌(Chtistensenella minuta).如上图所示,与肥胖的人相比,苗条的人具有更高水平的C.minuta,而无菌小鼠接受C.minuta移植后,小鼠体重减轻。
1. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, et al: Human genetics shape the gut microbiome. Cell 2014, 159:789-799.抗生素/质子泵抑制剂等 & 肠道菌群抗生素和质子泵抑制剂等试剂的大量使用可改变肠道菌群组成。
在我国,抗生素滥用问题十分严重,而兽用抗生素滥用问题更是触目惊心。
1. Modi SR, Collins JJ, Relman DA: Antibiotics and the gut microbiota. J Clin Invest 2014, 124:4212-4218.2. Jackson MA, Goodrich JK, Maxan ME, Freedberg DE, Abrams JA, Poole AC, Sutter JL, Welter D, Ley RE, Bell JT, et al: Proton pump inhibitors alter the composition of the gut microbiota. Gut 2016, 65:749-756.Tfh-IgA & 肠道菌群宿主体内的TLR配体与DC等抗原提呈细胞表面的TLR结合后诱导Tfh细胞产生,进而辅助B细胞产生IgA抗体来调控肠道菌群组成[1-3]。
1. Kubinak JL, Petersen C, Stephens WZ, Soto R, Bake E, O'Connell RM, Round JL: MyD88 signaling in T cells directs IgA-mediated control of the microbiota to promote health. Cell Host Microbe 2015, 17:153-163.2. Proietti M, Cornacchione V, Rezzonico Jost T, Romagnani A, Faliti CE, Perruzza L, Rigoni R, Radaelli E, Caprioli F, Preziuso S,et al: ATP-gated ionotropic P2X7 receptor controls follicular T helper cell numbers in Peyer's patches to promote host-microbiota mutualism. Immunity 2014, 41:789-801.3. Kawamoto S, Maruya M, Kato LM, Suda W, Atarashi K, Doi Y, Tsutsui Y, Qin H, Honda K, Okada T, et al: Foxp3(+) T cells regulate immunoglobulin a selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 2014, 41:152-165.母体IgG和IgA抗体促进子代肠道菌群稳态母体内IgG和IgA抗体协同作用抑制子代体内肠道菌群特异性的CD4+T细胞反应,从而维持子代肠道菌群稳态。
1. Koch MA, Reiner GL, Lugo KA, Kreuk LS, Stanbery AG, Ansaldo E, Seher TD, Ludington WB, Barton GM: Maternal IgG and IgA Antibodies Dampen Mucosal T Helper Cell Responses in Early Life. Cell 2016, 165:827-841.饮食 & 肠道菌群如上图所示,高纤维饮食可以很好的维持母代、子代和孙代的肠道菌群多样性,而低纤维饮食可显著减少母代、子代和孙代的肠道菌群多样性。
而且这种多样性减少的过程是不可逆的[1]。
1. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL: Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016, 529:212-215.肠道菌群的作用肠道菌群除了和肠内疾病的发生发展(如IBD、IBS、克罗恩病、结直肠癌等)有直接关系,和肿瘤、肥胖、糖尿病、关节炎、哮喘等肠外疾病也有密切关系,甚至还能影响机体的行为、学习和记忆等过程。
实验小白这一期重点为大家介绍肠道菌群与这些肠外疾病的关系。
肠道菌群 & 免疫在具体介绍肠道菌群和疾病的关系之前,我们首先总体了解一下肠道菌群是如何调控机体的免疫系统的。
肠道菌群的代谢物肠道菌群的代谢产物中的短链脂肪酸(Short-chain fatty acids)、芳香烃受体的配体(AHR liagands)、多胺类物质(polymines)等都可显著影响机体的免疫功能。
尤其是短链脂肪酸可结合多种GPR分子,诱导Treg细胞和耐受型的DC细胞产生,维持肠道稳态,最终降低Th2、过敏等炎症反应[1]。
1.Rooks MG, Garrett WS: Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016, 16:341-352.肠道菌群的组分肠道菌群很多自身组分(如LPS、鞭毛蛋白、肽聚糖、甲酰基多肽以及特殊的核酸等)都可调控机体的免疫反应。
例如(如上图所示),肠道拟杆菌的成分PSA经过DC细胞提呈后,诱导Treg细胞产生,同时抑制Th1和Th17细胞反应[1]。
1.Rooks MG, Garrett WS: Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016, 16:341-352.Gut-Liver axis肠道菌群稳态被环境或免疫等因素破坏后,可产生大量的病原体相关分子模式,这些分子随着门静脉进入肝脏进而与TLR等模式识别受体结合而活化肝内巨噬细胞、星状细胞等固有免疫细胞,最终可导致肝纤维化、肝炎、NAFLD以及肝癌的发生[1]。
1.Chassaing B, Etienne-Mesmin L, Gewirtz AT: Microbiota-liver axis in hepatic disease. Hepatology 2014, 59:328-339.Gut-Brain axis肠道菌群还可通过代谢产物、刺激免疫细胞分泌细胞因子、诱导肠内分泌细胞产生5-HT、影响神经细胞等方式调节大脑活动,最终影响人的行为、心情、学习和记忆等活动[1-2]。
1.Collins SM, Surette M, Bercik P: The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol 2012, 10:735-742.2. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY: Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161:264-276.肠道菌群 & 肿瘤(详细内容请点击这里)肠道菌群 VS. 肿瘤形成在TLR信号存在的卡波氏肉瘤模型中,肠道共生菌产物触发TLR5信号,促进肿瘤生长。