Gut-Liver Axis and Sensing Microbes
- 格式:pdf
- 大小:335.52 KB
- 文档页数:8
Microbial Ecology of the Gut The microbial ecology of the gut is a fascinating and complex aspect of human biology that has been the subject of extensive research in recent years. The gut microbiome, which consists of trillions of microorganisms, including bacteria, viruses, fungi, and other microbes, plays a crucial role in maintaining theoverall health and functioning of the human body. This intricate ecosystem of microorganisms has been found to have a significant impact on various aspects of human health, including digestion, immunity, metabolism, and even mental health. One of the most significant aspects of the gut microbiome is its role in digestion and nutrient absorption. The microorganisms in the gut play a key role in breaking down complex carbohydrates, producing essential vitamins, and aiding in the absorption of nutrients. This symbiotic relationship between the gut microbiota and the human body is crucial for maintaining a healthy digestive system and overall well-being. Imbalances in the gut microbiome, known as dysbiosis, have been linked to various digestive disorders, such as irritable bowel syndrome, inflammatory bowel disease, and even obesity. In addition to its role in digestion, the gut microbiome also plays a crucial role in modulating the immune system. The presence of beneficial microorganisms in the gut helps to train the immune system to distinguish between harmful pathogens and beneficial microbes, thus helping to prevent the development of autoimmune disorders and allergies. Furthermore, the gut microbiome has been found to produce various metabolites and signaling molecules that can directly influence immune cell function, further highlighting the intricate relationship between the gut microbiota and the immune system. Moreover, recent research has also uncovered the significant impact of the gut microbiome on mental health and brain function. The gut-brain axis, a bidirectional communication system between the gut and the central nervous system, has been found to play a crucial role in regulating mood, cognition, and behavior. The production of neurotransmitters, such as serotonin and dopamine, by the gut microbiota, as well as the modulation of the immune system and inflammation, can have a profound impact on mental health. Imbalances in the gut microbiome have been linked to various mental health disorders, including depression, anxiety, and even neurodegenerative diseases. Furthermore, the composition of the gutmicrobiome has also been found to be influenced by various factors, including diet, lifestyle, and environmental exposures. A diet rich in fiber and diverse plant-based foods has been associated with a more diverse and beneficial gut microbiome, while a diet high in processed foods and saturated fats has been linked to dysbiosis and various health problems. Additionally, lifestyle factors, such as stress, sleep, and physical activity, have also been found to influence the composition and functioning of the gut microbiome. Moreover, exposure to antibiotics, environmental toxins, and other external factors can also have a significant impact on the gut microbiome, highlighting the delicate balance ofthis complex ecosystem. In conclusion, the microbial ecology of the gut is a multifaceted and essential aspect of human biology that plays a crucial role in maintaining overall health and well-being. The intricate relationship between the gut microbiota and various physiological systems, including digestion, immunity, and mental health, highlights the significant impact of the gut microbiome on human health. Furthermore, the influence of various factors, such as diet, lifestyle, and environmental exposures, on the composition and functioning of the gut microbiome underscores the importance of maintaining a healthy and diverse gut microbiome. As research in this field continues to advance, further insights into the gut microbiome's role in human health will undoubtedly emerge, paving the way for innovative approaches to promoting gut health and preventing disease.。
关系仍需要进一步探究。
综上所述,TLR4介导的炎症反应在帕金森病发病机制中的作用被广泛关注,本研究证实,MPTP 诱导的帕金森病小鼠模型同样存在TLR4活化的现象,并可能通过“肠-脑”轴双向作用加剧炎症级联反应。
而白藜芦醇可能通过抑制LPS 诱导的TLR4/MyD88/NF-κB 信号通路的激活缓解炎症反应,修复肠道屏障,并减少α-syn 在肠道的累积,进而发挥保护多巴胺能神经元的效应。
参考文献:[1]Pajares M,I Rojo A,Manda G,et al.Inflammation in Parkinson'sdisease:mechanisms and therapeutic implications [J ].Cells,2020,9(7):1687-93.[2]Wang R,Shih LC.Parkinson's disease-current treatment [J ].CurrOpin Neurol,2023,36(4):302-8.[3]Bashir Y ,Khan AU.The interplay between the gut-brain axis and themicrobiome:a perspective on psychiatric and neurodegenerative dis-orders [J ].Front Neurosci,2022,16:1030694-9.[4]Su M,Tang T,Tang WW,et al.Astragalus improves intestinal barrierfunction and immunity by acting on intestinal microbiota to treat T2DM:a research review [J ].Front Immunol,2023,14:1243834-42.[5]Wasiak J,Gawlik-Kotelnicka O.Intestinal permeability and itssignificance in psychiatric disorders-A narrative review and future perspectives [J ].Behav Brain Res,2023,448:114459-63.[6]Su Y ,Shi CH,Wang T,et al.Dysregulation of peripheral monocytesand pro-inflammation of alpha-synuclein in Parkinson's disease [J ].J Neurol,2022,269(12):6386-94.[7]Li JP,Liu X,Wu YZ,et al.Aerobic exercise improves intestinalmucosalbarrierdysfunctionthroughTLR4/MyD88/NF-κBsignaling pathway in diabetic rats [J ].Biochem Biophys Res Commun,2022,634:75-82.[8]郭野,陈美霓,李家萱.白藜芦醇的临床应用研究进展[J ].新乡医学院学报,2023,40(3):290-4.[9]Jiang H,Ni J,Hu LS,et al.Resveratrol may reduce the degree ofperiodontitis by regulating ERK pathway in gingival-derived MSCs [J ].Int J Mol Sci,2023,24(14):11294-9.[10]Mannino D,Scuderi SA,Casili G,et al.Neuroprotective effects ofGSK-343in an in vivo model of MPTP-induced nigrostriatal degeneration [J ].J Neuroinflammation,2023,20(1):155-63.[11]Li JY ,Zhao JW,Chen LM,et al.α-Synuclein induces Th17differentiation and impairs the function and stability of Tregs by promoting RORC transcription in Parkinson's disease [J ].Brain Behav Immun,2023,108:32-44.[12]安云英.白藜芦醇改善帕金森病小鼠相关症状及其对肠道菌群的影响[D ].新乡:新乡医学院,.[13]李琪,陈碾,罗金定,等.二氢杨梅素经激活AMPK/ULK1通路改善2型糖尿病大鼠帕金森病样病变[J ].生理学报,2023,9(1):59-68.[14]Yan YQ,Zheng R,Liu Y ,et al.Parkin regulates microglial NLRP3and represses neurodegeneration in Parkinson's disease [J ].Aging Cell,2023,22(6):e13834-40.[15]Su CF,Jiang L,Zhang XW,et al.Resveratrol in rodent models ofParkinsons disease:a systematic review of experimental studies [J ].图6RES 下调脑黑质区TLR4的表达,上调TH 的表达Fig.6RES downregulates the expression of TLR4and upregulates the expression of TH in the substantia nigra of the mice.A:Immunofluorescence double-stained image of TLR4-positive neurons (red fluorescence)and TH-positive neurons (green fluorescence)in the substantia nigra of the mice.The fluorescence intensity represents the level of expression (Scale bar:100μm).B:Expression of TLR4.C:Expression of TH.*P <0.05,**P <0.01.M P T P +R E S 30M P T PC o n t r o lT L R 4e x p r e s s i o n500450400350300250200150100500ControlMPTP MPTP+RES30Control MPTP MPTP+RES30T H e x p r e s s i o n40003500300025002000150010005000ABC MERGETLR4TH*****J South Med Univ,2024,44(2):270-279··278Front Pharmacol,2021,12:644219-25.[16]Shahcheraghi SH,Salemi F,Small S,et al.Resveratrol regulates inflammation and improves oxidative stress via Nrf2signaling pathway:therapeutic and biotechnological prospects[J].Phytother Res,2023,37(4):1590-605.[17]Gao YN,Meng QW,Qin JW,et al.Resveratrol alleviates oxidative stress induced by oxidized soybean oil and improves gut function via changing gut microbiota in weaned piglets[J].J Anim Sci Biotechnol,2023,14(1):54-63.[18]Shaito A,Al-Mansoob M,Ahmad SMS,et al.Resveratrol-mediated regulation of mitochondria biogenesis-associated pathways in neurodegenerative diseases:molecular insights and potential therapeutic applications[J].Curr Neuropharmacol,2023,21(5): 1184-201.[19]Zhang L,Kang QZ,Kang MX,et al.Regulation of main ncRNAs by polyphenols:a novel anticancer therapeutic approach[J].Phytomedicine,2023,120:155072-83.[20]Tao J,An YY,Xu LY,et al.The protective role of microbiota in the prevention of MPTP/P-induced Parkinson's disease by resveratrol [J].Food Funct,2023,14(10):4647-61.[21]Yang LC,Wang YB,Li ZW,et al.Brain targeted peptide-functionalized chitosan nanoparticles for resveratrol delivery: impact on insulin resistance and gut microbiota in obesity-related Alzheimer's disease[J].Carbohydr Polym,2023,310:120714-21.[22]Cannon T,Gruenheid S.Microbes and Parkinson's disease:from associations to mechanisms[J].Trends Microbiol,2022,30(8):749-60.[23]Goya ME,Xue F,Sampedro-Torres-Quevedo C,et al.Probiotic Bacillus subtilis protects againstα-synuclein aggregation in C.elegans[J].Cell Rep,2020,30(2):367-80.e7.[24]Ramezani M,Wagenknecht-Wiesner A,Wang T,et al.Alpha synuclein modulates mitochondrial Ca2+uptake from ER during cell stimulation and under stress conditions[J].bioRxiv,2023: 2023.04.23.537965.[25]Duan YN,Wang YX,Liu YH,et al.Circular RNAs in Parkinson's disease:reliable biological markers and targets for rehabilitation[J].Mol Neurobiol,2023,60(6):3261-76.[26]Aho VTE,Houser MC,Pereira PAB,et al.Relationships of gut microbiota,short-chain fatty acids,inflammation,and the gut barrier in Parkinson's disease[J].Mol Neurodegener,2021,16(1):6-12.[27]Kumari S,Taliyan R,Dubey prehensive review on potential signaling pathways involving the transfer ofα-synuclein from the gut to the brain that leads to Parkinson's disease[J].ACS Chem Neurosci,2023,14(4):590-602.[28]Roe K.An alternative explanation for Alzheimer's disease and Par-kinson's disease initiation from specific antibiotics,gut microbiotadysbiosis and neurotoxins[J].Neurochem Res,2022,47(3):517-30.[29]Kleine Bardenhorst S,Cereda E,Severgnini M,et al.Gut microbiota dysbiosis in Parkinson disease:a systematic review and pooledanalysis[J].Eur J Neurol,2023,30(11):3581-94.[30]Zhao Z,Ning JW,Bao XQ,et al.Fecal microbiota transplantation protects rotenone-induced Parkinson's disease mice via suppressinginflammation mediated by the lipopolysaccharide-TLR4signalingpathway through the microbiota-gut-brain axis[J].Microbiome,2021,9(1):226-35.[31]Wang WX,Bale S,Yalavarthi B,et al.Deficiency of inhibitory TLR4 homolog RP105exacerbates fibrosis[J].JCI Insight,2022,7(21):e160684-90.[32]De Ciucis CG,Fruscione F,De Paolis L,et al.Toll-like receptors and cytokine modulation by goat milk extracellular vesicles in a modelof intestinal inflammation[J].Int J Mol Sci,2023,24(13):11096-105.[33]Wang QH,Botchway BOA,Zhang Y,et al.Ellagic acid activates the Keap1-Nrf2-ARE signaling pathway in improving Parkinson'sdisease:a review[J].Biomed Pharmacother,2022,156:113848-55.[34]da Costa RO,Gadelha-Filho CVJ,de Aquino PEA,et al.Vitamin D (VD3)intensifies the effects of exercise and prevents alterations ofbehavior,brain oxidative stress,and neuroinflammation,inhemiparkinsonian rats[J].Neurochem Res,2023,48(1):142-60.[35]Sarkar S.Microglial ion channels:key players in non-cell autono-mous neurodegeneration[J].Neurobiol Dis,2022,174:105861-8.[36]Chen MT,Hou PF,Zhou M,et al.Resveratrol attenuates high-fat diet-induced non-alcoholic steatohepatitis by maintaining gut barrierintegrity and inhibiting gut inflammation through regulation of theendocannabinoid system[J].Clin Nutr,2020,39(4):1264-75.[37]Lei JR,Tu XK,Wang Y,et al.Resveratrol downregulates the TLR4 signaling pathway to reduce brain damage in a rat model of focalcerebral ischemia[J].Exp Ther Med,2019,17(4):3215-21.[38]Wang P,Chen Q,Tang ZQ,et al.Uncovering ferroptosis in Parkinson's disease via bioinformatics and machine learning,andreversed deducing potential therapeutic natural products[J].FrontGenet,2023,14:1231707-18.[39]黄庆洋,纪东东,田绣云,等.小檗碱通过激活Nrf2-HO-1/GPX4通路抑制小鼠海马神经元HT22细胞的铁死亡[J].南方医科大学学报,2022,42(6):937-43.[40]Mirzaei H,Sedighi S,Kouchaki E,et al.Probiotics and the treatment of Parkinson's disease:an update[J].Cell Mol Neurobiol,2022,42(8):2449-57.(编辑:林萍) J South Med Univ,2024,44(2):270-279··279肺癌是全球最常诊断的癌症之一,也是癌症相关死亡的主要原因,其中非小细胞肺癌(NSCLC )是肺癌最常见的一种亚型,占比达80%[1,2]。
The Gut Microbiota and the Development of the BrainThe human gut host 1014 bacterial organisms, an amount that exceeds eventhe number of somatic cells within the body. When combined with thebacteria living both inside and on the human body (a community collectivelyknown as microbiota, dwelling in the human microbiome), it is estimated that these (mostly) friendlyintruders outnumber the somatic and germ cells of their human home by a factor of 10 (Neufeld, Turnbaugh). Specific to the human gut is the commensal microflora, the microbial community thatenters into an important symbiotic association with the human host beginning with the colonization of the gastrointestinal (GI) tract by the bacteria within the first few postnatal days. The systematic takeover of the GI tract by this diverse array of microorganisms is actually a fairlyuseful and peaceful invasion, not the hostile takeover its numbers suggest it would be(Neufeld). Called “developmental processing,” the phenomenon begins at birth, continues throughearly development, and remains for life. Developmental processing is actually defined as a process by which environmental factors act during vulnerable or sensitive developmental periods and thus exert influences that impact the structure and function of organs that last throughout life (Heijtz). However, although the colonization of microbiota is due to postnatal environmental factors and is also affected by genetics, the relative abundance and distribution along the GI tract of the human microbiome is similar across healthy individuals, even those residing in different countries with different diets (Duff). The number and type of helpful bacteria is relatively consistent and is difficult to change. This stability is important because of the influences the microflora has on the development of a range of systems in the body. For example, the microbiota is essential to the proper development of the mucosal and systemic immune systems and in nutrient uptake and metabolism (Neufeld). In order to more fully understand the significance of these microorganisms, the Human Microbiome Project (HMP) has been launched worldwide. The HMP is an almost natural offshoot of the Human Genome Project and is hoped to not only bridge the gap between medical and environmental microbiology, but to reveal new insights into the world of health and diseases. Already, much has been found regarding the useful and important contributions these microbial symbionts make to an individual’s physiology (Turnbaugh, et al). Rec ent research regarding the HMP has revealed evidence that suggests these gut bacteria also influence the function of the central nervous system (CNS) and behavior (Neufeld). Of particular interest, though, is the impact on the functional development of the mammalian brain. Studies have shown the brain to be susceptible to internal and external cues during its perinatal life, an important point when considering the association between common neurodevelopmental disorders (e.g. autism, schizophrenia) and microbial pathogen infections during this same period. Related studies have shown support for this, at least in regards to mice; exposure to microbial pathogens within the perinatal period resulted in behavioral abnormalities such as anxiety-like behavior and impaired cognitive functions (Heijtz).Gut FeelingsThe relationship between the gut microflora and the brain, the so-called gut-brain axis, has been an important component for many years in gastrointestinal research. It is especially influential in such functional bowel disorders as Irritable Bowel Syndrome (IBS) and may play a part in ulcers. However, as with IBS, most of the studies have focused on a brain-to-gut control, a type of top-down relationship that examines mainly the impact of the brain on the function of the gut. New research in this field has also revealed that there may be close ties between the microbiota of the gut and the development and function of the central nervous system (CNS). This, in turn, has led to a discovery of a link between early life stress, altered stress activity later in life, and a change in the landscape of the microflora, all examined using the top-down approach. Further studies demonstrated the influence of microbiota over energy balance and their integral connections to the pathophysiology of obesity. Energy balance and the intake of food are both centrally mediated processes, and although the direct link between the gut microbiome and the central feeding circuits has not been discovered yet, it reveals the presence of the alternative bottom-up control, where the microbiota have an influence over the development and functionality of the brain. This discovery of bottom-up controlswill be important in the research regarding the relationship between brain development and the establishment of the gut microbiota (Neufeld). It also brings into light the possible role of the human microbiome in the incidence of neurodevelopmental disorders (Heijtz)./image.axd?id=b2487c81-5d3e-4bdc-a4bb-8b7102af2b5e&t=634164293398800000Evidence in MiceIn 2006, immunologist Sven Pettersson of the Karolinska Institute in Stockholm developed an interest in the bottom-up approach to the gut-brain axis when he and genomicistShugui Wang of the Genome Institute of Singapore discovered that the key brain chemical was regulated by the microbes in the gut (Pennisi). This finding led him to collaboration with a neurobiologist at the Karolinska Institute, Rochellys Diaz Heijtz , in order to further study the effects of gut microbiota on brain development and function using mice (Heigtz).For the study, they engineered germ-free (GF) mice that had no commensal intestinal microflora and therefore an underdeveloped immune system and specific pathogen free (SPF) mice with normal gut microbiota and put them through a series of tests in order to observe their exploratory activity and anxiety. Such tests included placing the mice in an open field box and measuring their spontaneous motor activity for an hour. Another test that assessed whether nonpathogenic gut microbiota could affect anxiety-like behavior involved both a light-dark box test and an elevated plus maze. In every case, the GF mice what are considered riskier behaviors, spending more time in the light box and in the open arms or end of the open arms of the elevated plus maze than the SPF mice. They also generated more spontaneous activity, traveling a greater total distance with more exploration of the open field than the SPF mice in the open field box. However, when the same battery of tests were performed with adult conventionalized offspring (CON) (offspring of GF mice that were exposed to the microbial pathogens early in their development), the CON mice displayed similar tendencies to the SPF mice instead of their GF parents. In order to check that this normalization of behavior patterns takes place only during a sensitive or critical period wherein the effects of the normal gut microbiota can occur, Heijtz et. al. then conventionalized adult GF mice and retested them. The new findings of the conventionalized adult GF mice showed no significant changes, displaying a failure to normalize their behavior at this late stage (Heijtz). Dr. Heijtz inferred from this data “that there is a critical period early in l ife when gut microorganisms affect the brain and change behavior in later life” (Pennisi).nerve-cell (Pennisi, Heijtz).This study clearly supports the hypothesis that the gut microbiota can have a significant impacton normal brain development and behavioral functions during a critical period early in the postnatal growth that lasts throughout life. It suggests also that during the course of evolution, microbiota colonization became an integral part of the development and programming of the brain with its effects extending to motor control and anxiety-like behaviors (Heijtz). However, Pettersson cautions that “it is importa nt to note that this knowledge can be applied only to mice, and that it is too early to say anything about the effect of gut bacteria on the human brain” (“Bacteria in the gut…”).Evidence in AutismEven with such cautions, it is hard to ignore the implications to the world of neurodevelopmental disorders. In fact, many GI disorders have shown a high comorbidity with psychiatric illnesses (Neufeld). The most widely hoped for link is between the gut microbiota and schizophrenia or autism. Children with autistic spectrum disorders, particularly, demonstrate a high incidence of severe gastrointestinal problems, combined with an onset in early childhood, perhaps linking to the critical period discovered by Heijtz et al (Parracho). In addition to several bodily and behavioral changes and difficulties, there often is an inflammation of the intestinal lining, causing the gut to be more permeable. Some of these symptoms have even been linked to changes in the gut microbiota due to treatment with antibiotics (Duff).Several studies have been done hoping to find an association between the microbiota and autism in order to begin finding a cause and then a cure. One such study tested 86 children with autism and revealed a substantial change in the metabolism of amino acids and in the normal microorganisms that inhabit the gut of a healthy person. These major differences were mostly concerned with the aerobic bacteria Escherichia coli (E. coli) and Enterococcus, and with the anaerobic bacteria Bacteroides and Bifidobacteria. E. coli is the most common aerobe of the microflora, accounting for around 80-90% of all aerobic bacteria. The second, then,is Enterococcus which accounts for just 5% of aerobes. When interpreting the results of the Bioscreenfaecal microbiology, it was found that the average amount of E. coli had considerably decreased, accounting for only 56% instead of its usual 90%. In 18 of the children, the amount of E. coli was actually less than 10%. Conversely, the amount of Enterococcus had risen to 40%, with a complete swap of the two aerobes in 19% of the children, as Enterococcus accounted for 95% of the aerobic bacteria. Relatedly, the anaerobes changed as well. There was a measurable decreasein Bacteroides whereas the Bifidobacteria increased (Duff).This study, however, is very basic and does not take into account many variables that would have to be considered in further examination of the role of the gut microorganisms in autism. For example, repeated microbial use (such as those commonly taken for autism) could cause disruptions in the indigenous microflora of the gut, accounting for some of the discrepencies. As Pettersson stated, at this point in the game, it is far too early to make any assumptions about the significance of the microbiota in the development of the human brain. In the future, who knows what will be discovered? Perhaps we will just have to listento our guts.References1. Duff, Jacques. "Autism and ADHD: Intestinal Dysbiosis." ADHD,ADD, AttentionDeficit Hyperactivity Disorder. BehaviouralNeurotherapy Clinic. Web.<.au/Intestinal_Dysbiosis.htm>.2. Heijtz, Rochellys Diaz, Shugui Wang, FarhanaAnuar, Yu Qian, Britta Björkholm,Annika Samuelsson, Martin L. Hibberd, Hans Forssberg, and SvenPettersson."Normal gut microbiota modulates brain development andbehavior." PNAS 108.7 (2011): 3047-052. PNAS: Proceedings of the NationalAcademy of Sciences of the United States of America. National Academy of Sciences,31 Jan. 2011. Web.</content/early/2011/01/26/1010529108.full.pdf+html>.3. KarolinskaInstitutet. "Bacteria in the gut may influence braindevelopment." ScienceDaily, 1 Feb. 2011. Web.4. Neufeld, BSc, Karen-Anne, and Jane A. Foster, PhD. "Effects of gut microbiota onthe brain: implications for psychiatry." Journal of Psychiatry & Neuroscience 34.3 (2009): 230-31. PubMed Central. Canadian Medical Association, May 2009. Web.</pmc/articles/PMC2674977/>.5. Parracho, H. M. "Differences between the gut microflora of children with autisticspectrum disorders and that of healthy children." Journal of MedicalMicrobiology 54.10 (2005): 987-91. . National Center for Biotechnology Information, U.S. National Library of Medicine, Aug. 2005. Web.</pubmed/16157555>.6. Pennisi, Elizabeth. "Do Gut Bugs Practice Mind Control?" Science/AAAS | News - Upto the Minute News and Features from Science. American Association for theAdvancement of Science, 31 Jan. 2011. Web.</sciencenow/2011/01/do-gut-bugs-practice-mind-contro.html>.7. Turnbaugh, Peter J., Ruth E. Ley, Micah Hamady, Claire M. Fraser-Liggett, RobKnight, and Jeffrey I. Gordon. "The Human Microbiome Project." Nature 449.7164 (2007): 804-10. Nature. Nature Publishing Group, 17 Oct. 2007. Web.</nature/journal/v449/n7164/abs/nature06244.html>.。
肠道微生物群-肠-脑轴在神经精神系统疾病中的研究进展崔佳瞿1, 2 陈启仪1, 2 李宁1, 2[1. 同济大学附属上海市第十人民医院肠道微生态诊疗中心(结直肠病专科);2. 同济大学消化系统疾病临床研究中心 上海 200072]摘要 肠-脑轴在维持机体内平衡方面起着重要作用,而肠道微生物群在肠道和大脑的双向沟通中扮演着重要角色,故学者们建立了肠道微生物群-肠-脑轴这一概念。
肠道微生物群可通过神经、免疫、神经内分泌和代谢途径对宿主产生影响,包括神经发育、传递和行为,并参与多种神经精神系统疾病的发生发展。
本文根据目前国内外的研究进展,结合本中心的临床经验,对肠道微生物群、肠道、神经系统之间的相互作用关系,肠道微生物群-肠-脑轴在神经精神系统疾病发生发展中扮演的角色,以及以肠道微生物群为神经精神系统疾病治疗靶点的肠-脑轴干预策略进行讨论,以期为神经精神系统疾病治疗提供一些新的理念和方法。
关键词 肠道微生物群 肠-脑轴 神经精神系统疾病中图分类号:R363.21; R454.9 文献标志码:A 文章编号:1006-1533(2023)01-0014-05引用本文崔佳瞿, 陈启仪, 李宁. 肠道微生物群-肠-脑轴在神经精神系统疾病中的研究进展[J]. 上海医药, 2023, 44(1): 14-18.Research progress of gut microbiota-gut-brain axis in neuropsychiatric disordersCUI Jiaqu1, 2, CHEN Qiyi1, 2, LI Ning1, 2[1. Intestinal Microenvironment Treatment Center (Department of Colorectal Disease ), Shanghai Tenth People’s Hospital;2. Clinical Research Center for Digestive Diseases, Tongji University, Shanghai 200072, China]ABSTRACT The gut-brain axis and gut microbiota play an important role in maintaining homeostasis and the bidirectional communication between the gut and the brain, respectively. Therefore, scholars established the concept of a gut microbiota-gut-brain axis. Gut microbiota may affect the host through neural, immune, neuroendocrine, and metabolic pathways, including nerve development, transmission and behavior, and participates in the occurrence and development of a variety of neuropsychiatric diseases. In this review, we discussed the interaction among gut microbiota, gut and brain, the role of gut microbiota-gut-brain axis in the occurrence and development of neuropsychiatric diseases including depression, anxiety, autism spectrum disorders, Alzheimer’s disease, Parkinson’s disease and irritable bowel syndrome, and the intervention strategy of gut-brain axis with gut microbiota as therapeutic target for neuropsychiatric diseases based on the current research progress at home and abroad and the clinical experience of the center, so as to provide some new ideas and methods for the treatment of neuropsychiatric diseases.KEY WORDS gut microbiota; gut-brain axis; neuropsychiatric disorders肠道中有数以万亿计的微生物,它们与人类的健康息息相关。
微生物组学英语Microbiome: The Unseen World Within UsThe human body is a complex and intricate ecosystem, teeming with trillions of microorganisms that play a vital role in our overall health and well-being. This vast and diverse community of microbes, collectively known as the microbiome, has been the subject of extensive research in recent years, as scientists strive to unravel the mysteries of this unseen world within us.The microbiome is a term that encompasses the entirety of the microbial communities that reside in various parts of the human body, including the gut, skin, oral cavity, and even the respiratory system. These microorganisms, which include bacteria, viruses, fungi, and archaea, have evolved alongside humans over millions of years, forming a symbiotic relationship that is essential for our survival.One of the most well-studied aspects of the microbiome is its role in the gut. The human gut is home to a vast and diverse array of microbes, with an estimated 100 trillion bacteria residing in thedigestive tract. These gut microbes play a crucial role in digesting and metabolizing the food we consume, extracting essential nutrients and energy that our bodies can then utilize.Beyond their role in digestion, gut microbes also have a profound impact on our immune system. They help to train and regulate the immune cells, ensuring that they are able to effectively fight off harmful pathogens while also maintaining a delicate balance that prevents autoimmune disorders. This intricate relationship between the gut microbiome and the immune system has been the focus of numerous studies, with researchers exploring the potential of probiotics and other microbial-based therapies to treat a wide range of health conditions.The skin microbiome is another area of intense research. The skin is the largest organ in the human body and is home to a diverse array of microbes, including bacteria, fungi, and viruses. These skin-dwelling microbes play a crucial role in maintaining the skin's barrier function, protecting us from harmful environmental factors and pathogens. They also contribute to the skin's overall health, helping to regulate inflammation, prevent the overgrowth of harmful microbes, and even influence the appearance of the skin.The oral microbiome is another important aspect of the human microbiome. The mouth is a complex ecosystem, with a diverse arrayof microbes that play a critical role in maintaining oral health. These microbes help to break down food, regulate pH levels, and prevent the overgrowth of harmful bacteria that can lead to dental problems such as cavities and gum disease.In addition to these well-known aspects of the microbiome, there is growing evidence that the microbial communities in other parts of the body, such as the respiratory system and the urogenital tract, also play important roles in human health and disease.One of the most exciting areas of microbiome research is the potential for microbiome-based therapies to treat a wide range of health conditions. By understanding the composition and function of the microbiome, researchers are exploring ways to manipulate it to improve human health. This includes the use of probiotics, which are live microorganisms that can be consumed to help restore the balance of the microbiome, as well as the development of personalized therapies that target specific microbial imbalances.Another promising area of research is the role of the microbiome in mental health. Emerging evidence suggests that the gut microbiome may play a significant role in the development and maintenance of mental health disorders, such as depression and anxiety. This has led to the concept of the "gut-brain axis," which posits that the bidirectional communication between the gut and the brain can havea profound impact on our emotional and cognitive well-being.As our understanding of the microbiome continues to grow, it is clear that this unseen world within us is a critical component of human health and well-being. By unraveling the complexities of the microbiome, researchers and clinicians are paving the way for new and innovative approaches to disease prevention and treatment. From improving gut health to enhancing mental well-being, the potential of the microbiome is limitless, and the future of personalized, microbiome-based medicine is rapidly taking shape.。
gut microbes 引文格式Gut Microbes: The Key Players in Health and DiseaseIntroductionGut microbes, also known as gut microbiota or gut microbiome, refer to the trillions of microorganisms that reside in our gastrointestinal tract. These microscopic organisms play a crucial role in maintaining our overall health and have been the focus of extensive research in recent years. This article aims to explore the importance of gut microbes in various aspects of human health and disease.The Complex World of Gut MicrobesThe gut microbiota is a highly diverse ecosystem consisting of bacteria, viruses, fungi, and other microorganisms. These microbes have co-evolved with humans, establishing a symbiotic relationship that influences our physiology, immunity, and metabolism. The composition of gut microbes varies greatly among individuals, influenced by factors such as genetics, diet, age, and environment.Regulating Digestion and Nutrient AbsorptionOne of the primary functions of gut microbes is to aid in the digestion and absorption of nutrients from our food. They break down complex carbohydrates, proteins, and fats that our own digestive enzymes cannot handle. This process produces short-chain fatty acids, vitamins, and other metabolites that contribute to our overall health. Additionally, gut microbes help regulate the integrity of the intestinal barrier, preventing harmful substances from entering the bloodstream.Immune System ModulationThe gut microbiota has a profound impact on our immune system. It helps educate and train our immune cells, distinguishing between harmless substances and potential threats. Imbalance or dysbiosis in the gut microbial community can lead to immune dysfunction and an increased risk of inflammatory diseases, such as allergies,autoimmune disorders, and inflammatory bowel disease (IBD). Research has shown that certain gut microbes can influence the production of immune cells and the development of immune tolerance.Metabolic InfluenceRecent studies have demonstrated the crucial role of gut microbes in metabolic disorders, including obesity, diabetes, and metabolic syndrome. Gut microbes are involved in the regulation of energy extraction from food and fat storage. Imbalances in the gut microbial composition have been associated with increased caloric extraction from the diet, leading to weight gain and metabolic disturbances. Certain gut microbes also produce metabolites that affect insulin sensitivity, inflammation, and lipid metabolism.Neurological ImplicationsThe gut microbiota has a bidirectional communication system with the brain, known as the gut-brain axis. This communication occurs through various pathways, including the immune system, neural connections, and the production of neurotransmitters. Emerging research suggests that the gut microbiota plays a role in anxiety, depression, and neurodevelopmental disorders, such as autism spectrum disorder. Specific gut microbial species are capable of producing neurotransmitters and other molecules that influence brain function and behavior.Clinical ApplicationsUnderstanding the importance of gut microbes has paved the way for various clinical applications. Probiotics, live microorganisms that confer health benefits when consumed, have become increasingly popular. They can help restore gut microbial balance, alleviate gastrointestinal symptoms, and prevent antibiotic-associated diarrhea. Additionally, fecal microbiota transplantation (FMT) has shown promising results in the treatment of recurrent Clostridium difficile infections and other conditions characterized by dysbiosis.ConclusionGut microbes are essential for maintaining human health and well-being. They contribute to digestion, nutrient absorption, immune regulation, metabolism, and even neurological function. The complex interplay between gut microbes and human physiology underscores the importance of maintaining a healthy gut microbial community. Further research in this area will undoubtedly yield new insights into the intricate relationship between gut microbes and human health, opening doors to innovative therapeutic approaches and personalized medicine.。
人血管内皮生长因子受体2胞外段在毕赤氏巴斯德酵母中的表达(英文)左秋;田聆;侯健梅;王永生;文艳君;李炯;魏于全【期刊名称】《四川大学学报:医学版》【年(卷),期】2006(37)1【摘要】目的探讨在毕赤氏巴斯德酵母(PICHIA PASTORIS)中高效表达有真核蛋白结构的人血管内皮生长因子受体2胞外段(HEVEGFR-2)的可行性。
方法从重组质粒PORF-HEVEGFR-2经PCR获全长HEVEGFR-2 DNA,构建重组毕赤氏巴斯德酵母分泌性表达载体,电转化PICHIA PASTORIS X-33。
用抗药性表型和甲醇诱导筛选出重组HEVEGFR-2蛋白表达阳性的转化子(X-33.HEVEGFR-2)。
结果SDS-PAGE显示。
获分子量约108 KDA的重组HEVEGFR-2蛋白。
约占X-33-HEVEGFR-2分泌性表达蛋白总量的45%。
该重组蛋白在表达上清中的质量浓度达80 MG/L。
其HEVEGFR-2部分分子量约106 KDA。
WESTERNBLOT证实,该蛋白能特异地与大鼠抗小鼠VEGFR-2单克隆抗体结合。
结论毕赤氏巴斯德酵母能高效表达有真核蛋白结构的人血管内皮生长因子受体2胞外段蛋白全段。
【总页数】4页(P1-4)【关键词】人血管内皮生长因子受体2;毕赤氏巴斯德酵母;基因表达【作者】左秋;田聆;侯健梅;王永生;文艳君;李炯;魏于全【作者单位】四川大学人类疾病生物治疗国家重点实验室肿瘤生物治疗研究室【正文语种】中文【中图分类】R818.74;R392【相关文献】1.人血管内皮细胞生长因子受体Flt-1胞外区cDNA在毕赤酵母中的表达和鉴定[J], 马骊;张智清;周小明;曾革非;陈爱君;姚立红;王小宁2.人血管内皮细胞生长抑制因子在巴斯德毕赤酵母中的分泌表达 [J], 刘麟;陈宇光;谈立松;唐亮;张颉3.克氏原螯虾i-型溶菌酶在巴斯德毕赤酵母中的高效胞外表达及其抑菌活性 [J], 水燕; 管政兵; 叶俊贤; 史永红; 刘国锋; 徐增洪4.异种同源表皮生长因子受体EGFR胞外段在毕赤巴斯德酵母中的表达 [J], 方芳;李炯;文艳君;田聆;魏于全5.鹌鹑血管内皮生长因子受体Quek1胞外段第2~4区cDNA在毕赤酵母中的表达和鉴定 [J], 刁鹏;文艳君;王永生;杜小波;周行;魏于全因版权原因,仅展示原文概要,查看原文内容请购买。
用去掉免疫球蛋白IgG的腹水提高体外培养的单克隆抗体产
量(英文)
周若愚;叶静;谢弘
【期刊名称】《中国生物工程杂志》
【年(卷),期】2005(25)8
【摘要】发展了一种在实验室大幅度提高抗体产量的方法,即采用添加2%的去免疫球蛋白IgG的腹水于无血清培养基中的方法,培养杂交瘤细胞,获得了高达3.55 ×106/ml的细胞密度,纯化后获得了135μg/ml的单克隆抗体产量,比通常的用无血清培养的单位体积培养液抗体产量高4倍。
对于体外生产单克隆抗体而言,这种方法经济并且易于推广,它的成功是抗体产量提高的一个巨大进步。
【总页数】6页(P87-92)
【关键词】腹水;细胞培养;单克隆抗体生产;无血清培养基;单克隆抗体;免疫球蛋白;产量高;IgG;体外培养;水提
【作者】周若愚;叶静;谢弘
【作者单位】中国科学院上海生命科学研究院生化细胞研究所生物治疗实验室;上海第二医科大学附属瑞金医院急诊部
【正文语种】中文
【中图分类】S852.43;S852.4
【相关文献】
1.抗人免疫球蛋白IgG单克隆抗体制备及其特性鉴定 [J], 王香菊;徐秀兰
2.小鼠腹水IgG类单克隆抗体的制备及活性研究 [J], 李涛;刘代成
3.小鼠腹水IgG类单克隆抗体的制备及活性研究 [J], 李涛;刘代成
4.抗鸡免疫球蛋白(IgM、IgG)单克隆抗体的制备与鉴定 [J], 刘晓明;刘(王子);李润萍;马从林;冯书章
5.大鼠单克隆抗体——Ⅶ.用降植烷和福氏佐剂预处理大鼠后腹水产生及单克隆抗体产量的增加 [J], J.P.Kints;周雪慧
因版权原因,仅展示原文概要,查看原文内容请购买。
·137·白首乌为萝藦科(Asclepiadaceae)植物白首乌Gynanchum…bungei…Decne.的干燥块根,味甘、苦,性微温,历代医家视为养身防老的珍品,有养血益肝、固肾益精、强筋健骨、乌黑须发及延年益寿的功效,为贵州苗族常用药材[1]。
为控制不同炮制方法对白首乌的质量,本文对其指标成分2,4-二羟基苯乙酮含量进行测定,初步探讨了其炮制意义。
1仪器与试药1.1…仪器美国ThermoscientificU-3000型高效液相色谱仪(二元梯度泵,紫外检测器色谱工作站);BS-210S型电子天平(北京赛多利斯仪器有限公司);分析天平(AG285,MetlerToledo)。
1.2…试药2,4-二羟基苯乙酮对照品(上海源叶生物科技有限公司,HPLC≥99%),乙腈、甲醇为色谱纯(天津市科密欧化学试剂有限公司);无水乙醇(重庆川东化工有限公司);水为超纯水。
2方法与结果2.1…炮制品的制备2.1.1…取白首乌饮片,加米泔水加以浸泡24h,取出,在50℃以下烘干,即得。
2.1.2…将锅烧热,加入规定量大米,中火炒至冒烟时,投入白首乌饮片,拌炒至米呈焦黄或焦褐色,饮片表面呈黄白至浅黄褐色,及时取出,过筛,放凉。
每100kg白首乌饮片用大米30kg。
2.1.3…每100kg白首乌块,用黑豆10kg,黄酒10kg。
取黑豆加适量水煮3小时,熬汁15kg,豆渣再加水煮2小时,熬汁10kg,共得黑豆汁25kg。
取白首乌饮片,加入黑豆汁、黄酒搅拌均匀,密闭闷2小时,至汁液基本吸尽,倒入高压灌内,压力下加热润1小时,停止加热,候冷取出,摊凉,于50℃烘干,即得。
2.1.4…综合2.1.3项和2.1.2项方法。
2.2…含量测定2.2.1…色谱条件色谱柱:Thermo…Hypersil…GOLD…C18色谱柱(4.6…mm…×…250…mm,5…um);流动相:甲醇(A)-…0.…1%磷酸水溶液(B),梯度洗脱(…0~24…min,15%A→21%…A;…24~35…min,21%…A;…35~36…min,21%…A→23%…A;…36~60…min,23%…A)…,流速1.0…mL·min -1,检测波长…280…nm,柱温35℃,进样量10μL。
中国免疫学杂志2023 年第 39 卷肠道微生物-免疫轴与疾病王艳婷 金延春 任科雨 崔铭娟 赵坤 曹彬① (青岛大学附属医院消化内科,青岛 266000)中图分类号 R392 文献标志码 A 文章编号 1000-484X (2023)05-1112-04[摘要] 肠道微生物对人体的健康至关重要,其改变与许多全身性疾病的发生、发展密切相关。
随着生活方式及饮食习惯的不断转变,肠道微生物正在发生改变,人类的疾病谱也发生重大改变,越来越多的研究致力于阐明肠道微生物对人类健康与疾病的影响,其中,肠道微生物-免疫轴的提出对疾病的发生发展有了更深刻的认识。
本综述总结了肠道微生物如何通过调节机体免疫系统影响某些全身性疾病发生发展,通过认识肠道微生物与机体免疫系统的相互作用,为某些疾病的诊治提供新思路。
[关键词] 肠道微生态;自身免疫性疾病;中枢神经系统疾病;心血管疾病;肿瘤Research progress of intestinal microbial -immune axis and diseaseWANG Yanting , JIN Yanchun , REN Keyu , CUI Mingjuan , ZHAO Kun , CAO Bin. Gastroenterology Department , the Affiliated Hospital of Qingdao University , Qingdao 266000, China[Abstract ] Intestinal microbe is very important to human health , and the change of intestinal microbe is closely related to theoccurrence and development of many systemic diseases. As the lifestyle and eating habits changing , intestinal microbes are changing , the human disease is also a major change , a growing number of studies to clarify the intestinal microbial effects on human health and disease , among them , the intestinal microbes -immune axis is put forward on the happening of the disease development we have a more profound understanding. This review summarizes how intestinal microorganisms affect the occurrence and development of some systemic diseases by regulating the immune system of the body. By understanding the interaction between intestinal microorganisms and theimmune system of the body , it will provide new ideas for the diagnosis and treatment of some diseases.[Key words ] Intestinal microecology ;Autoimmune diseases ;Central nervous system diseases ;Cardiovascular diseases ;Tumors 肠道是人体与肠道微生物的重要屏障,一个成年人的肠道内约有1×1014个细菌,约占自身细胞的10 倍。
Gut Microbiota and Neurodegenerative Disorders The gut microbiota is a complex ecosystem of microorganisms that reside in the gastrointestinal tract. The microbiota plays a critical role in maintaining the health of the host organism, including regulating immune function, metabolism, and the central nervous system. Recent research has found that disruptions in the gut microbiota may be linked to the development of neurodegenerative disorders, such as Parkinson's disease and Alzheimer's disease.One perspective on the relationship between gut microbiota and neurodegenerative disorders is that the microbiota may play a role in the development and progression of these diseases. Studies have found that individuals with Parkinson's disease and Alzheimer's disease have different gut microbiota compositions compared to healthy individuals. Additionally, animal studies have shown that altering the gut microbiota can affect the development of neurodegenerative diseases. For example, mice that were given antibiotics to disrupt their gut microbiota showed increased levels of amyloid-beta protein, a hallmark of Alzheimer's disease.Another perspective is that neurodegenerative disorders may disrupt the gut microbiota, leading to further health complications. For example, individuals with Parkinson's disease often experience gastrointestinal symptoms, such as constipation and inflammation, which may alter the gut microbiota. Additionally, studies have found that individuals with Alzheimer's disease have increased intestinal permeability, which may allow harmful bacteria to enter the bloodstream and cause further inflammation.A third perspective is that the gut microbiota may provide a potential target for treatment or prevention of neurodegenerative disorders. Studies have found that probiotics and prebiotics, which can alter the gut microbiota, may have beneficial effects on cognitive function and brain health. Additionally, fecal microbiota transplantation, a procedure in which fecal matter from a healthy donor is transplanted into the gut of an individual with a disrupted microbiota, has shown promise in treating gastrointestinal symptoms in individuals with Parkinson's disease.However, there are also limitations to the current research on gut microbiota and neurodegenerative disorders. Many studies have been conducted on animal models, and it is unclear how well these findings translate to humans. Additionally, the mechanisms by which the gut microbiota may contribute to neurodegenerative diseases are not yet fully understood, and more research is needed to elucidate these mechanisms.Overall, the relationship between gut microbiota and neurodegenerative disorders is a complex and multifaceted one. While there is evidence to suggest that disruptions in the microbiota may be linked to the development and progression of these diseases, more research is needed to fully understand the mechanisms involved and to develop effective treatments and prevention strategies.。
肠道菌群通过脑肠轴影响肿瘤的发生发展胥婧;刘齐雨;李可;康玉;徐丛剑【摘要】正常情况下,人体肠道内菌群保持相对稳定的状态,共生菌、益生菌和病原菌的比例维持相对稳定.肠道菌群失调可改变肠道内的生理环境,还能通过各种途径引起人体肠外组织器官生理状态的改变,甚至影响肿瘤的发生发展.脑肠轴是其中一条通路,由免疫、代谢、神经内分泌和迷走神经等途径构成.本文旨在讨论肿瘤和肠道菌群之间的关系以及肠道菌群通过脑肠轴的4条途径改变机体的应激水平,从而影响肿瘤发生发展的相关机制,同时就肠道菌群调控在肿瘤治疗中的作用进行了探讨.%Under normal circumstances, the gut microbiota and the proportion of symbiotic bacteria, probiotics, and pathogens re-main relatively stable, but dysbiosis in the gut can change not only the physiological environment of the intestine but also the physio-logical state of some distant tissues and organs. This condition can even lead to tumor development. Gut-brain axis is a pathway through which gut microbiota can influence tumor growth and is associated with other pathways, such as immune, metabolic, neuro-endocrine, and vagal neural pathways. This paper reports the relationship between tumor growth and microbial groups in the gut and the mechanisms of gut dysbiosis inducing a chronic stress state via the gut–brain axis, which affects tumor development. This paper also presents the role of gut microbiota in tumor treatment.【期刊名称】《中国肿瘤临床》【年(卷),期】2017(044)017【总页数】4页(P886-889)【关键词】肠道菌群;脑肠轴;慢性应激;肿瘤【作者】胥婧;刘齐雨;李可;康玉;徐丛剑【作者单位】复旦大学附属妇产科医院中西医结合科上海市200011;复旦大学附属妇产科医院中西医结合科上海市200011;复旦大学附属妇产科医院中西医结合科上海市200011;复旦大学附属妇产科医院中西医结合科上海市200011;复旦大学附属妇产科医院中西医结合科上海市200011【正文语种】中文Abstract Under normal circumstances,the gut microbiota and the proportion of symbiotic bacteria,probiotics,and pathogens remain relatively stable,but dysbiosis in the gut can change not only the physiological environment of the intestine but also the physiological state of some distant tissues and organs.This condition can even lead to tumor development.Gut-brain axis is a pathway through which gut microbiota can influence tumor growth and is associated with other pathways,such as immune,metabolic,neuroendocrine,and vagal neural pathways.This paper reports the relationship between tumor growth and microbial groups in the gut and the mechanisms of gut dysbiosis inducing a chronic stress state via the gut–brain axis,which affects tumor development.This paper also presents the role of gut microbiota in tumor treatment. Keywords:gut microbiota,gut-brain axis,chronic stress,tumor肿瘤发生发展机制一直是研究的热点,受体内和体外多种信号通路直接或间接的调节,与机体的免疫代谢等生命活动密切相关。
急性胰腺炎患者肠道菌群变化及益生菌的应用吕彦青,李嘉杰,刘坤禹,郭汝华,季慧范吉林大学白求恩第一医院肝胆胰内科,长春 130021通信作者:季慧范,************.cn(ORCID: 0000-0001-8816-1906)摘要:肠道菌群是人体中最复杂和最重要的微生态系统,肠道菌群失调与急性胰腺炎的发生、发展密切相关,近年来靶向调控肠道微生态辅助治疗急性胰腺炎逐步被人们重视。
本文详细叙述了急性胰腺炎患者肠道菌群变化情况及机制,总结了益生菌应用的研究现状,指出益生菌辅助治疗方案的研究方向,并提出了预测急性胰腺炎患者优势菌群的新方法,以期为急性胰腺炎的治疗带来新思路。
关键词:胰腺炎,急性坏死性;胃肠道微生物组;鼠李糖乳杆菌基金项目:吉林市人口与健康发展研究(371170143428)Changes in intestinal microbiota and application of probiotics in patients with acute pancreatitisLYU Yanqing, LI Jiajie, LIU Kunyu, GUO Ruhua, JI Huifan.(Department of Hepatobiliary and Pancreatic Medicine, The First Hospital of Jilin University, Changchun 130021, China)Corresponding author: JI Huifan,************.cn(ORCID: 0000-0001-8816-1906)Abstract:Intestinal microbiota is the most complex and important microecosystem in the human body, and gut microbiota dysbiosis is closely associated with the development and progression of acute pancreatitis. Targeted regulation of intestinal microecology in assisting the treatment of acute pancreatitis has attracted more attention in recent years. This article describes the changes in intestinal microbiota and related mechanisms in patients with acute pancreatitis,summarizes the current research status of the use of probiotics, points out the research direction of probiotics as the adjuvant treatment regime, and proposes a new method for predicting the dominant flora in patients with acute pancreatitis, in order to bring new ideas for the treatment of acute pancreatitis.Key words:Pancreatitis, Acute Necrotizing; Gastrointestinal Microbiome; Lactobacillus RhamnosusResearch funding:Research on Population and Health Development of Jilin City (371170143428)急性胰腺炎(acute pancreatitis,AP)是多种病因导致胰腺组织自身消化所致的胰腺水肿、出血及坏死等炎症性损伤,组织学上以腺泡细胞破坏为特征。
肠道里的会议读后感英文回答:I recently read an article titled "The Gut Meeting: Insights from the Microbiome". It discussed the fascinating world of the gut microbiome and its impact on our overall health. The article provided insights into the gut microbiome's role in digestion, immune function, and even mental health.One of the key takeaways from the article was the importance of a diverse gut microbiome. A healthy gut is populated by a wide variety of bacteria, fungi, and other microorganisms. These microbes work together to break down food, absorb nutrients, and protect against harmful pathogens. Without a diverse microbiome, our digestive system may struggle to function properly.The article also highlighted the concept of "leaky gut syndrome". This occurs when the lining of the intestinesbecomes permeable, allowing toxins and bacteria to leakinto the bloodstream. This can lead to inflammation and various health issues. Maintaining a balanced gut microbiome is crucial in preventing leaky gut syndrome.Furthermore, the article discussed the gut-brain axis, which refers to the bidirectional communication between the gut and the brain. The gut microbiome produces neurotransmitters and other chemicals that can influence our mood, emotions, and even cognitive function. For example, serotonin, a key neurotransmitter involved in regulating mood, is primarily produced in the gut. This connection between the gut and the brain highlights the importance of maintaining a healthy gut for overall mental well-being.In addition to the scientific information, the article also included some interesting anecdotes. One story shared the experience of a woman who suffered from chronic digestive issues. After undergoing a fecal microbiota transplant, where healthy bacteria from a donor's stool is transferred to the patient's gut, her symptomssignificantly improved. This example demonstrated the potential of manipulating the gut microbiome to treatcertain health conditions.Overall, reading this article was eye-opening. It made me realize the immense importance of a healthy gut microbiome in maintaining overall health. It also shedlight on the potential for targeted interventions, such as probiotics or fecal transplants, to restore and optimizegut health.中文回答:我最近读了一篇标题为《肠道会议,来自微生物群落的洞察力》的文章。
基于“肠—肾轴”浅谈CKD与肠道微生态变化的关系发表时间:2016-04-26T13:30:07.583Z 来源:《健康文摘》2015年12期作者:付璐1,钱小方1,赵龙1, 陈明2 [导读] 1成都中医药大学临床医学院,成都 610072;2成都中医药大学附属医院肾病内科 “肠—肾轴”理论已有多年历史。
慢性肾脏疾病(chronic kidney disease,CKD)与肠道微生态密切相关且相互影响。
( 1成都中医药大学临床医学院,成都 610072;2成都中医药大学附属医院肾病内科,成都 610072)摘要:“肠—肾轴”理论已有多年历史。
慢性肾脏疾病(chronic kidney disease,CKD)与肠道微生态密切相关且相互影响。
本文概述了“肠—肾轴”的定义,肠、肾在能量物质代谢、免疫炎症、肠道黏膜及肠道细菌等方面的联系,简述“肠—肾轴”的中医认识,CKD患者肠道微生态的中、西医变化及其机制,肠道微生态对CKD及其并发症的影响机制等,综述CKD发生发展与肠道微生态变化关系的相关性的研究进展。
关键词:肠肾轴;中医;慢性肾脏疾病;肠道微生态中图分类号:R5 文献标志码:A 文章编号:Abcract:The Theory of Gut-kidney Axis has presence many years in the history,Chronic kidney disease(CKD)and intestinal microecology closely related and influence each other.This paper outlines the definition of “gut-kidney axis”, and the connection of intestinal and kidney in metabolism of energy substances,immune inflammation,intestinal mucosa and intestinal bacterria;saummarizing the understanding of “gut-kidney axis” with traditional Chinese medicine,the transformation and mechanism of intestinal microecology of the patients with CKD in western medicine and traditional Chinese medicine,and the intestinal microecology on the infuence mechanism of CKD and its complications,and so on.Overview the correlation of research progress on the relationship between the occurrence and development of CKD and the changes of intestinal microecology. Keyword: The gut-kidney axis; Chronic kidney axis; Intestinal microecology [中图分类号] R25 [文献标识码] A 文章编号:肠道微生态中厌氧菌、兼性厌氧菌占细菌总数99%以上,其中专性厌氧杆菌与双歧杆菌占90%以上。
Gut Microbiota and Liver DiseasesThe gut microbiota plays a crucial role in the maintenance of human health. It is a complex ecosystem of microorganisms that resides in the gastrointestinal tract and helps in the digestion and absorption of nutrients. However, recent studies have shown that alterations in the gut microbiota composition can lead to the development of liver diseases.Liver diseases are a major health concern worldwide, and they can be caused by a variety of factors such as alcohol consumption, viral infections, and metabolic disorders. The gut microbiota has been shown to play a role in the pathogenesis of liver diseases such as non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), and liver cirrhosis.NAFLD is a condition in which fat accumulates in the liver, leading to inflammation and damage to liver cells. Studies have shown that alterations in the gut microbiota composition can contribute to the development of NAFLD. The gut microbiota is involved in the metabolism of dietary nutrients, and changes in its composition can lead to an imbalance in the production of short-chain fatty acids (SCFAs) and bile acids, which can contribute to the development of NAFLD.ALD is a condition in which the liver is damaged due to excessive alcohol consumption. The gut microbiota has been shown to play a role in the development of ALD. Alcohol consumption can lead to alterations in the gut microbiota composition, which can contribute to the production of harmful metabolites such as endotoxins and acetaldehyde. These metabolites can lead to inflammation and damage to liver cells, contributing to the development of ALD.Liver cirrhosis is a condition in which the liver is damaged and scarred, leading to a loss of liver function. The gut microbiota has been shown to play a role in the development of liver cirrhosis. Studies have shown that alterations in the gut microbiota composition can contribute to the production of harmful metabolites such as ammonia, which can lead to inflammation and damage to liver cells, contributing to the development of liver cirrhosis.In addition to the role of the gut microbiota in the development of liver diseases, there is also evidence to suggest that the gut microbiota can influence the response to treatment for liver diseases. For example, studies have shown that the gut microbiota can influence the efficacy of treatments for hepatitis C virus (HCV) infection and liver transplantation.In conclusion, the gut microbiota plays a crucial role in the development and progression of liver diseases. Alterations in the gut microbiota composition can contribute to the development of NAFLD, ALD, and liver cirrhosis. Furthermore, the gut microbiota can influence the response to treatment for liver diseases. Therefore, further research is needed to better understand the role of the gut microbiota in the pathogenesis of liver diseases and to develop new therapies that target the gut microbiota to treat liver diseases.。
C URRENTO PINION Gut microbiota and host defense in critical illnessMax C.Jacobs a,Bastiaan W.Haak a,Floor Hugenholtz a,and W.Joost Wiersinga a,bPurpose of reviewThe review aims to discuss emerging evidence in the field of microbiome-dependent roles in host defenseduring critical illness with a focus on lung,kidney,and brain inflammation.Recent findingsThe gut microbiota of critical ill patients is characterized by lower diversity,lower abundances of keycommensal genera,and in some cases overgrowth by one bacterial genera,a state otherwise known asdysbiosis.Increasing evidence suggests that microbiota-derived components can reach the circulatorysystem from the gut and modulate immune homeostasis.Dysbiosis might have greater consequences for thecritically ill than previously imagined and could contribute to poor outcome.Preclinical studies suggest thatimpaired communication across the gut–organ axes is associated with brain,lung–and kidney failure.SummaryIn health,a diverse microbiome might enhance host defense,while during critical illness,the dysbioticmicrobiome might contribute to comorbidity and organ dysfunction.Future research should be aimed atfurther establishing the causes and consequences of dysbiosis seen in the critically ill,which will provideperspective for developing new strategies of intervention.Keywordscritically ill,gut microbiota,host defense,innate immunity,ICUINTRODUCTIONThe gut microbiota refers to the community of microbes that reside in our gastrointestinal tract, with whom we have developed a symbiotic relation-ship over the course of millions of years.These microbes,as we have learned in recent times,con-tribute to various physiological processes that are crucial for human homeostasis and health[1].These observations have guided the current paradigm that the gut microbiome could in fact be perceived as the latest discovered human organ.Critically ill patients in the ICU could potentially benefit from more insight into this nascent area of research,as virtually all of these patients will have a perturbed micro-biome.On any given day almost three-quarter of the patients treated on the ICU will receive antibiotics [2].Although antibiotics are administered to fight pathogens and/or to try to decrease the occurrence of nosocomial infections,we are becoming increas-ingly aware that this strategy may simultaneously open the door to a new and uncharted problem:the coinciding depletion of commensal gut bacteria[3].Despite the heterogeneity of disease states among critically ill patients,it has become apparent that ICU treatment is associated with an extreme and long-lasting shift in microbiome composition,with potentially negative impact on both short and long-term outcome[4&].Unfortunately,clinical research assessing therapeutic modalities that could strengthen the microbiome of patients on the ICU are sparse.This is probably in part explained be a clear lack of mechanistic insights on how a healthy and stable microbiome confers protection against (opportunistic)pathogens as well as organ damage.The review aims to discuss emerging evidence in the field of microbiome-dependent roles in host defense during critical illness with a focus on lung, kidney,and brain inflammation.Ultimately,we aim to breach the gap on how these insights could be used in the development of novel microbiome-based diagnostics and treatment modalities that a Center for Experimental and Molecular Medicine and b Division of Infectious Diseases,Department of Medicine,Academic Medical Center, University of Amsterdam,Amsterdam,the Netherlands Correspondence to Max C.Jacobs,Center for Experimental and Mol-ecular Medicine,Academic Medical Center,University of Amsterdam, Meibergdreef9,Room F0–117,1105AZ Amsterdam,the Netherlands. Tel:+31205665247;e-mail:m.c.jacobs@amc.uva.nl;w.j.wiersinga@amc.uva.nlCurr Opin Crit Care2017,23:000–000DOI:10.1097/MCC.0000000000000424REVIEWcould potentially enhance host defense in critically ill patients.FUNCTION OF THE GUT MICROBIOME: BASICSThe intestinal microbiota are hard to cultivate and study in the laboratory;so far up to77%of the bacteria in the gut have been cultivated[5].How-ever,the fast development of novel molecular tech-niques in the last few decades has considerably increased our insights into this complex ecosystem [6].In the large intestine the phyla Bacteroidetes and Firmicutes dominate and Actinobacteria,Pro-teobacteria,and Verrucomicrobia are important and lower abundance phyla[7].These gut microbiota degrade dietary plant polysaccharides and–in a lesser degree–proteins,which are fermented into a wide range of metabolites.The main end products of fermentation are short-chain fatty acids(SCFA), such as butyrate,acetate,and propionate.The SCFA profiles produced by members of the two dominant phyla differ,with a tendency for butyrate pro-duction by members of the Firmicutes phyla,while propionate production tends to be dominated by Bacteroidetes[8].SCFA play an essential role in the maintenance of colonic integrity and metabolism, as it has been shown that butyrate serves as the main energy source for the colonocytes[9,10].The optimal composition of a healthy microbiome is under debate[11];however,general features have been noted.Diversity indices need to be relatively high,where bacterial groups of Firmicutes,Bacter-oidetes,and Actinobacteria provide the majority of the richness(that is the number of different bacterial species observed;see Fig.1for an overview of the major taxonomic groups).Proteobacteria are usually also present,albeit in low numbers and preferably low richness.The above-mentioned features need to be in balance,and when this balance shifts we speak of a disturbed homeostasis or dysbiosis.Of special interest is the relationship of the gut microbiome in the host defense against specific pathogens,the role of which we are only just begin-ning to uncover.Within the gut,an intact micro-biome is able to maintain stability and homeostasis through numerous mechanisms.First,our commen-sals are able to directly outcompete intestinal patho-gens for available nutrients,or even kill potential invaders through the production of defensins and production of signaling(quorum sensing)mol-ecules[12&].Additionally,the microbiota are potent inducers of the immune system,which in turn target potential invaders in the gut.Elimination of these commensal bacteria by external factors,for example,antibiotics,is therefore hypothesized to lead to overgrowth of opportunistic pathogens[13]. THE GUT MICROBIOME DYNAMICS IN CRITICALLY ILL PATIENTSCritically ill patients admitted to the ICU have severely altered and unstable microbiomes [4&,14&&,15,16,17&&].However,so far,the number and types(e.g.,septic,nonseptic,location of sepsis) of ICU patients in clinical studies that investigated the microbiome have been limited.When compared to healthy controls,the gut microbiome of patients with sepsis is characterized by lower diversity,lower abundances of key commensal genera(such asKEY POINTSCritically ill patients have a severely perturbed gut microbiome.This is characterized by a loss of diversity, and susceptibility to overgrowth ofopportunistic pathogens.Dysbiosis can have a myriad of effects,which should be further investigated in critically ill patients.In general,it is suggested that the microbiome in a healthy setting might enhance host defense,whereas during illness,the dysbiotic gut microbiome might contribute to poor outcome.Impaired communication across the gut organ axes has been associated with organ dysfunction.It is warranted to translate the enormous amount of associative findings into causative mechanisms to better understand the immunomodulatory role of the microbiome in critical illness.The gut microbiome remains a high potential future therapeutic or diagnostic target.However,current evidence into microbiota-targeted therapies in critical illness remains sparse andambiguous.FIGURE1.Schematic overview of the microbiome gradient between homeostasis in healthy people and dysbiosis in critical ill.Metabolic supportFaecalibacterium,Blautia,Ruminococcus),and some-times overgrowth(over50%relative abundance)by one genera,like Clostridium difficile,Staphylococcus spp.,Escherichia spp.,Shigella spp.,Salmonella spp., and Enterococcus spp.[4&,15,16,14&&].The occurrence of the last feature–overgrowth by a single bacterial species–is only observed in roughly a third of cases, but does seem to increase with the time a patient stays on the ICU[14&&,15].Explanations for gut micro-biome dysbiosis in critically ill patients include(but are not limited to)the use of(par)enteral feeding, gastric acid inhibitory drugs,and antibiotics[18&]. Additionally,mechanical ventilation was recently shown to be an influencer of microbiome dynamics and,most probably as a result,the susceptibility to nosocomial infection[19&].DISTANT EFFECT OF THE GUT MICROBIOME DURING CRITICAL ILLNESS: THE CONCEPT OF THE GUT–ORGANAXESCurrently,it is hypothesized that microbiota-derived components,such as pathogen-associated molecular patterns and metabolites that derive from the gut can reach the circulatory system and interact at the systemic level to influence immune homeo-stasis[20].Murine models exerting manipulated gut microbiomes through breeding in sterile environ-ments(i.e.,germ-free mice),supplementation of antibiotics,or altered diets have been instrumental in obtaining these insights.Germ-free mice exhibit decreased numbers of immune cells residing in bone marrow,which was explained to be resulting from aberrant toll-like receptor signaling[21].Further-more,hematopoietic abnormalities seen in micro-biome depleted or germ-free mice models include aberrations in granulopoiesis and myelopoiesis as well as T-cell development,which,in turn,have been linked to deteriorated immunity against viruses,bacteria,and fungi[22–27].Diet is key in shaping the ecosystem of the gut microbiome and important in host metabolism[28,29&].Fiber-rich diets have demonstrated positive effects in intesti-nal barrier integrity,amelioration of intestinal inflammatory disease,and even protection against invading pathogens and sepsis in preclinical models [30–32].Of note,nondigestible dietary fiber intake is correlated to levels of circulating SCFA,which are implicated as important factors in pathogenesis of various inflammatory diseases(see review[33&]). Furthermore,in a murine study investigating the association between obesity and susceptibility to asthma,it has been suggested that a high-fat diet might result in impaired immune activation of air-way dendritic and regulatory T cells[34].Despite the mounting evidence emerging from preclinical models on the role of the gut microbiome in the host defense against inflammation,surpris-ingly little studies have been conducted in humans. One comprehensive study in a cohort of500healthy adults of whom fecal and blood samples were col-lected indicated that variations in cytokines responses,that is,interleukin1b,interleukin6, interleukin17,and interleukin22,tumor necrosis factor a and interferon g,are to some extent linked to specific gut microbiome organisms and sub-sequent functions[35&&].The authors suggested that host defense modulation most likely is orchestrated in part through specific gut microbiome-derived metabolites.In line,a small study conducted in 12healthy human adult study participants who were treated for1week with broad-spectrum anti-biotics,showed that gut microbiome depletion resulted in an impaired production of tumor necrosis factor a,in peripheral blood mononuclear cells,and interleukin6and interleukin1b,in whole blood,after ex-vivo lipopolysaccharide stimulation [36].Cytokine responses returned to normal6weeks after cessation of the antibiotics.However,a follow-up intervention trial in which healthy human study participants were pretreated with antibiotics prior to the administration of lipopolysaccharide intrave-nously showed that gut microbiota disruption by broad-spectrum antibiotics does not affect systemic innate immune responses during human endotox-emia[37&].These novel insights on the role of the microbiome in systemic immunity are increasingly being used in disease research,resulting in the development of theories of gut microbiome to organ interactions,popularly coined gut–organ axes. Gut–lung axisThe gut–lung axis refers to the crosstalk that is hypothesized to take place between the gut micro-biota and the lungs which might be important in diseases ranging from allergic asthma to lung infec-tion(reviewed[38&]).Evidence that the gut and the lung were closer connected than was initially thought emerged over20years ago from studies in rat models of trauma and hemorrhagic shock (THS)[39].It was demonstrated that during THS, gut-derived compounds translocate through mesen-teric lymph and cause distal lung injury[40].This phenotype could be rescued by ligation of the thora-cic lymph duct and,furthermore,it was shown that mesenteric lymph from postshock rats transferred into naı¨ve animals again provokes distal organ injury[41,42].Interestingly,vagal nerve stimulation was shown to decrease gut barrier permeability and abrogates THS-induced distal organ injury[43]. Gut microbiota and host defense in critical illness Jacobs et al.FIGURE 2.Schematic overview of gut microbiome states and their proposed effect on systemic host defense in critical illness;an example of the gut–lung axis.(a)The healthy gut microbiome resides in the intestinal lumen and comprises a diverse set of commensal bacteria that interact with one another and the mensal bacteria can metabolize dietary fiber andproduce short-chain fatty acids (SCFA)which provide an important energy source for intestinal epithelial cells.Besides,SCFA,pathogen-associated molecular patterns and bacteria themselves are believed to be translocated into the lamina propria andMetabolic supportConversely,a protective role of the gut microbiome along the gut–lung axis was suggested in several murine models of bacterial pneumonia and sepsis. Gut microbiome dysbiosis,caused by antibiotic administration,resulted in impaired immune acti-vation and increased mortality during murine Pneu-mococcal pneumonia[44].Additionally,it was demonstrated that specific absence of segmented filamentous bacteria in the gut of mice led to impaired Th17mucosal immunity and suscepti-bility to Staphylococcus aureus pneumonia[45]. Furthermore,recent work has described a crucial window of early exposure to and colonization with intestinal commensal bacteria in the effective devel-opment of innate lymphoid cell-mediated immun-ity against bacterial pneumonia in neonatal mice [46].Finally,translocation of gut bacteria along the gut–lung axis was recently reported in human study participants.[47&&].Sequencing provided evidence of lung microbiome enrichment with gut-associated bacteria in both mice suffering of experimental sepsis and human patients with acute respiratory distress syndrome,but not healthy controls.Gut–brain axisRecent research has shown extensive crosstalk between the gut microbiome and the brain,that is, through neuropeptides or endocrine,immune,and nerve signaling[48,49],which have resulted in the suggestion that the gut microbiome might play a role in cognitive function and behavior[50].Also,in critically ill patients this so-called gut–brain axis can be of importance.Mouse models showed that microbiota depletion could ameliorate outcome after ischemic brain injury,which was attributed to defec-tive trafficking of specific types of T cells to the ischemic brain area[51].Surprisingly,a similar study in mice showed that acute brain injury itself resulted in gut microbiome dysbiosis,intestinal barrier dys-function,and reduced intestinal motility,which underscores the notion that gut–brain communi-cation is bidirectional[52&].Of interest,(secondary) infections,mainly pneumonia,are a common medical complication and a leading cause of post-stroke death not directly attributable to stroke itself [53].However,conventional cultures of sputum derived from poststroke patients with lung infection samples are often negative suggesting that anaerobic bacteria might play a role.This,in turn,suggests that secondary pulmonary infection in poststroke patients could very well be gut derived.In correspondence,it was reported that gut-derived bacteria to comprise 70%of cultures samples taken from poststroke patients with secondary lung infection[54&&].These results were confirmed and expanded in mice exper-iments.Therein,it was shown that stroke leads to impaired intestinal barrier function,which could be causative for gut commensals to spread systemically and cause infection.Excitingly,the b-adrenergic receptor blocker propranolol could improve barrier function,which coincided with marked reduction of poststroke secondary infection[54&&].Gut–kidney axisThe overall incidence of acute kidney injury(AKI)in ICU patients ranges from20to50%[55].Recent murine studies have implicated a role for the gut microbiome and its derived metabolites in the out-come of this condition.It was shown that therapy with the SCFA’s acetate,propionate,and butyrate, was protective in an ischemia/reperfusion model of AKI injury[56&].Interestingly,another study using a similar model showed that gut microbiome depletion before induction resulted in protection against renal dysfunction,damage,and inflam-mation[57],suggestive of a deleterious effect of ‘normal’gut microbiome during AKI.For an overview of the proposed role of various gut microbiome states in immune priming and susceptibility to(secondary)infections,see Fig.2. CONCLUSION:THE ROAD AHEADAlthough the knowledge of the role of the gut microbiome in critical illness is increasing rapidly, numerous questions remain to be answered.One ofcirculatory systems were they contribute to priming of the immune system,(b)A singular course of antibiotics(depicted by the dark circles showing a white(a)may be prescribed to fight infections.Sensitive commensal bacteria can be depleted leading to reduction in diversity.This results in less effective production of SCFA,which may contribute to decreased local epithelial cell condition and reduced translocation and immune priming.Reduced immune priming may cause susceptibility to secondary infections(c)In a scenario were infections are severe or recurrent,repeated,or broad-spectrum antibiotics treatment can result in marked reduction in commensal bacteria and dysbiosis.This can lead to overgrowth of(opportunistic)pathogens that can lead to local infections and immune problems.Impaired or absent production of SCFA can contribute to intestinal epithelium damage and reduced barrier function,and impaired immune priming.Translocation of pathogenic bacteria through leaky barrier may contribute to secondary infections.(c)The poststroke gut microbiome becomes(e.g.,with concomitant antibiotics treatment)dysbiotic.Also,stroke can lead to decreased barrier function.In this condition commensal bacteria may translocate in systemic circulation,turn pathogenic and cause secondary infections.Gut microbiota and host defense in critical illness Jacobs et al.the most pressing unanswered question is what comprises a healthy gut microbiome,and to what extent is microbiota composition generalizable on a population wide scale?To begin to answer these questions a multidisciplinary scientific approach is required,as it entails wide ranges of fundamental and medical biology.Furthermore,it will be of particular interest to learn which gut microbiome species and metabolites account for the hypothes-ized effects on host health.What can be stated is that in a healthy situation at least two factors appear to be present,that is,a diverse,symbiotic gut microbiome and an intact intestinal barrier.We have discussed research wherein it was shown that environmental factors, for example,diet and oral antibiotics,as well as critical disease itself(or its treatment)can influence either one or both of these factors,which in turn might contribute to an unfavorable outcome.Para-doxically,when intestinal barrier function is com-promised,which is seen in sepsis or after stroke,an otherwise‘healthy’microbiome might be account-able for medical complications such as secondary infections.Overall it appears that–at least for now –no simple black and white answers can be formu-lated with respect to the gut microbiome’s role in the host defense during critical illness.However, despite reasonable gaps in our knowledge base,cur-rent therapeutic developments based on the manip-ulation of gut microbiomes are showing great potential.The success story of the usage of fecal microbiota transplantation to fight recurrent C.dif-ficile infection–also in the ICU setting–serves as a leading example[58&].Other therapeutic develop-ments were reviewed here recently[59&].Now that the field moves from association studies toward establishing causation it will be of importance to see where these scientific endeavors will bring us in the near future.Particularly,whether we will be able to learn from fundamental studies into microbiome–host interaction and(pre)clinical studies to the point that they initiate the development of novel strategies to manipulate gut microbiomes of patient to ameli-orate clinical outcome of critically ill patients suffer-ing from sepsis or brain injury.Overall,a concept emerges that the gut microbiome makes for a high potential target for intervention to improve clinical outcome.Future research should be aimed at estab-lishing causation in gut microbiome dysbiosis seen in critically ill,which will provide perspective for devel-oping new strategies of intervention. AcknowledgementsWe would like to thank our colleagues of the Marie Curie Innovative Training Network(MC-ITN)European Sepsis Academy for fruitful discussions leading to this article.Financial support and sponsorshipThe work was supported by the Netherlands Organiz-ation for Scientific Research(NWO).Conflicts of interestThere are no conflicts of interest.REFERENCES AND RECOMMENDED READINGPapers of particular interest,published within the annual period of review,have been highlighted as:&of special interest&&of outstanding interest1.Sekirov I,Russell SL,Antunes LCM,Finlay BB.Gut microbiota in health anddisease.Physiol Rev2010;90:859–904.2.Vincent J,Marshall J,Anzueto A,et al.,EPIC II group of investigators.International study of the prevalence and outcomes of infection in intensive care units.JAMA2009;302:2323–2329.3.Blaser M.Antibiotic overuse:stop the killing of beneficial bacteria.Nature2011;476:393–394.4.&Wischmeyer PE,McDonald D,Knight R.Role of the microbiome,probiotics, and‘dysbiosis therapy’in critical illness.Curr Opin Crit Care2016;22:347–353.Excellent review on current status of microbiota targeted therapies,such as novel probiotics and fecal transplants.gier JC,Khelaifia S,Alou MT,et al.Culture of previously unculturedmembers of the human gut microbiota by culturomics.Nat Microbiol2016;1:16203.6.Rajilic-Stojanovic M,Smidt H,de Vos WM.Diversity of the humangastrointestinal tract microbiota revisited.Environ Microbiol2007;9:2125–2136.7.Rajilic-Stojanovic M,de Vos WM.Thefirst1000cultured species ofthe human gastrointestinal microbiota.FEMS Microbiol Rev2014;38:996–1047.8.Mun˜oz-Tamayo R,Laroche B,Walter E´,et al.Kinetic modelling of lactateutilization and butyrate production by key human colonic bacterial species.FEMS Microbiol Ecol2011;76:615–624.9.Cook SI,Sellin JH.Review article:short chain fatty acids in health anddisease.Aliment Pharmacol Ther1998;12:499–507.10.Morrison DJ,Preston T.Formation of short chain fatty acids by the gutmicrobiota and their impact on human metabolism.Gut Microbes2016;7:189–200.11.Zhernakova A,Kurilshikov A,Bonder MJ,et al.,LifeLines cohort study.Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity.Science2016;352:565–569.12.&Klingensmith NJ,Coopersmith CM.The gut as the motor of multiple organ dysfunction in critical illness.Crit Care Clin2016;32:203–212.Excellent overview of the role of the gut and gut microbiota in multiorgan failure and critical illness.13.Pamer EG.Resurrecting the intestinal microbiota to combat antibiotic-resis-tant pathogens.Science2016;352:535–538.14.&&Lankelma JM,van Vught LA,Belzer C,et al.Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation:a pilot study.Intensive Care Med2017;43:59–68.Recent pilot study to investigate the intestinal microbiota in critical ill patients,with a special focus to septic patients.Here,critical ill patients had less or no abundances of butyrate-producing genera in their intestinal microbiota compared with human volunteers.15.Zaborin A,Smith D,Garfield K,et al.Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during pro-longed critical illness.MBio2014;5:e01361–e1414.16.Ojima M,Motooka D,Shimizu K,et al.Metagenomic analysis reveals dynamicchanges of whole gut microbiota in the acute phase of intensive care unit patients.Dig Dis Sci2016;61:1628–1634.17.&&McDonald D,Ackermann G,Khailova L,et al.Extreme dysbiosis of the microbiome in critical illness.mSphere2016;1:.doi:10.1128/mSphere.00199-16.Editor.First study to sample fecal,oral,and skin for microbiota analysis in critical ill patients admitted at the ICU within2days and at10days or discharge.Indicated increasing microbiota dysbiosis in the patients during their stay,with health promoting commensal disappearing and overgrowth of pathogens.18.&Haak BW,Wiersinga WJ.The role of the gut microbiota in ncet Gastroentrol Hepatol2017;2:135–143.Excellent overview of the latest studies on the role of the intestinal microbiota in (acquiring)sepsis.Metabolic support19. &Zakharkina T,Martin-loeches I,Matamoros S,et al.The dynamics of the pulmonary microbiome during mechanical ventilation in the intensive care unit and the association with occurrence of pneumonia.Thorax2017.Interesting study in ICU patients showing the effect of mechanical ventilation on the respiratory microbiome.20.Levy M,Blacher E,Elinav E.Microbiome,metabolites and host immunity.CurrOpin Microbiol2016;35:8–15.21.Balmer ML,Schurch CM,Saito Y,et al.Microbiota-derived compounds drivesteady-state granulopoiesis via MyD88/TICAM signaling.J Immunol2014;193:5273–5283.22.Josefsdottir KS,Baldridge MT,Kadmon CS,King KY.Antibiotics impairmurine hematopoiesis by depleting intestinal microbiota.Blood2016;129:729–739.23.Khosravi A,Ya´n˜ez A,Price JG,et al.Gut microbiota promote hematopoiesis tocontrol bacterial infection.Cell Host Microbe2014;15:374–381.24.Trompette A,Gollwitzer ES,Yadava K,et al.Gut microbiota metabolism ofdietaryfiber influences allergic airway disease and hematopoiesis.Nat Med 2014;20:159–166.25.Zhang D,Chen G,Manwani D,et al.Neutrophil ageing is regulated by themicrobiome.Nature2015;525:528–532.26.Abt MC,Osborne LC,Monticelli LA,et mensal bacteria calibrate theactivation threshold of innate antiviral immunity.Immunity2012;37:158–170.27.McAleer JP,Nguyen NL,Chen K,et al.Pulmonary Th17antifungal immunity isregulated by the gut microbiome.J Immunol2016;197:97–107.28.Sonnenburg ED,Smits SA,Tikhonov M,et al.Diet-induced extinctions in thegut microbiota compound over generations.Nature2016;529:212–215.29. &Sonnenburg JL,Ba¨ckhed F.Diet-microbiota interactions as moderators of human metabolism.Nature2016;535:56–64.Excellent updated review on how the diet,microbiota,and its metabolites interact with the host and play a crucial role in the host metabolism.30.Monk JM,Lepp D,Wu W,et al.Chickpea-supplemented diet alters the gutmicrobiome and enhances gut barrier integrity in C57Bl/6male mice.J Funct Foods2017;In press.31.Morowitz MJ,Di Caro V,Pang D,et al.Dietary supplementation with non-fermentablefiber alters the gut microbiota and confers protection in murine models of sepsis.Crit Care Med2017;45:e516–e523.32.Monk JM,Lepp D,Zhang CP,et al.Diets enriched with cranberry beans alterthe microbiota and mitigate colitis severity and associated inflammation.J Nutr Biochem2016;28:129–139.33. &Richards JL,Yap YA,McLeod KH,et al.Dietary metabolites and the gut microbiota:an alternative approach to control inflammatory and autoimmune diseases.Clin Transl Immunol2016;5:e82–e182.The review summarizes how diet,microbiota and SCFAs can modulate the progression of inflammatory diseases and autoimmunity.34.Pizzolla A,Oh DY,Luong S,Prickett SR.High fat diet inhibits dendritic cell andT cell response to allergens but does not impair inhalational respiratory tolerance.PLoS One2016;11:1–18.35. &&Schirmer M,Smeekens SP,Vlamakis H,et al.Linking the human gut microbiome to inflammatory cytokine production capacity.Cell2016; 167:1125–1136.Human cohort study showing that microbiome-dependent metabolic pathways drive distinct immune responses in healthy volunteers.nkelma JM,Belzer C,Hoogendijk AJ,et al.Antibiotic-induced gut micro-biota disruption decreases TNF-a release by mononuclear cells in healthy adults.Clin Transl Gastroenterol2016;7:e186.37. &Lankelma JM,Cranendonk DR,Belzer C,et al.Antibiotic-induced gut micro-biota disruption during human endotoxemia:a randomised controlled study. Gut2016;1–8.Epub ahead of print.The study indicated that the systemic innate immune responses are not affected by broad spectrum antibiotics in healthy volunteers.38. &Budden KF,Gellatly SL,Wood DL,et al.Emerging pathogenic links between microbiota and the gut-lung axis.Nat Rev Microbiol2017;15:55–63.Excellent review on the link of the microbiota in the gut–lung axis.39.Magnotti LJ,Upperman JS,Xu DZ,et al.Gut-derived mesenteric lymph but notportal blood increases endothelial cell permeability and promotes lung injury after hemorrhagic shock.Ann Surg1998;228:518–527.40.Deitch EA,Xu DZ,Lu Q.Gut lymph hypothesis of early shock and trauma-induced multiple organ dysfunction syndrome:a new look at gut origin sepsis.J Organ Dysfunct2006;2:70–79.41.Wohlauer MV,Moore EE,Harr J,et al.Cross-transfusion of postshockmesenteric lymph provokes acute lung injury.J Surg Res2011;170:314–318.42.Reino DC,Pisarenko V,Palange D,et al.Trauma Hemorrhagic shock-inducedlung injury involves a Gut-Lymph-induced TLR4pathway in mice.PLoS One 2011;6:e14829.43.Levy G,Fishman JE,Xu D,et al.Vagal nerve stimulation modulates gut injuryand lung permeability in trauma-hemorrhagic shock.J Trauma Acute Care Surg2012;73:338–342.44.Schuijt TJ,Lankelma JM,Scicluna BP,et al.The gut microbiota plays aprotective role in the host defence against pneumococcal pneumonia.Gut 2015;65:575–583.45.Gauguet S,Ortona SD,Ahnger-pier K,et al.Intestinal microbiota of miceinfluences resistance to Staphylococcus aureus pneumonia.Infect Immun 2015;83:4003–4014.46.Gray J,Oehrle K,Worthen G,et al.Intestinal commensal bacteria mediatelung mucosal immunity and promote resistance of newborn mice to infection.Sci Transl Med2017;9:1–14.47.&&Dickson RP,Singer BH,Newstead MW,et al.Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome.Nat Microbiol2016;1:16113.The study reports evidence that the lung microbiome is enriched with intestinal microbiota both in a murine model of sepsis and in humans with acute respiratory distress syndrome.The authors demonstrate that these diseases have potentially a shared mechanism of pathogenesis.48.Holzer P,Farzi A.Neuropeptides and the microbiota-gut-brain axis.Adv ExpMed Biol2014;817:195–219.49.Carabotti M,Scirocco A,Maselli MA,Severi C.The gut-brain axis:Interactionsbetween enteric microbiota,central and enteric nervous systems.Ann Gas-troenterol2015;28:203–209.50.Foster JA,McVey Neufeld KA.Gut-brain axis:how the microbiome influencesanxiety and depression.Trends Neurosci2013;36:305–312.51.Benakis C,Brea D,Caballero S,et mensal microbiota affectsischemic stroke outcome by regulating intestinal gd T cells.Nat Med 2016;22:516–523.52.&Singh XV,Roth XS,Llovera G,et al.Microbiota dysbiosis controls the neuroinflammatory response after stroke.J Neurosci2016;36:7428–7440.Demonstration of a bidirectional association between the gut microbiome and the brain in a model of stroke.53.Chamorro A´,Urra X,Planas AM.Infection after acute ischemic stroke:amanifestation of brain-induced immunodepression.Stroke2007;38:1097–1103.54.&&Stanley D,Mason LJ,Mackin KE,et al.Translocation and dissemination of commensal bacteria in poststroke infection.Nat Med2016;22:1277–1284.A comprehensive study showing evidence that the gut microbiome might be animportant source of secondary infections after stroke.55.Case J,Khan S,Khalid R,Khan A.Epidemiology of acute kidney injury in theintensive care unit.Crit Care Res Pract2013;2013:479730.56.&Andrade-Oliveira V,Amano MT,Correa-Costa M,et al.Gut bacteria products prevent AKI induced by ischemia-reperfusion.J Am Soc Nephrol2015;26:1877–1888.Interesting study suggesting at a therapeutic role for SCFAs in a murine model of AKI.57.Emal D,Rampanelli E,Stroo I,et al.Depletion of gut microbiota protectsagainst renal ischemia-reperfusion injury.J Am Soc Nephrol2016;28:1–12.58.&van Nood E,Vrieze A,Nieuwdorp M,et al.Duodenal infusion of donor feces for recurrent Clostridium difficile.N Engl J Med2013;368:407–415.Clinical trial showing fecal microbiota transplants benificial in patients with recurrent C.difficile infection.59.&Haak BW,Levi M,Wiersinga WJ.Microbiota-targeted therapies on the intensive care unit.Curr Opin Crit Care Rev2017;23:167–174.Recent overview on current and new possibilities for microbiota-targeted therapies.Gut microbiota and host defense in critical illness Jacobs et al.。
You Are What You EatIntroductionThe saying “You are what you eat” emphasizes the importance of a healthy and balanced diet in maintaining overall well-being. Our food choices play a crucial role in shaping our physical health, mental state, and even our personalities. This article delves into the intricate relationship between our diet and our identity, exploring how what weeat impacts various aspects of our lives.The Physical Impact1.Nutrient Intake: The nutrients we consume directly affect ourphysical health. A well-rounded diet consisting of essentialvitamins, minerals, proteins, carbohydrates, and fats ensuresoptimal bodily functions. On the other hand, a poor diet lackingin these nutrients can lead to deficiencies and various healthproblems, such as weak immune system, fatigue, and impaired growth.2.Weight Management: Our food choices significantly impact ourweight. Consuming excessive amounts of high-calorie, processedfoods can lead to weight gain and obesity, while a balanced andnutritious diet helps maintain a healthy weight. An adequateintake of fruits, vegetables, whole grains, and lean proteinspromotes satiety, prevents overeating, and supports weightmanagement.3.Physical Performance: Whether it’s an athlete striving for peakperformance or an individual engaging in daily activities, a well-planned diet directly affects physical performance. Providing thebody with the right fuel, such as complex carbohydrates forsustained energy and protein for muscle recovery, enhances stamina, strength, and endurance.The Mental Connection1.Brain Health: The food we consume influences our cognitiveabilities and mental well-being. Omega-3 fatty acids found in fish,nuts, and seeds, for instance, support brain health and improvemood. Conversely, a diet high in added sugars and processed foodsmay contribute to cognitive decline and increase the risk ofmental health disorders, such as depression and anxiety.2.Focus and Concentration: Certain foods and nutrients have adirect impact on our cognitive functions, including focus andconcentration. Incorporating sources of antioxidants, such asberries and dark chocolate, and essential vitamins like B vitamins and iron can enhance brain function, memory, and attention span.3.Gut-Brain Axis: Emerging research has highlighted the intricatelink between our gut and brain, forming what is known as the gut-brain axis. The food we eat affects our gut microbiota, thecollection of microorganisms residing in our digestive system.These microbes produce neurotransmitters that influence our mood,emotions, and mental health. A diet rich in fiber, fermented foods, and probiotics supports a healthy gut microbiome and improvesoverall mental well-being.The Emotional Aspectfort Eating: Many individuals turn to food for emotionalcomfort, seeking solace in certain types of food during times ofstress, sadness, or loneliness. This phenomenon, known as comforteating, can lead to overeating and weight gain. Recognizingemotional triggers and developing alternative coping strategies is essential for maintaining a healthy relationship with food.2.Food Preferences and Identity: Our food preferences are ofteninfluenced by cultural background, upbringing, and personalbeliefs. What we choose to eat reflects a part of our identity and may evoke feelings of nostalgia, connection, or pride. Exploringdifferent cuisines and embracing diverse food choices can broadenour horizons and foster cultural understanding.3.Mindful Eating: Mindful eating involves paying full attention tothe eating experience, including the taste, texture, and aroma offood. It emphasizes awareness of hunger and fullness cues,fostering a healthier relationship with food. Practicingmindfulness while eating can enhance enjoyment, reduce emotionaleating, and promote overall well-being.Conclusion“You are what you eat” holds true wh en considering the physical, mental, and emotional impact of our diet on our overall identity. Choosing a healthy, balanced diet not only supports our physical health but also influences our mental well-being and emotional connection to food. By recognizing the power of our food choices, we can strive to nourish our bodies and minds, ultimately shaping our identity for the better.。