1. Trang chủ
  2. » Giáo án - Bài giảng

inflammatory disease caused by intestinal pathobionts

6 1 0

Đang tải... (xem toàn văn)

THÔNG TIN TÀI LIỆU

Nội dung

Available online at www.sciencedirect.com ScienceDirect Inflammatory disease caused by intestinal pathobionts Ellen L Zechner1,2 Environmental and intrinsic factors that alter microbiota structure can trigger aberrant immune responses The resulting states of dysbiosis take many forms characterized by overrepresentation of pro-inflammatory organisms and pathobionts and loss of beneficial commensals further aggravating the inflammatory state The pathogenic potential of the dysbiotic community can be linked to specific organisms in some cases but accumulating evidence suggests that intestinal inflammatory diseases are driven by collective functions of highly variable polymicrobial communities Key challenges are to gain sufficient knowledge of the structure and function of a given disease-causing consortium to understand how inflammation is perpetuated, to identify the protective mechanisms lost in the absence of specific commensals and test interventions to shift a persistent dysbiotic community to a more benign state Addresses Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/1, A-8010 Graz, Austria BioTechMed-Graz, Austria Corresponding author: Zechner, Ellen L (ellen.zechner@uni-graz.at) Current Opinion in Microbiology 2017, 35:64–69 This review comes from a themed issue on Host–microbe interactions: bacteria Edited by Samuel Miller and Rene´e Tsolis http://dx.doi.org/10.1016/j.mib.2017.01.011 1369-5274/ã 2017 The Author Published by Elsevier Ltd This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/) Introduction Enteric bacteria and their vertebrate hosts have coevolved over millions of years [1] Colonization and development of a stable microbiota within the intestinal tract is crucial for host physiology and a fully functional immune system [2,3] The intestinal epithelium is a single layer of cells that facilitates absorption of nutrients and vitamins Remarkably this thin barrier also prevents pathogen invasion and dissemination of commensals Coping with the trillions of microbes present in the gut lumen and continuous threats from newly ingested infectious and non-infectious organisms is a tremendous challenge for the mucosal immune system Achieving a healthy steady state requires mounting an effective frontline defense while enabling the host to accommodate the symbiotic Current Opinion in Microbiology 2017, 35:64–69 community Intestinal epithelial cells (IECs) take an active role in both processes through perception of microbial signals and working in concert with immunocompetent cells from the underlying lamina propria Preservation of homeostasis relies on highly efficient innate and adaptive mucosal immunity and continuous epithelial renewal Historically bacteria have been classified according to their relationship with the host: commensal or pathogen However, as the field of human microbiome research has exploded in the last decade, more differentiated terms are needed to adequately describe the host– microbe commensal relationship (for an excellent discussion see Ref [4]) Homeostasis, and the composition and activities of the symbiotic community can be challenged by host genetics [5] or environmental factors such as diet [6,7] or medication [8] The altered host–microbe relationship can be defined as dysbiosis when the microbial shift has pathophysiological consequences Pathobionts are members of the symbiotic community that expand as a result of the imbalance and exert pathogenic effects on the host Here I discuss how microbial dysbiosis contributes to the development and pathology of inflammatory diseases of the intestine, use recent advances with selected experimental models to illustrate microbial-driven pathologies and focus on the particular role of pathobionts in this dysregulated state Individualists vs community level pathogenic potential Antibiotics alter microbiota composition and functions producing both acute and long-lasting deleterious effects for the host [9] Depletion of microbial density diminishes signaling to the intestinal mucosa, impairs colonization resistance and allows expansion by the antibiotic resistant population A healthy human microbiota harbors a rich and diverse reservoir of resistance genes, which amplify dramatically during treatment [10] Moreover, antibiotic use lowers resistance to intestinal domination by bacteria associated with hospital-acquired infections [11] Overgrowth and dissemination to extraintestinal organs by highly resistant bacteria such as vancomycin-resistant Enterococci and multidrug-resistant Enterobacteriaceae create clinical problems that increasingly defy treatment [12,13] The induced dysbiosis can also result in overgrowth of toxigenic members The organisms causing antibiotic associated diarrhea remain unidentified in many cases, www.sciencedirect.com Inflammatory disease caused by intestinal pathobionts Zechner 65 yet antibiotic associated colitis is a disease model that has enabled single organisms and their specific pathophysiological effects to be causally linked to the development of inflammatory disorders Resident members such as Clostridium difficile or Klebsiella oxytoca flourish under these conditions and their ability to produce protein or small molecule enterotoxins, once high microbial densities are reached [14], is responsible for some but not all forms of antibiotic associated colitis [15,16] Although these organisms appear to exert their pathogenic properties individually, a complex interplay between pathobiont, microbial community, and host response actually determines disease outcome [17] Other inflammatory diseases are instigated not by single organisms but by the collective activities of a multispecies community Interactions between community members create synergies and determine the pathogenic potential of the consortium, such as the capacity to overgrow, produce noxious substances, sustain inflammation and support pathogens [18] Hajishengallis and Lamont [4] suggested the term nososymbiocity [nosos (Greek for disease) arising from host–microbe symbiosis] to describe the capacity of an indigenous community to cause disease in a context-dependent manner The potential for a dysbiotic microbiota to induce an immune response that is uncontrolled and destructive is a key component in inflammatory mucosal diseases Crohn’s disease (CD) and ulcerative colitis (UC) are two intestinal inflammatory conditions collectively called inflammatory bowel disease (IBD) that are caused by multiple factors involving host genetics, the environment, and microbes Hallmark shifts in microbial abundances in CD are expansion of pathobiont microbes from Bacteroidetes and Enterobacteria and a concomitant depletion of symbiont microbes including Firmicutes, Bifidobacteria and Clostridia [19] Nonetheless evidence implicating a single pathobiont has not emerged from studies of different IBD cohorts Perturbation of the host–microbe commensal relationship is widely accepted as a leading factor driving the inflammatory tissue injury Yet the microbial component of IBD has been more difficult to define primarily because the bacterial role in pathogenesis occurs indirectly, via stimulation of the immune system The emerging view based on numerous studies is that collective interactions of the community drive disease through dysregulation of mucosal immunity and disruption of the mucosal barrier The resulting loss of tolerance to antigens present in the commensal microbiota induces chronic intestinal inflammation and disease Knowledge of how immunopathological communities become established has advanced dramatically (Figure 1) Functional mechanistic insights into how the host response shapes the microbial community and the consequences of these fluctuations are also beginning to emerge The challenge remains to delineate the www.sciencedirect.com respective roles of community members to understand causalities between gut microbes and immunity Host genetic deficiencies and colonic microbial ecology Large-scale genome-wide association studies reveal IBD as complex multigenic disorders [20] The genetic variants that confer risk to IBD indicate the importance of genes involved in recognition and intracellular killing of bacteria including autophagy genes involved in bacterial clearance [21] Although genetic polymorphisms associated with IBD predispose the host, it is increasingly appreciated that both genetic background and inflammation itself impacts the microbiota The resulting shift in microbial structure and function is required for induction and perpetuation of chronic inflammation As an illustration of this, both loss and hyperactivity of inflammasomes – complexes that act as steady-state sensors of pathogenassociated molecular patterns and regulators of the colonic microbiota – are linked to dysbiosis, pathobiont expansion and disease [22–24] Lack of the inflammasomes NLRP6 and NLRP3, for example, results in dysbiosis marked by expansion of several specific taxa not found in wild type littermates [23,24] The altered gut microbiota generated by inflammasome deficiency triggered an enhanced inflammatory response in the intestine and predisposed the host to IBD Importantly, the colitogenic phenotypes were communicable to wild type mice through co-housing Moreover, genetically susceptible mice treated with antibiotics to reduce the microbiota or those born via caesarian section then raised germfree developed little or no colitis Thus, genetic alterations play a role, yet it is the associated changes in microbial composition and pathogenic potential that are believed to regulate not only the rate of progression of IBD, but also colorectal cancer and multiple metabolic syndrome-associated abnormalities [25,26] This interpretation raises the question of whether genetics predispose individuals to a less effective microbiota Role of the microbiota in the inflammatory cascade Identifying the causal molecular mechanisms that enable pathobionts to contribute to disease development has been challenging but important insights are emerging In the healthy intestine, there is normally a low oxygen level and a large obligate anaerobe population Fermentation of complex polysaccharides supports anaerobic growth of the resident community However, conditions and diseases leading to intestinal inflammation disrupt the microbiota composition severely [27,28] Inflammatory conditions are mainly associated with an overall drop in species richness and an alteration in the abundance of several taxa [29,30] Obligate anaerobes from the phyla Bacteroidetes and Firmicutes are lost and facultative anaerobes belonging to the Gammaproteobacteria increase The bloom of Enterobacteriaceae is notably Current Opinion in Microbiology 2017, 35:64–69 66 Host–microbe interactions: bacteria Figure INITIAL SYMBIOSIS DYSBIOSIS NOSOSYMBIOCITY Healthy Healthy Antibiotics Infections Intestinal inflammatory disease Inflammation Intestinal lumen Exacerbations Early determinants Initial colonization Geography Evolutionary history Host genetics Diet Intestinal lumen Pathological Pathological TIME Protective organism Commensal Pathobiont Inflammophiles Pathogen (persistent) Pathogen (transient) Current Opinion in Microbiology Variable microbiota composition and the interplay of pathobionts and dysbiotic community activities determine community pathogenic potential or ‘nososymbiocity’ Composition of an individual’s microbiota determines functional outcomes ranging from resilient homeostasis to predisposition to dysbiosis and inflammation (scale bar, left) Multiple factors influence initial development of the gut microbiota, while diet has long-term impact on community maintenance Exposure to exogenous factors such as antibiotics eliminate commensal organisms and allow expansion of the resistant population including pathobionts Suppression of this population can mediate recovery Alternatively, the altered microbial structure stabilizes with increased pro-inflammatory potential Transient exposure to pathogens may also shift community structure and function with lasting impact despite pathogen clearance Inflammation, triggered extrinsically or from host immune deficiencies, drives loss of obligate anaerobes and their protective functions and favors growth of inflammophiles and pathobionts better equipped to withstand host antimicrobial responses Progressive dysregulation, loss of barrier function, loss of tolerance, hyperimmunoactivity, and depletion of even more beneficial organisms exacerbate immunopathologic potential (scale bar, right) more pronounced in the microbiota associated with the oxygen containing environment of the mucosa, than in fecal samples [31] Chronic inflammation of the colon leads to the production of various inflammatory cytokines, free radicals and other antimicrobials Winter et al [32] used murine models with chemically-induced colitis or animals lacking the antiinflammatory cytokine IL-10 to establish a mechanistic link between elevated inducible nitric oxide synthase in the inflamed intestine, abundant nitric oxide and a competitive advantage for facultative anaerobic commensal Enterobacteriaceae Members of this community are able to utilize the highly oxidized by-products as electron acceptors for anaerobic respiration The local inflammatory response thus creates a unique nutritional environment that is conducive to a bloom of bacterial species able to consume inflammation-derived nutrients, survive oxidative stress and evade released antimicrobials [33] Current Opinion in Microbiology 2017, 35:64–69 Oxygen availability is another important component in the relationship between host and microbiota, which initiates and perpetuates dysbiosis in IBD patients [34] Albenberg et al [35] examined intraluminal oxygen levels in the colon of mice and humans and observed a radial gradient of microbes linked to distribution of oxygen and nutrients provided by host tissue Oxygen produced by the epithelium enriches for aerotolerant bacteria closest to the mucosa, while anaerobic organisms dominate the gut lumen [35] Since chronic inflammation is expected to elevate oxygen levels in the intestine through increased blood flow and immunological responses, oxygen-consuming organisms are selected and obligate anaerobic communities disrupted [34,36] Consistent with this view, comparison of the microbiome in new-onset CD patients showed an altered mucosalassociated bacterial community favoring aerotolerant species [36,37] Antibiotic therapy is another driver of dysbiosis that can alter gut oxygenation [38,39] www.sciencedirect.com Inflammatory disease caused by intestinal pathobionts Zechner 67 Bacteria-derived butyrate is a preferred energy source for IECs, which oxidize this short chain fatty acid to CO2 The epithelium becomes hypoxic as a result Antibiotic depletion of butyrate-producing Clostridia in the mouse alters colonocyte energy metabolism with a concomitant increase in tissue oxygenation [38] The shift in pO2 selects for an altered microbiota and may lead to higher diffusible oxygen in the lumen, conditions that support post-antibiotic expansion of pathogens such as S Typhimurium [39] and very likely pathobionts within the altered gut microbiota In summary these data underscore the importance of the microbial component, gut oxygenation and the inflammatory reaction itself as perpetrators of dysbiosis in IBD patients (Figure 1) The condition enriches for inflammophilic pathobionts, more irritating proteobacteria, such as adherent-invasive E coli [40,41] and perhaps other pathogens better equipped to compete in the inflamed gut This more aggressive community may instigate chronic inflammation in susceptible hosts by shedding pro-inflammatory effectors and activating innate immunity, which worsens inflammation and interferes with its resolution Participation of these organisms in the inflammation process exacerbates the destructive spiral characteristic of IBD Microbial pathogenesis in colitis-associated colorectal cancer Colorectal cancer (CRC) has a multifactorial etiology that includes microbiota-mediated effects [42–44] Chronic inflammation impacts gut microbial composition and metabolic activities Moreover it is becoming increasingly clear that the microbiota associated with inflammation has a direct role in tumorigenesis Community-wide effects involving the gain and loss of bacterial populations and general metabolic functions have been shown Zackular et al [45] compared tumor formation in germ-free mice colonized either with microbiota from tumor-bearing mice or conventional animals They found that the gut microbiota associated with inflammation and CRC directly contributed to tumorigenesis and that antibiotic treatment reduced both the number and size of tumors Pro-carcinogenic activities for specific bacteria associated with colorectal adenomas and IBD are also becoming clear Human colonic isolates of Fusobacterium nucleatum accelerated intestinal tumorigenesis when introduced to genetically susceptible mice [46,47] Certain strains of Bacteroides fragilis and E coli promote CRC in murine models through the production of tumorigenic factors Up to 30% of E coli strains of the B2 phylotype harbor a polyketide synthase ( pks) gene cluster for synthesizing the DNA crosslinking metabolite colibactin [48,49] Arthur et al [50] used germ-free IL-10À/À mice with the colon-specific carcinogen azoxymethane as a model for colitis-associated CRC and linked carriage of the pks gene cluster by a commensal E coli strain to higher www.sciencedirect.com incidence of invasive adenocarcinoma Use of boronbased compounds to block colibactin production controlled tumorigenic effects of pks+ E coli in preclinical models [51] Early gut colonization by pks+ E coli contributes to defective intestinal homeostasis consistent with their frequent association with the mucosa of IBD and CRC patients [52] All these data fuel concerns over the increasing prevalence of B2 group E coli strains colonizing infants [53] Enterotoxigenic B fragilis (ETBF) colonize humans of all ages [54], yet ETBF also causes acute diarrhea and is associated with active IBD and CRC The metalloprotease toxin secreted by ETBF cleaves cadherin family proteins leading to disruption of tight junctions and increased barrier permeability Chronic colonization with ETBF in humans [55] may result in persistent subclinical but potentially tumor promoting colitis [56] E-cadherin cleavage, Wnt signaling and secretion of proinflammatory cytokines contribute further to the toxin’s carcinogenic potential [42] Intriguingly, Hecht et al [57] recently observed that symbiotic non-toxigenic B fragilis could limit acquisition of pathogenic ETBF and prevent disease in a murine host The mechanism of interbacterial competition relies on type VI secretion, thus highlighting the importance of antagonistic factors for microbiota composition and stability Better understanding of bacterial competition may support probiotic interventions to restrict pathobionts or even modulate dysbiotic communities that have reached a metastable state Conclusions Although this Commentary has focused on the role of pathobionts and the positive feedback loop that exacerbates the inflammatory state, an important property of dysbiosis that compounds the problem is the loss of protective microorganisms Appreciation of interdependencies linking beneficial bacteria and the immune system is rising, thus mechanistic understanding of the protective role of anti-inflammatory bacteria such as Faecalibacterium prausnitzii, absent in patients with inflammatory disease [58], will be equally important In a promising approach Miquel and coworkers [59] established a stable dual-association of E coli and the extremely oxygen-sensitive F prausnitzii in a gnotobiotic murine model Variations in the gut metabolomes associated with the presence of F prausnitzii during acute colitis identified a specific metabolic signature linked to anti-inflammatory effects Metabolites monitored with this approach can be of host or bacterial origin, thus through studies of this kind we may begin to understand reciprocal functional adaptation occurring between host and microbes Acknowledgements Work in the author’s laboratory is supported by the Austrian Science Fund (FWF) W901 DK Molecular Enzymology and BioTechMed-Graz, flagship Current Opinion in Microbiology 2017, 35:64–69 68 Host–microbe interactions: bacteria project Secretome S Roier is gratefully acknowledged for providing the graphics and helpful discussion 14 Darkoh C, DuPont HL, Norris SJ, Kaplan HB: Toxin synthesis by Clostridium difficile is regulated through quorum signaling MBio 2015, 6:e02569 References and recommended reading 15 Kuehne SA, Cartman ST, Heap JT, Kelly ML, Cockayne A, Minton NP: The role of toxin A and toxin B in Clostridium difficile infection Nature 2010, 467:711-713 Papers of particular interest, published within the period of review, have been highlighted as:  of special interest  of outstanding interest Moeller AH, Caro-Quintero A, Mjungu D, Georgiev AV, Lonsdorf EV, Muller MN, Pusey AE, Peeters M, Hahn BH, Ochman H: Cospeciation of gut microbiota with hominids Science 2016, 353:380-382 Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T et al.: Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells Nature 2013, 504:446-450 Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR et al.: Gut immune maturation depends on colonization with a host-specific microbiota Cell 2012, 149:1578-1593  Hajishengallis G, Lamont RJ: Dancing with the stars: how choreographed bacterial interactions dictate nososymbiocity and give rise to keystone pathogens, accessory pathogens, and pathobionts Trends Microbiol 2016, 24:477-489 In this excellent review Hajishengallis and Lamont develop the concept that commensal and pathogenic properties of organisms are not intrinsic features but need to be perceived in the context of a given microbial community and the host immune status 16 Schneditz G, Rentner J, Roier S, Pletz J, Herzog KA, Bucker R, Troeger H, Schild S, Weber H, Breinbauer R et al.: Enterotoxicity of a nonribosomal peptide causes antibiotic-associated colitis Proc Natl Acad Sci U S A 2014, 111:13181-13186 17 El Feghaly RE, Stauber JL, Deych E, Gonzalez C, Tarr PI, Haslam DB: Markers of intestinal inflammation, not bacterial burden, correlate with clinical outcomes in Clostridium difficile infection Clin Infect Dis 2013, 56:1713-1721 18 Short FL, Murdoch SL, Ryan RP: Polybacterial human disease: the ills of social networking Trends Microbiol 2016, 22:508-516 19 Walters WA, Xu Z, Knight R: Meta-analyses of human gut microbes associated with obesity and IBD FEBS Lett 2014, 588:4223-4233 20 Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, Lee JC, Schumm LP, Sharma Y, Anderson CA et al.: Host– microbe interactions have shaped the genetic architecture of inflammatory bowel disease Nature 2012, 491:119-124 21 Butto LF, Schaubeck M, Haller D: Mechanisms of microbe–host interaction in Crohn’s disease: dysbiosis vs pathobiont selection Front Immunol 2015, 6:555 22 Ayres JS, Trinidad NJ, Vance RE: Lethal inflammasome activation by a multidrug-resistant pathobiont upon antibiotic disruption of the microbiota Nat Med 2012, 18:799-806 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, 159:789799 23 Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI et al.: NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis Cell 2011, 145:745-757 Wang J, Linnenbrink M, Kunzel S, Fernandes R, Nadeau MJ, Rosenstiel P, Baines JF: Dietary history contributes to enterotype-like clustering and functional metagenomic content in the intestinal microbiome of wild mice Proc Natl Acad Sci U S A 2014, 111:E2703-2710 24 Hirota SA, Ng J, Lueng A, Khajah M, Parhar K, Li Y, Lam V, Potentier MS, Ng K, Bawa M et al.: NLRP3 inflammasome plays a key role in the regulation of intestinal homeostasis Inflamm Bowel Dis 2011, 17:1359-1372  David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA et al.: Diet rapidly and reproducibly alters the human gut microbiome Nature 2014, 505:559-563 These authors analyze the impact of short-term variation in macronutrients on the human gut microbiome underscoring the view that diet is one of the most potent driving forces shaping gut microbial communities Forslund K, Sunagawa S, Kultima JR, Mende DR, Arumugam M, Typas A, Bork P: Country-specific antibiotic use practices impact the human gut resistome Genome Res 2013, 23:1163-1169 Jernberg C, Lofmark S, Edlund C, Jansson JK: Long-term ecological impacts of antibiotic administration on the human intestinal microbiota ISME J 2007, 1:56-66 10 Sommer MO, Dantas G, Church GM: Functional characterization of the antibiotic resistance reservoir in the human microflora Science 2009, 325:1128-1131 25 Hu B, Elinav E, Huber S, Strowig T, Hao L, Hafemann A, Jin C, Wunderlich C, Wunderlich T, Eisenbarth SC et al.: Microbiotainduced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer Proc Natl Acad Sci U S A 2013, 110:9862-9867 26 Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, Thaiss CA, Kau AL, Eisenbarth SC, Jurczak MJ et al.: Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity Nature 2012, 482:179-185 27 Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR: Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases Proc Natl Acad Sci U S A 2007, 104:13780-13785 28 Lupp C, Robertson ML, Wickham ME, Sekirov I, Champion OL, Gaynor EC, Finlay BB: Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae Cell Host Microbe 2007, 2:204 11 Swaminathan M, Sharma S, Poliansky Blash S, Patel G, Banach DB, Phillips M, LaBombardi V, Anderson KF, Kitchel B, Srinivasan A et al.: Prevalence and risk factors for acquisition of carbapenem-resistant Enterobacteriaceae in the setting of endemicity Infect Control Hosp Epidemiol 2013, 34:809-817 29 Papa E, Docktor M, Smillie C, Weber S, Preheim SP, Gevers D, Giannoukos G, Ciulla D, Tabbaa D, Ingram J et al.: Non-invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease PLoS One 2012, 7:e39242 12 Becattini S, Taur Y, Pamer EG: Antibiotic-induced changes in the intestinal microbiota and disease Trends Mol Med 2016, 22:458-478 30 Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, LeLeiko N, Snapper SB et al.: Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment Genome Biol 2012, 13:R79 13 Ubeda C, Taur Y, Jenq RR, Equinda MJ, Son T, Samstein M, Viale A, Socci ND, van den Brink MR, Kamboj M et al.: Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans J Clin Investig 2010, 120:4332-4341 Current Opinion in Microbiology 2017, 35:64–69 31 Chassaing B, Darfeuille-Michaud A: The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases Gastroenterology 2011, 140:1720-1728 32 Winter SE, Winter MG, Xavier MN, Thiennimitr P, Poon V, Keestra AM, Laughlin RC, Gomez G, Wu J, Lawhon SD et al.: www.sciencedirect.com Inflammatory disease caused by intestinal pathobionts Zechner 69 Host-derived nitrate boosts growth of E coli in the inflamed gut Science 2013, 339:708-711 33 Winter SE, Lopez CA, Baăumler AJ: The dynamics of gutassociated microbial communities during inflammation EMBO Rep 2013, 14:319-327 34 Rigottier-Gois L: Dysbiosis in inflammatory bowel diseases: the oxygen hypothesis ISME J 2013, 7:1256-1261 35 Albenberg L, Esipova TV, Judge CP, Bittinger K, Chen J,  Laughlin A, Grunberg S, Baldassano RN, Lewis JD, Li H et al.: Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota Gastroenterology 2014, 147:1055-1063 e1058 The authors used phosphorescence quenching and a specially designed intraluminal oxygen probe to dynamically quantify gut luminal oxygen levels in mice Combined with 16S ribosomal RNA gene sequencing to characterize the microbiota the study revealed a radial gradient of microbes linked to the distribution of oxygen and nutrients provided by host tissue 36 Haberman Y, Tickle TL, Dexheimer PJ, Kim MO, Tang D, Karns R, Baldassano RN, Noe JD, Rosh J, Markowitz J et al.: Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature J Clin Investig 2014, 124:3617-3633 37 Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, Schwager E, Knights D, Song SJ, Yassour M et al.: The treatment-naive microbiome in new-onset Crohn’s disease Cell Host Microbe 2014, 15:382-392 38 Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A et al.: Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function Cell Host Microbe 2015, 17:662-671 39 Rivera-Chavez F, Zhang LF, Faber F, Lopez CA, Byndloss MX,  Olsan EE, Xu G, Velazquez EM, Lebrilla CB, Winter SE et al.: Depletion of butyrate-producing Clostridia from the gut microbiota drives an aerobic luminal expansion of Salmonella Cell Host Microbe 2016, 19:443-454 This work describes a remarkable bacterial strategy to control epithelial oxygenation.S Typhimurium uses its virulence arsenal for selective antagonism of Clostridia The resulting switch in colonocyte energy metabolism to glucose fermentation raises epithelial oxygenation, which drives aerobic luminal expansion and enhanced pathogen transmission 40 Chassaing B, Koren O, Carvalho FA, Ley RE, Gewirtz AT: AIEC  pathobiont instigates chronic colitis in susceptible hosts by altering microbiota composition Gut 2014, 63:1069-1080 This study shows that, remarkably, even transient colonization by the pathobiont AIEC during microbiota acquisition in mice lacking the flagellin receptor, TLR5, altered the gut microbiota composition so as to give it an inherently greater ability to activate innate immunity/pro-inflammatory gene expression and resulted in chronic colitis 41 Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, Barnich N, Bringer MA, Swidsinski A, Beaugerie L, Colombel JF: High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease Gastroenterology 2004, 127:412-421 42 Sears CL, Garrett WS: Microbes, microbiota, and colon cancer Cell Host Microbe 2014, 15:317-328 43 Kostic AD, Chun E, Meyerson M, Garrett WS: Microbes and inflammation in colorectal cancer Cancer Immunol Res 2013, 1:150-157 44 Irrazabal T, Belcheva A, Girardin SE, Martin A, Philpott DJ: The multifaceted role of the intestinal microbiota in colon cancer Mol Cell 2014, 54:309-320 45 Zackular JP, Baxter NT, Iverson KD, Sadler WD, Petrosino JF, Chen GY, Schloss PD: The gut microbiome modulates colon tumorigenesis MBio 2013, 4:e00692-00613 46 Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL et al.: Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment Cell Host Microbe 2013, 14:207-215 www.sciencedirect.com 47 Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW: Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin Cell Host Microbe 2013, 14:195-206 48 Vizcaino MI, Crawford JM: The colibactin warhead crosslinks  DNA Nat Chem 2015, 7:411-417 Structural analyses of colibactin have been hindered due to high instability These authors employ a combination of NMR spectroscopy and bioinformatics-guided isotopic labeling studies to characterize the colibactin substituted spirobicyclic ‘warhead’ structure This structure crosslinks duplex DNA in vitro, providing direct experimental evidence for colibactin’s DNA-damaging activity 49 Nougayrede JP, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, Buchrieser C, Hacker J, Dobrindt U, Oswald E: Escherichia coli induces DNA double-strand breaks in eukaryotic cells Science 2006, 313:848-851 50 Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB et al.: Intestinal inflammation targets cancer-inducing activity of the microbiota Science 2012, 338:120-123 51 Cougnoux A, Delmas J, Gibold L, Fais T, Romagnoli C, Robin F,  Cuevas-Ramos G, Oswald E, Darfeuille-Michaud A, Prati F et al.: Small-molecule inhibitors prevent the genotoxic and protumoural effects induced by colibactin-producing bacteria Gut 2016, 65:278-285 This study shows that development of small molecule inhibitors of colibactin biosynthesis can control the tumorigenic capacity of pks+ bacteria in a mouse model of CRC 52 Payros D, Secher T, Boury M, Brehin C, Menard S, SalvadorCartier C, Cuevas-Ramos G, Watrin C, Marcq I, Nougayrede JP et al.: Maternally acquired genotoxic Escherichia coli alters offspring’s intestinal homeostasis Gut Microbes 2014, 5:313-325 53 Secher T, Brehin C, Oswald E: Early settlers: which E coli strains you not want at birth? Am J Physiol Gastrointest Liver Physiol 2016, 311:G123-129 54 Sears CL: Enterotoxigenic Bacteroides fragilis: a rogue among symbiotes Clin Microbiol Rev 2009, 22:349-369 Table of Contents 55 Boleij A, Hechenbleikner EM, Goodwin AC, Badani R, Stein EM, Lazarev MG, Ellis B, Carroll KC, Albesiano E, Wick EC et al.: The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients Clin Infect Dis 2015, 60:208-215 56 Rhee KJ, Wu S, Wu X, Huso DL, Karim B, Franco AA, Rabizadeh S, Golub JE, Mathews LE, Shin J et al.: Induction of persistent colitis by a human commensal, enterotoxigenic Bacteroides fragilis, in wild-type C57BL/6 mice Infect Immun 2009, 77:1708-1718 57 Hecht AL, Casterline BW, Earley ZM, Goo YA, Goodlett DR,  Bubeck Wardenburg J: Strain competition restricts colonization of an enteric pathogen and prevents colitis EMBO Rep 2016, 17:1281-1291 These authors report that colonization of a murine host with symbiotic non-toxigenic Bacteroides fragilis can limit acquisition of pathogenic enterotoxigenic B fragilis and prevent intestinal inflammatory disease 58 Sokol H, Pigneur B, Watterlot L, Lakhdari O, BermudezHumaran LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G et al.: Faecalibacterium prausnitzii is an antiinflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients Proc Natl Acad Sci U S A 2008, 105:16731-16736 59 Miquel S, Leclerc M, Martin R, Chain F, Lenoir M, Raguideau S,  Hudault S, Bridonneau C, Northen T, Bowen B et al.: Identification of metabolic signatures linked to anti-inflammatory effects of Faecalibacterium prausnitzii MBio 2015, These authors gain insights to mechanisms underlying beneficial effects using a novel gnotobiotic diassociation rodent model in which E coli primed the gastrointestinal tract allowing a stable establishment of the extreme oxygen-sensitive F prausnitzii Metabolic profiling along the GI tract and in the peripheral blood identified signature metabolites linked to anti-inflammatory effects Current Opinion in Microbiology 2017, 35:64–69 ... in inflammatory mucosal diseases Crohn’s disease (CD) and ulcerative colitis (UC) are two intestinal inflammatory conditions collectively called inflammatory bowel disease (IBD) that are caused. . .Inflammatory disease caused by intestinal pathobionts Zechner 65 yet antibiotic associated colitis is a disease model that has enabled single organisms... of dysbiosis that can alter gut oxygenation [38,39] www.sciencedirect.com Inflammatory disease caused by intestinal pathobionts Zechner 67 Bacteria-derived butyrate is a preferred energy source

Ngày đăng: 04/12/2022, 14:54

TÀI LIỆU CÙNG NGƯỜI DÙNG

TÀI LIỆU LIÊN QUAN