Therapeutic Potential of the Gut Microbiota in the Prevention and Treatment of Sepsis

Bastiaan W Haak, Hallie C Prescott, W Joost Wiersinga, Bastiaan W Haak, Hallie C Prescott, W Joost Wiersinga

Abstract

Alongside advances in understanding the pathophysiology of sepsis, there have been tremendous strides in understanding the pervasive role of the gut microbiota in systemic host resistance. In pre-clinical models, a diverse and balanced gut microbiota enhances host immunity to both enteric and systemic pathogens. Disturbance of this balance increases susceptibility to sepsis and sepsis-related organ dysfunction, while restoration of the gut microbiome is protective. Patients with sepsis have a profoundly distorted composition of the intestinal microbiota, but the impact and therapeutic potential of the microbiome is not well-established in human sepsis. Modulation of the microbiota consists of either resupplying the pool of beneficial microbes by administration of probiotics, improving the intestinal microenvironment to enhance the growth of beneficial species by dietary interventions and prebiotics, or by totally recolonizing the gut with a fecal microbiota transplantation (FMT). We propose that there are three potential opportunities to utilize these treatment modalities over the course of sepsis: to decrease sepsis incidence, to improve sepsis outcome, and to decrease late mortality after sepsis. Exploring these three avenues will provide insight into how disturbances of the microbiota can predispose to, or even perpetuate the dysregulated immune response associated with this syndrome, which in turn could be associated with improved sepsis management.

Keywords: fecal microbiota transplantation; microbiota; pathogenesis; probiotics and synbiotics; sepsis; therapeutics.

Figures

Figure 1
Figure 1
Overview of systemic immunomodulatory mechanisms associated with the microbiota. Structural components of gut microbiota, otherwise known as microbe-associated molecular patterns (MAMPs), can elicit a systemic pro-inflammatory response by activating pattern recognition receptors of both the innate and the adaptive immune system. Microbial metabolites, such as short chain fatty acids (SCFAs) modulate epigenetic changes in host leukocytes, which can induce both pro- and anti-inflammatory responses. The presence of the SCFAs butyrate and propionate drives the generation of regulatory T cells (Treg), which dampen inflammation. In addition, the gut metabolite desaminotyrosine enhances clearance of respiratory viruses by inducing type 1 interferon (IFN) responses. Direct interactions with epithelial cells by segmented filamentous bacteria (SFB) can enhance mucosal immunity by upregulating T helper 17 (Th 17) cells in both in the gut and in the lung. It is important to realize that our knowledge on microbiota-derived host-resistance is fragmented and it remains to be determined how these individual mechanisms fit in an overarching framework of systemic immunity. DC, dendritic cell; ILC3, type 3 innate lymphoid cell; Treg cell, regulatory T cell; IgA, Immunoglobulin A; IgG, Immunoglobulin G; IgM, immunoglobulin M; LPS, lipopolysaccharide; LTA, lipoteichoic acid; MLP, murein lipoprotein.
Figure 2
Figure 2
Timeline of potential microbiota-associated interventions prior to, during and post-sepsis. (A) Hypothetical diagram of the associations between the risk of dysbiosis and sepsis development and outcome. Dysbiosis occurs due to the administration of antibiotics and/or hospitalizations, recover quickly upon hospital discharge or cessation of antibiotic treatment, but it often takes long for the microbiota to recover entirely. These periods of dysbiosis predispose to the development of opportunistic infections, which in turn further worsens the state of dysbiosis and predispose to sepsis development. During sepsis, the microbiota composition is extremely hampered, which has been associated with an increased risk of secondary infections, immunosuppression and potentially even organ dysfunction. Recolonization of a homeostatic microbiota is slow upon hospital discharge and sepsis recovery, which might contribute to prolonged immunosuppression, rehospitalization due to infections and increased mortality. (B) Probiotics and synbiotics have the potential to enhance microbiota colonization in neonates, as well as accelerate the recovery of the microbiota after periods of dysbiosis, which in turn could provide protection against the development of sepsis. During the course of sepsis, microbiota disruption could be ameliorated by the administration of pro- and synbiotics, which potentially reduces the occurrence of secondary infections. In addition, future treatment with fecal microbiota transplantation (FMT) or targeted restoration with microbiota-associated metabolites, such as short-chain fatty acids (SCFAs), could reduce the risk of prolonged immunosuppression and organ dysfunction. Finally, microbiota restoration after sepsis recovery could be accelerated with pro- and synbiotics, or by an autologous FMT. *Time depiction is schematic.

References

    1. Singer M, Deutschman CS, Seymour C, Shankar-Hari M, Annane D, Bauer M, et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA (2016) 315:801–10. 10.1001/jama.2016.0287
    1. van der Poll T, van de Veerdonk FL, Scicluna BP, Netea MG. The immunopathology of sepsis and potential therapeutic targets. Nat Rev Immunol. (2017) 17:407–20. 10.1038/nri.2017.36
    1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. (2013) 13:862–74. 10.1038/nri3552
    1. Gotts JE, Matthay MA. Sepsis: pathophysiology and clinical management. BMJ (2016) 353:i1585. 10.1136/bmj.i1585
    1. Arefian H, Heublein S, Scherag A, Brunkhorst FM, Younis MZ, Moerer O, et al. . Hospital-related cost of sepsis: a systematic review. J Infect. (2017) 74:107–17. 10.1016/j.jinf.2016.11.006
    1. Fleischmann C, Scherag A, Adhikari NK, Hartog CS, Tsaganos T, Schlattmann P, et al. . Assessment of global incidence and mortality of hospital-treated sepsis. Current estimates and limitations. Am J Respir Crit Care Med. (2016) 193:259–72. 10.1164/rccm.201504-0781OC
    1. Prescott HC, Dickson RP, Rogers MA, Langa KM, Iwashyna TJ. Hospitalization type and subsequent severe sepsis. Am J Respir Crit Care Med. (2015) 192:581–88. 10.1164/rccm.201503-0483OC
    1. Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. (2017) 2:135–43. 10.1016/S2468-1253(16)30119-4
    1. Ojima M, Motooka D, Shimizu K, Gotoh K, Shintani A, Yoshiya K, et al. . Metagenomic analysis reveals dynamic changes of whole gut microbiota in the acute phase of intensive care unit patients. Dig Dis Sci. (2016) 61:1628–34. 10.1007/s10620-015-4011-3
    1. Zaborin A, Smith D, Garfield K, Quensen J, Shakhsheer B, Kade M, et al. . Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio (2014) 5:e01361–14. 10.1128/mBio.01361-14
    1. McDonald D, Ackerman G, Khailova L, Baird C, Heyland D, Kozar R, et al. . Extreme dysbiosis of the microbiome in critical illness. mSphere (2016) 1:e00199-16. 10.1128/mSphere.00199-16
    1. Lankelma JM, van Vught LA, Belzer C, Schultz MJ, van der Poll T, de Vos WM, et al. Critically ill patients demonstrate large interpersonal variation in intestinal microbiota dysregulation: a pilot study. Intensive Care Med. (2017) 43:59–68. 10.1007/s00134-016-4613-z
    1. Sender R, Fuchs S, Milo R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell (2016) 164:337–40. 10.1016/j.cell.2016.01.013
    1. Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. (2017) 8:172–84. 10.1080/19490976.2017.1290756
    1. Li X, Watanabe K, Kimura I. Gut microbiota dysbiosis drives and implies novel therapeutic strategies for diabetes mellitus and related metabolic diseases. Front Immunol. (2017) 8:1882. 10.3389/fimmu.2017.01882
    1. Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, et al. . Gut immune maturation depends on colonization with a host-specific microbiota. Cell (2012)149:1578–93. 10.1016/j.cell.2012.04.037
    1. Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, Schmidt TM, et al. . Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis. (2008) 197:435–8. 10.1086/525047
    1. Harris VC, Haak BW, Boele van Hensbroek M, Wiersinga WJ. The intestinal microbiome in infectious diseases: the clinical relevance of a rapidly emerging field. Open Forum Infect Dis. (2017) 4:ofx144. 10.1093/ofid/ofx144
    1. Kim S, Covington A, Pamer EG. The intestinal microbiota: antibiotics, colonization resistance, and enteric pathogens. Immunol Rev. (2017) 279:90–105. 10.1111/imr.12563
    1. Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell (2016) 167:1125–36. 10.1016/j.cell.2016.10.020
    1. Lankelma JM, Belzer C, Hoogendijk AJ, de Vos AF, de Vos WM, van der Poll T, et al. . Antibiotic-induced gut microbiota disruption decreases TNF-α release by mononuclear cells in healthy adults. Clin Transl Gastroenterol. (2016) 7:e186. 10.1038/ctg.2016.43
    1. Lankelma JM, Cranendonk DR, Belzer C, de Vos AF, de Vos WM, van der Poll T, et al. . Antibiotic-induced gut microbiota disruption during human endotoxemia: a randomised controlled study. Antibiotic-induced gut microbiota disruption during human endotoxemia: a randomised controlled study. Gut (2017) 66:1623–30. 10.1136/gutjnl-2016-312132
    1. Schuijt TJ, van der Poll T, de Vos WM, Wiersinga WJ. The intestinal microbiota and host immune interactions in the critically ill. Trends Microbiol. (2013) 21:221–29. 10.1016/j.tim.2013.02.001
    1. Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, et al. . Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. (2017) 15:55–63. 10.1038/nrmicro.2016.142
    1. Gray J, Oehrle K, Worthen G, Alenghat T, Whitsett J, Deshmukh H. Intestinal commensal bacteria mediate lung mucosal immunity and promote resistance of newborn mice to infection. Sci Transl Med. (2017) 9:aaf9412. 10.1126/scitranslmed.aaf9412
    1. Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nature Med. (2010) 16:228–31. 10.1038/nm.2087
    1. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJ, de Boer JD, et al. . The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut (2016) 65:575–83. 10.1136/gutjnl-2015-309728
    1. Lankelma JM, Birnie E, Weehuizen TAF, Scicluna BP, Belzer C, Houtkooper RH, et al. . The gut microbiota as a modulator of innate immunity during melioidosis. PLoS Negl Trop Dis. (2017) 11:e0005548. 10.1371/journal.pntd.0005548
    1. Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, et al. . Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity (2012) 37:158–70. 10.1016/j.immuni.2012.04.011
    1. McAleer JP, Nguyen NLH, Chen K, Kumar P, Ricks DM, Binnie M, et al. Pulmonary Th17 anti-fungal immunity is regulated by the gut microbiome. J Immunol. (2016) 197:97–107. 10.4049/jimmunol.1502566
    1. Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, et al. . Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci USA. (2011) 108:5354–9. 10.1073/pnas.1019378108
    1. Rosshart SP, Vassallo BG, Angeletti D, Hutchinson DS, Morgan AP, Takeda K, et al. . Wild mouse gut microbiota promotes host fitness and improves disease resistance. Cell (2017) 171:1015–28.e13. 10.1016/j.cell.2017.09.016
    1. Galvão I, Tavares LP, Corrêa RO, Fachi JL, Rocha VM, Rungue M, et al. . The metabolic sensor GPR43 receptor plays a role in the control of Klebsiella pneumoniae infection in the lung. Front Immunol. (2018) 9:142. 10.3389/fimmu.2018.00142
    1. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. . Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature (2013) 504:451–5. 10.1038/nature12726
    1. Chakraborty K, Raundhal M, Chen BB, Morse C1, Tyurina YY, Khare A, et al. . The mito-DAMP cardiolipin blocks IL-10 production causing persistent inflammation during bacterial pneumonia. Nat Commun. (2017) 8:13944. 10.1038/ncomms13944
    1. Haak BW, Littmann ER, Chaubard J-L, Pickard AJ, Fontana E, Adhi F, et al. . Impact of gut colonization with butyrate producing microbiota on respiratory viral infection following Allo-HCT. Blood (2018) 131:2978–86. 10.1182/blood-2018-01-828996
    1. Steed AL, Christophi GP, Kaiko GE, Sun L, Goodwin VM, Jain U, et al. . The microbial metabolite desaminotyrosine protects from influenza through type I interferon. Science (2017) 357:498–502. 10.1126/science.aam5336
    1. Deshmukh HS, Liu Y, Menkiti OR, Mei J, Dai N, O'Leary CE, et al. . The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat Med. (2014) 20:524–30. 10.1038/nm.3542
    1. Zeng MY, Cisalpino D, Varadarajan S, Hellman J, Warren HS, Cascalho M, et al. . Gut microbiota-induced immunoglobulin G controls systemic infection by symbiotic bacteria and pathogens. Immunity (2016) 44:647–58. 10.1016/j.immuni.2016.02.006
    1. Proietti M, Cornacchione V, Rezzonico Jost T, Romagnani A, Faliti CE, Perruzza L, et al. . ATP-gated ionotropic P2X7 receptor controls follicular T helper cell numbers in Peyer's patches to promote host-microbiota mutualism. Immunity (2014) 41:789–801. 10.1016/j.immuni.2014.10.010
    1. Wilmore JR, Gaudette BT, Gomez Atria D, Hashemi T1, Jones DD1, Gardner CA, et al. . Commensal microbes induce serum IgA responses that protect against polymicrobial sepsis. Cell Host Microbe. (2018) 23:302–11. 10.1016/j.chom.2018.01.005
    1. Dickson RP. The microbiome and critical illness. Lancet Respir Med. (2016) 4:59–72. 10.1016/S2213-2600(15)00427-0
    1. Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, et al. . International study of the prevalence and outcomes of infection in intensive care units. JAMA (2009) 302:2323–9. 10.1001/jama.2009.1754
    1. Benus RF, Harmsen HJ, Welling GW, Spanjersberg R, Zijlstra JG, Degener JE, et al. . Impact of digestive and oropharyngeal decontamination on the intestinal microbiota in ICU patients. Intensive Care Med. (2010) 36:1394–402. 10.1007/s00134-010-1826-4
    1. Haak BW, Levi M, Wiersinga WJ. Microbiota-targeted therapies on the intensive care unit. Curr Opin Crit Care. (2017) 23:167–74. 10.1097/MCC.0000000000000389
    1. Donskey CJ. The role of the intestinal tract as a reservoir and source for transmission of nosocomial pathogens. Clin Infect Dis. (2004) 39:219–26. 10.1086/422002
    1. Huber-Lang M, Lambris JD, Ward PA. Innate immune responses to trauma. Nat Immunol. (2018) 18:327–41. 10.1038/s41590-018-0064-8
    1. Krezalek MA, DeFazio J, Zaborina O, Zaborin A, Alverdy JC. The shift of an intestinal “Microbiome” to a “Pathobiome” governs the course and outcome of sepsis following surgical injury. Shock (2016) 45:475–82. 10.1097/SHK.0000000000000534
    1. Klingensmith NJ, Coopersmith CM. The gut as the motor of multiple organ dysfunction in critical illness. Crit Care Clin. (2016) 32:203–12. 10.1016/j.ccc.2015.11.004
    1. Jang HR, Ko GJ, Wasowska BA, Rabb H. The interaction between ischemia-reperfusion and immune responses in the kidney. J Mol Med. (2009) 87:859–64. 10.1007/s00109-009-0491-y
    1. Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A, Felizardo RJ, de Almeida DC, et al. . Gut bacteria products prevent AKI induced by ischemia-reperfusion. J Am Soc Nephrol. (2015) 26:1877–88. 10.1681/ASN.2014030288
    1. Schieber AM, Lee YM, Chang MW, Leblanc M, Collins B, Downes M, et al. . Disease tolerance mediated by microbiome E. coli involves inflammasome and IGF-1 signaling. Science (2015) 350:558–63. 10.1126/science.aac6468
    1. Deitch EA. Gut-origin sepsis: evolution of a concept. Surgeon (2012) 10:350–6. 10.1016/j.surge.2012.03.003
    1. Carrico CJ, Meakins JL, Marshall JC, Fry D, Maier RV. Multiple-organ-failure syndrome. Arch Surg. (1986) 121:196–208. 10.1001/archsurg.1986.01400020082010
    1. Dickson RP, Singer BH, Newstead MW, Falkowski NR, Erb-Downward JR, Standiford TJ, et al. . Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. (2016) 1:16113. 10.1038/nmicrobiol.2016.113
    1. Singer BH, Dickson RP, Denstaedt SJ, Newstead MW, Kim K, Falkowski NR, et al. . Bacterial dissemination to the brain in sepsis. Am J Respir Crit Care Med. (2018) 197:747–56. 10.1164/rccm.201708-1559OC
    1. Sundman MH, Chen NK, Subbian V, Chou YH. The bidirectional gut-brain-microbiota axis as a potential nexus between traumatic brain injury, inflammation, and disease. Brain Behav Immun. (2017) 66:31–44. 10.1016/j.bbi.2017.05.009
    1. Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, et al. . Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. (2012) 55:905–14. 10.1093/cid/cis580
    1. Baggs J, Jernigan JA, Halpin AL, Epstein L, Hatfield KM, McDonald LC. Risk of subsequent sepsis within 90 days after a hospital stay by type of antibiotic exposure. Clin Infect Dis. (2018) 66:1004–12. 10.1093/cid/cix947
    1. Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Critical Care (2016) 20:262 10.1186/s13054-016-1434-y
    1. Kasatpibal N, Whitney JD, Saokaew S, Kengkla K, Heitkemper MM, Apisarnthanarak A. Effectiveness of probiotic, prebiotic, and synbiotic therapies in reducing postoperative complications: a systematic review and network meta-analysis. Clin Infect Dis. (2017) 64:S153–60. 10.1093/cid/cix114
    1. Panigrahi P, Parida S, Nanda NC, Satpathy R, Pradhan L, Chandel DS, et al. . A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature (2017) 548:407–12. 10.1038/nature23480
    1. Jacobs SE, Tobin JM, Opie GF, Donath S, Tabrizi SN, Pirotta M, et al. . Probiotic effects on late-onset sepsis in very preterm infants: a randomized controlled trial. Pediatrics (2013) 132:1055–62. 10.1542/peds.2013-1339
    1. Costeloe K, Hardy P, Juszczak E, Wilks M, Millar MR. Bifidobacterium breve BBG-001 in very preterm infants: a randomised controlled phase 3 trial. Lancet (2016) 387:649–60. 10.1016/S0140-6736(15)01027-2
    1. Morrow LE, Wischmeyer P. Blurred lines: dysbiosis and probiotics in the intensive care unit. Chest (2016) 151:492–9. 10.1016/j.chest.2016.10.006.35
    1. Bongaerts GP, Severijnen RS. A reassessment of the PROPATRIA study and its implications for probiotic therapy. Nat Biotechnol. (2016) 34:55–63. 10.1038/nbt.3436
    1. Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. . Successful treatment of severe sepsis and diarrhea after vagotomy utilizing fecal microbiota transplantation: a case report. Crit Care (2015) 19:37. 10.1186/s13054-015-0738-7
    1. Li Q, Wang C, Tang C, He Q, Zhao X, Li N, et al. . Therapeutic modulation and reestablishment of the intestinal microbiota with fecal microbiota transplantation resolves sepsis and diarrhea in a patient. Am J Gastroenterol. (2014) 109:1832–4. 10.1038/ajg.2014.299
    1. Wei Y, Yang J, Wang J, Yang Y, Huang J, Gong H, et al. . Successful treatment with fecal microbiota transplantation in patients with multiple organ dysfunction syndrome and diarrhea following severe sepsis. Crit Care (2016) 20:332. 10.1186/s13054-016-1491-2
    1. Dessein R, Gironella M, Vignal C, Peyrin-Biroulet L, Sokol H, Secher T, et al. . Toll-like receptor 2 is critical for induction of Reg3 beta expression and intestinal clearance of Yersinia pseudotuberculosis. Gut (2009) 58:771–6. 10.1136/gut.2008.168443
    1. Nascimento DC, Melo PH, Piñeros AR, Ferreira RG, Colón DF, Donate PB, et al. . IL-33 contributes to sepsis-induced long-term immunosuppression by expanding the regulatory T cell population. Nat Commun. (2017) 8:14919. 10.1038/ncomms14919
    1. Nascimento DC, Alves-Filho JC, Sônego F, Fukada SY, Pereira MS, Benjamim C, et al. . Role of regulatory T cells in long-term immune dysfunction associated with severe sepsis. Crit Care Med. (2010) 38:1718–25. 10.1097/CCM.0b013e3181e78ad0
    1. Iwashyna TJ, Cooke CR, Wunsch H, Kahn JM. Population burden of long-term survivorship after severe sepsis in older Americans. J Am Geriatr Soc. (2012) 60:1070–7. 10.1111/j.1532-5415.2012.03989.x
    1. Prescott HC, Langa KM, Iwashyna TJ. Readmission diagnoses after hospitalization for severe sepsis and other acute medical conditions. JAMA (2015) 313:1055–7. 10.1001/jama.2015.1410
    1. Shen HN, Lu CL, Yang HH. Risk of recurrence after surviving severe sepsis: a matched cohort study. Crit Care Med. (2016) 44:1833–41. 10.1097/CCM.0000000000001824
    1. Prescott HC, Osterholzer JJ, Langa KM, Angus DC, Iwashyna TJ. Late mortality after sepsis: propensity matched cohort study. BMJ (2016) 353:i2735. 10.1136/bmj.i2375
    1. Tosh PK, McDonald LC. Infection control in the multidrug-resistant era: tending the human microbiome. Clin Infect Dis. (2012) 54:707–13. 10.1093/cid/cir899
    1. Dickson RP, Erb-Downward JR, Falkowski NR, Hunter EM, Ashley SL, Huffnagle GB. The lung microbiota of healthy mice are highly variable, cluster by environment, and reflect variation in baseline lung innate immunity. Am J Respir Crit Care Med. (2018) 198:497–508. 10.1164/rccm.201711-2180OC
    1. Chen YE, Fischbach MA, Belkaid Y. Skin microbiota-host interactions. Nature (2018) 553:427–36. 10.1038/nature25177
    1. Pfeiffer JK, Virgin HW. Viral immunity. Transkingdom control of viral infection and immunity in the mammalian intestine. Science (2016) 351:aad5872. 10.1126/science.aad5872
    1. Huffnagle GB, Noverr MC. The emerging world of the fungal microbiome. Trends Microbiol. (2013) 21:334–41. 10.1016/j.tim.2013.04.002
    1. Beura LK, Hamilton SE, Bi K, Schenkel JM, Odumade OA, Casey KA, et al. . Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature (2016) 532:512–6. 10.1038/nature17655
    1. Abolins S, King EC, Lazarou L, Weldon L, Hughes L, Drescher P, et al. . The comparative immunology of wild and laboratory mice, Mus musculus domesticus. Nat Commun. (2017) 8:14811. 10.1038/ncomms14811
    1. Rivard AL, Simura KJ, Mohammed S, Magembe AJ, Pearson HM, Hallman MR, et al. . Rat intubation and ventilation for surgical research. J Invest Surg. (2006) 19:267–74. 10.1080/08941930600778297
    1. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. . Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature (2015) 517:205–8. 10.1038/nature13828
    1. Caballero S, Kim S, Carter RA, Leiner IM, Sušac B, Miller L, et al. . Cooperating commensals restore colonization resistance to vancomycin-resistant enterococcus faecium. Cell Host Microbe. (2017) 21:592–602.e4. 10.1016/j.chom.2017.04.002

Source: PubMed

3
구독하다