Skip to main content
Erschienen in: Critical Care 1/2018

Open Access 01.12.2018 | Review

The emerging role of the microbiota in the ICU

verfasst von: Nora Suzanne Wolff, Floor Hugenholtz, Willem Joost Wiersinga

Erschienen in: Critical Care | Ausgabe 1/2018

Abstract

This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2018. Other selected articles can be found online at https://​www.​biomedcentral.​com/​collections/​annualupdate2018​. Further information about the Annual Update in Intensive Care and Emergency Medicine is available from http://​www.​springer.​com/​series/​8901.

Introduction

In recent years, the surge of culture-independent methods to study bacterial populations has led to an increasing body of evidence pointing towards the microbiome as an important player in the pathophysiology of a whole spectrum of diseases that affect the critically ill, including trauma and sepsis [14]. Techniques such as 16S rRNA and shotgun metagenomic sequencing have opened up a new area of research, enabling detailed investigations of complex populations of bacteria and their effects on health and disease [5, 6]. Some have even called the microbiome a separate organ given its numerous roles in metabolism, development of the immune system and host defense against pathogens [7].
Microbiota is an overarching term for all the microbes in a population, consisting of bacteria, archaea and eukarya [8]. Most studies that have been performed look at the bacterial microbiota because of its high abundance and high diversity. The collective of microbes in a population is referred to as the microbiota and the genetic content as the microbiome.
The real value of all this novel knowledge for the clinical care of patients on the intensive care unit (ICU) still has to be established. Nonetheless, data are accumulating that underscore the potential importance of the microbiome for intensive care medicine. On any given day, three-fourths of all patients on the ICU are treated with antibiotics, which are known to cause severe collateral damage to the microbiome [9]. Besides antibiotics, there are multiple external modulators of the gut microbiota applied during the clinical care of patients on the ICU, such as gastric acid inhibition, the route of feeding, sedatives and opioids [3, 10]. Novel strategies are being designed to intervene on the microbiome to prevent or treat trauma and sepsis. Excitingly, a very recent randomized, placebo-controlled trial among 4556 healthy infants in India showed that the oral administration of Lactobacillus plantarum in combination with fructooligosaccharide in the first week of life could reduce the occurrence of sepsis in the first 60 days of life [4]. Other evidence points towards the use of synbiotics as an adjunctive therapy to prevent postoperative complications, such as surgical site infections and sepsis among adult surgical patients [11]. Most research in this field has focused on the intestinal microbiome; however, current research is also starting to show the importance of the lung microbiome for ICU patients [1]. For example, enrichment of the lung microbiome with gut bacteria seems to play a role in the pathogenesis of acute respiratory distress syndrome (ARDS) [12].
In this chapter, we will discuss the emerging role of the bacterial microbiotas in the gut and the lung in the critically ill. First, we will discuss the techniques available to study the bacterial microbiome, then we will continue into the gut and lung microbiome and end with some key questions for future research in the field of microbiota-targeted therapies on the ICU.

Microbiota analyses

The composition of the microbiota in the gut has been studied extensively using 16S ribosomal RNA (rRNA) gene-targeted approaches. The use of this single genetic marker has revolutionized microbial ecology [13, 14]. It has become relatively easy to amplify the 16S rRNA encoding genes from environmental DNA. Nowadays, with next-generation sequencing techniques, many microbial environments can be studied in depth and at high resolution in space and time, using relatively straightforward procedures.
To address not only microbiota composition, but rather focus on the metabolic potential and actual activity of the intestinal microbiota, “meta’omic” approaches have emerged during the last decade and are now widely used [1517]. Each of the meta’omic approaches provides different information about the functional potential or activity profiles of a microbial community.
Metagenomics is used to determine the collective genomes of members present in a microbial community as well as their functional capacity. Metagenomics was used in the Meta HIT (Metagenomics of the Human Intestinal Tract) project and provided a human microbiome-derived gene catalogue with over 3 million genes, indicating a community of over 150 species in an individual and a 100-fold larger non-redundant gene set compared to the human gene complement [16].
Metatranscriptomics and metaproteomics are able to provide information about the functions expressed by the members of the community. For example, metaproteomics analysis in healthy humans revealed a difference in the amount of proteins expressed and the proteins predicted by metagenomics. Moreover, metaproteomics in fecal material not only gives information on the bacterial proteins, but also on the major human proteins, giving insights into the main host responses [18]. However, it is difficult and sometimes not possible to use conserved proteins to distinguish between species or even higher taxonomic levels (e.g., genus or family). Consequently, by using this technique, you lose some of the taxonomic information otherwise gained by using metagenomics, metatranscriptomics or targeted 16S rRNA sequencing.
Metatranscriptome analysis of the gastrointestinal tract microbiota enables elucidation of the specific functional roles microbes have in this complex community. Although initial studies on the human large intestine revealed that different functions are expressed among individuals, core functions of the microbiota appear to be consistently expressed in different individuals [19, 20]. Moreover, metatranscriptome analyses of the small intestinal microbiota underpinned the cross-feeding between two dominant members of the small intestinal microbiota, i.e., Streptococcus spp. and Veilonella spp., in which the lactate produced by Streptococcus spp. is used as a carbon and energy source by the Veillonella spp. [17]. Metatranscriptome analysis of the microbiota in humanized mice revealed that mice colonized with the microbiota obtained from a lean human donor had greater expression of genes involved in polysaccharide breakdown and in propionate and butyrate production as compared to those colonized with the microbiota of an obese human donor [21]. These findings imply that metatranscriptomics can provide insight into the differential activity profiles in the intestinal microbiota, and enables reconstruction of the metabolic activity profile of microbial communities.
Metabolomic approaches are used to detect and quantify the metabolites that are produced by the microbial community. This approach has been suggested to be applicable as a diagnostic tool in diseases that involve aberrations of the intestinal microbiota composition and activity [22]. However, as with metaproteomics, you also lose information on the specific bacteria producing these metabolites. To overcome the loss of taxonomic information in metaproteomics and metabolomics, these techniques are often combined with 16S rRNA sequencing, metagenomics or metatranscriptomics.
These tools often generate very complex datasets with many different measurements (e.g., species or function) under various conditions. Therefore, these multivariate meta’omics datasets need tools to simplify the datasets and focus on correlations between points of interest, such as dietary interventions and the bacterial community or the bacterial community and host responses. Multivariate statistics are used to handle these large datasets and enable a relatively quick focus on data of importance [23]. An overview of available techniques to analyze the microbiome is provided in Fig. 1.

The gut microbiota

Human individuals can harbor over 150 different microbial species in their gut, which collectively encode more than 100-fold more non-redundant genes than there are in the human genome [16, 24]. More recent data, however, has challenged this number, suggesting that the ratio between bacteria and human cells is closer to 1:1 [25]. In healthy humans, the intestinal microbiota consists of members of all three domains of life: bacteria, archaea and eukarya, of which the bacterial community is the most abundant and diverse [8]. Nine different bacterial phyla have been recorded in humans so far, of which the bacteroidetes and firmicutes dominate [8, 16, 24]. Many of these bacteria in the gut have not been cultivated. Recent breakthroughs in the successful culture of the previously ‘unculturable’ human microbiota have revealed a whole spectrum of novel bacterial species and taxa [26]. The application of novel sequencing techniques provides an opportunity to understand this complex ecosystem much better [8, 13].
The intestinal microbiota plays a critical role in priming the host’s immune system, gut maturation and gut functions, such as nutrient uptake and metabolism, mucosal barrier function, enteric nervous system and motility [2729]. Numerous host genes seem to be specifically altered in response to various members of the microbiota, showing the importance of the microbial composition to the body’s response [30, 31].

Modulators of the microbiota on the ICU

The healthy intestinal microbiota is affected primarily by the host and the diet. In critically ill patients, these two factors both play an important role. However, in the critically ill it is also important to note that pathogens are able to outcompete other members of the intestinal microbiota more easily than in healthy humans, also changing the microbial composition. These pathogens benefit from a changed environment in critically ill patients, which are (though not per se all at the same time) decreased transit time, lack of ‘normal’ nutrition, oxygen levels and antibiotic usage [1]. Most of these opportunistic pathogens, except for Clostridium difficile infections, are Gram-negative aerobic species belonging to the proteobacteria, which thrive under the changed environment in critically ill patients. The universal use of antibiotics is probably the main cause of the severe bacterial dysbiosis seen in patients admitted to the ICU [3236]. In addition, because of no or limited nutritional intake by patients on the ICU, nutrition for the microbiota in the intestine also decreases, affecting the microbiota composition. For example, in the large intestine, the microbiota ferments non-digested dietary fibers and dietary and host-derived proteins. The fermentation of fibers is necessary for the production of butyrate by the microbiota, which epithelial cells use as an energy source. The study of the clinical relevance of these effects in critically ill patients is, however, still in its infancy.

The potential clinical relevance of the gut microbiota on the ICU

An imbalance in the homeostasis or ‘dysbiosis’ of the gut microbiota has been associated with a range of different diseases, including diabetes, obesity, inflammatory bowel disease and rheumatoid arthritis [5, 6]. This association with disease has led to investigations into the gut microbiota’s involvement at the systemic level. In the critically ill, the intestinal microbiota has been analyzed in a couple of studies so far. In general, we can state that critically ill patients admitted to the ICU have a state of dysbiosis of the intestinal microbiota [3234]. Figure 2 shows a schematic representation of the composition of the intestinal microbiota in the critically ill in comparison to a healthy human. Overall, the intestinal microbiota of critically ill patients admitted to the ICU with sepsis is characterized by lower diversity, lower abundance of key commensal genera (such as Faecalibacterium, Blautia, Ruminococcus), and in some cases overgrowth (over 50% relative abundance) by one genera, such as Escherichia/Shigella, Salmonella, Enterococcus, C. difficile or Staphylococcus [3236].
A normal, healthy intestinal microbiota protects against an invasion of pathogens like Enterococcus faecium, Escherichia coli and C. difficile. It comes as no surprise that severe infections caused by these pathogens often occur in patients with a recent history of antibiotic use. The microbiota of these patients is therefore probably disrupted, which allows for the overgrowth of antibiotic-resistant pathogenic and opportunistic bacteria that are frequently encountered in hospital settings [37].
In sepsis, the focus often lies on the identification of a single pathogen as the causative agent. However, there is an increasing realization that most pathogens do not act in isolation and that infections have ‘polymicrobial’ phenotypes and are thus linked to the status of the microbiota in the patient. The initial state of the microbiota can influence susceptibility to infection [38] and severity of infection [39]. Recent preclinical data derived from animal models suggest that the gut microbiome plays a protective role in the host defense against sepsis [3941]. As an example, antibiotic induced disruption of the gut microbiome leads to increased inflammation and bacterial dissemination in murine models of both Gram-positive and Gram-negative pneumosepsis [39, 40]. ICU patient data have suggested that the loss of microbiome diversity could predict the length of stay of patients on the ICU further underscoring the potential clinical relevance of the gut microbiome for intensive care medicine [33].

The evolving role of the lung microbiota in critical illness

Healthy lungs were long thought to be sterile, until recent studies showed that bacteria can be cultured from healthy lungs [42]. The lung environment is less advantageous for bacteria to grow and flourish when compared to the intestine, leading to a less dense bacterial community compared to the gut. A lack of nutrients, the bidirectional movement of the lung, the aerobic environment, and the coating of alveoli with a lipid-rich surfactant that has bacteriostatic effects all contribute to this harsh environment for bacteria [42, 43]. Furthermore, obtaining a sample from the lung is far more invasive than obtaining a stool sample to study the microbiome. Contamination from the sampling device or from the upper respiratory tract is another point to take into consideration.
In a healthy lung, the microbiota is delicately balanced by the reproduction rate of present bacteria and the immigration and elimination rate of bacteria. Under normal circumstances, the reproduction rate of bacteria remains low and the immigration and elimination high. However, in critical illness, sedatives and endotracheal intubation can decrease the mucociliary clearance and cough reflex, leading to decreased microbial elimination. Furthermore, mechanical ventilation can cause an increase in alveolar edema, which can lead to an increase in available nutrients in the lung and areas where the oxygen levels are lower, allowing bacteria to thrive [1].
In healthy lungs, it is thought that most of the bacteria come from the oral microbiota. The lung microbiota most closely resembles the microbiota of the oropharynx, more so than that of the nasopharynx, gastrointestinal tract or inhaled air [42]. The healthy oropharynx contains benign Veillonella spp. and Prevotella spp., and, therefore, these are also found in healthy lungs. During critical illness, the oropharynx can become overpopulated with pathogenic proteobacteria, such as Pseudomonas aeruginosa and Klebsiella pneumoniae. Furthermore, in critical illness the stomach and small intestine can become the primary source of bacterial migration to the lung [1, 42].
ARDS and pneumonia are thought to cause alveolar injury and as a consequence induce changes in the microbiome. The alveoli become covered with protein-rich fluids and the bactericidal surfactant from the alveoli becomes inactivated, making the alveoli a more hospitable environment for bacteria [1]. Furthermore, Dickson et al. [44] proposed a theory regarding the existence of a positive feedback loop between the growth of certain bacterial species in the lung and the local inflammatory response; as the bacterial population grows, it starts to limit itself due to nutrient shortage while it also provokes an increased inflammatory response. This inflammatory response can consequently lead to endothelial and epithelial injury, releasing fluids that are rich in proteins and nutrients, thus stimulating bacterial growth. Further bacterial growth will increase the local inflammation, thus creating a positive feedback loop. This suggests that in some cases the body’s inflammatory response may be making the infection worse.
The intestinal microbiota has emerged as a key component of both local and systemic immunity. Epithelial and immune cells gain information directly from bacteria and local cytokine responses and subsequently adjust inflammatory responses. Microbiome research combining the gut and lung has started to show an association between the composition of the intestinal microbiota and lung health [45, 46]. Experimental germ-free and antibiotic murine models have shown that the body’s microbiota is important in the defense against influenza and several types of bacterial pneumonia [46]. Moreover, probiotics containing Lactobacillus spp. and Bifidobacterium spp. have been shown to improve incidence and outcome of respiratory infections. Additionally, exposure to Toll-like receptor (TLR) agonists and nucleotide-binding oligomerization domain (NOD)-like receptors in the intestine, by substances such as lipopolysaccharide (LPS), peptidoglycan and lipoteichoic acid, has been shown to increase the lungs’ ability to clear bacteria [45]. These studies suggest that the intestinal microbiota is important for airway defenses. Moreover, human and murine studies have shown that the lungs can contain gut-associated bacteria during sepsis and ARDS. However, this gut-lung axis does not seem to be a one-way street. Pulmonary infection with tuberculosis, influenza and Burkholderia pseudomallei have all been shown to have a significant effect on the composition of the gut microbiome in murine models [40, 47, 48]. Figure 3 shows a schematic overview of the potential consequences of microbial dysbiosis in the critically ill in both the lung and intestine.

The road ahead

Hospital-acquired infections are a huge problem in patients on the ICU. Antibiotic administration is the usual form of treatment or prevention of these infections and is lifesaving. However, as a side-effect, it severely affects the microbiota, risks induction of antibiotic resistant pathogens and potentially even the new onset of severe infection. Restoring the microbiota after antibiotic treatment has the potential to reduce infections [10, 23]. Different approaches can be utilized: probiotics, prebiotics (a dietary ingredient that promotes beneficial microbiota) and synbiotics (a combination of a probiotic with a prebiotic) have all been tested in various patient groups on the ICU with mixed results [4, 49]. Probiotics will not reestablish a complete microbiota. This can be achieved to a larger extent by performing a fecal microbiota transfer (FMT). FMT has been used successfully to restore re-occurring C. difficile infections in patients on the ICU. The first case reports on the successful use of FMT in patients with therapy-refractory sepsis have been published [10]. A key factor, however, will be establishing the exact mechanism by which certain components of the gut and/or lung microbiome play their presumed protective effects on the health of patients admitted to the ICU. This could enable us to develop targeted therapies to restore the microbiome in this very vulnerable patient population.
At the present time, we lack information on the dynamics of the microbiota in the critically ill in order to develop targeted therapies to restore the microbiome. There is a need for information on how the microbiota restores with and without interference or therapies in the critically ill. Moreover, these patients should be followed for long-term periods to monitor re-occurring infections. This information will aid in developing targeted approaches, such as the use of probiotics or FMT, to restore the microbiota and prevent infections during a patient’s recovery.

Conclusion

The importance of the intestinal and lung microbiotas is often overlooked on the ICU. Currently, we can explore the microbiome using a vast array of techniques, giving us ‘meta’ libraries of data, which has allowed researchers to show the potential crucial role of the microbiota for ICU patients. During a patient’s stay on the ICU, their microbiota is influenced by both their illness and the care provided. For example, the gut microbiota of critically ill patients admitted to the ICU with sepsis is characterized by lower diversity, lower abundance of key commensal genera and, in some cases, an overgrowth by one genera. These changes in the microbiota can, in turn, affect patient outcome and susceptibility to infection. Furthermore, the lung microbiota has only recently been shown to be important in the critically ill. Intriguingly, recent evidence also points to a bi-directional microbiota-mediated role between the gut and the lung, the so called gut-lung axis. The importance of these microbiotas is pushing us towards new types of treatments, in which we also start to treat the microbiota.

Acknowledgements

This work was supported by the Netherlands Organization for Scientific Research (NWO), the Horizon2020 Marie Skłodowska-Curie International Training Network “the European Sepsis Academy” and The Netherlands Organization for Health Research development (ZonMw).

Acknowledgements

Not Applicable.

Funding

Publication costs were funded by the Center for Experimental and Molecular Medicine (CEMM).

Availability of data and materials

Not applicable.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
3.
Zurück zum Zitat Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2:135–43.CrossRefPubMed Haak BW, Wiersinga WJ. The role of the gut microbiota in sepsis. Lancet Gastroenterol Hepatol. 2017;2:135–43.CrossRefPubMed
4.
Zurück zum Zitat Panigrahi P, Parida S, Nanda NC, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548:407–12.CrossRefPubMed Panigrahi P, Parida S, Nanda NC, et al. A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature. 2017;548:407–12.CrossRefPubMed
5.
Zurück zum Zitat Blaut M, Clavel T. Metabolic diversity of the intestinal microbiota: implications for health and disease. J Nutr. 2007;137(3 Suppl 2):751S–5S.CrossRefPubMed Blaut M, Clavel T. Metabolic diversity of the intestinal microbiota: implications for health and disease. J Nutr. 2007;137(3 Suppl 2):751S–5S.CrossRefPubMed
7.
Zurück zum Zitat Baquero F, Nombela C. The microbiome as a human organ. Clin Microbiol Infect. 2012;18(Suppl 4):2–4.CrossRefPubMed Baquero F, Nombela C. The microbiome as a human organ. Clin Microbiol Infect. 2012;18(Suppl 4):2–4.CrossRefPubMed
8.
Zurück zum Zitat Rajilic-Stojanovic M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38:996–1047.CrossRefPubMedPubMedCentral Rajilic-Stojanovic M, de Vos WM. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38:996–1047.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Vincent J-L, Rello J, Marshall J, et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA. 2009;302:2323–9.CrossRefPubMed Vincent J-L, Rello J, Marshall J, et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA. 2009;302:2323–9.CrossRefPubMed
10.
Zurück zum Zitat Haak BW, Levi M, Wiersinga WJ. Microbiota-targeted therapies on the intensive care unit. Curr Opin Crit Care. 2017;23:167–74.CrossRefPubMed Haak BW, Levi M, Wiersinga WJ. Microbiota-targeted therapies on the intensive care unit. Curr Opin Crit Care. 2017;23:167–74.CrossRefPubMed
11.
Zurück zum Zitat 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(suppl 2):S153–60.CrossRefPubMed 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(suppl 2):S153–60.CrossRefPubMed
12.
Zurück zum Zitat 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 Microbiol. 2016;1:16113.CrossRefPubMedPubMedCentral 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 Microbiol. 2016;1:16113.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Tringe SG, Hugenholtz P. A renaissance for the pioneering 16S rRNA gene. Curr Opin Microbiol. 2008;11:442–6.CrossRefPubMed Tringe SG, Hugenholtz P. A renaissance for the pioneering 16S rRNA gene. Curr Opin Microbiol. 2008;11:442–6.CrossRefPubMed
14.
Zurück zum Zitat Pace NR, Sapp J, Goldenfeld N. Phylogeny and beyond: scientific, historical, and conceptual significance of the first tree of life. Proc Natl Acad Sci U S A. 2012;109:1011–8.CrossRefPubMedPubMedCentral Pace NR, Sapp J, Goldenfeld N. Phylogeny and beyond: scientific, historical, and conceptual significance of the first tree of life. Proc Natl Acad Sci U S A. 2012;109:1011–8.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Fritz JV, Desai MS, Shah P, Schneider JG, Wilmes P. From meta-omics to causality: experimental models for human microbiome research. Microbiome. 2013;1:14.CrossRefPubMedPubMedCentral Fritz JV, Desai MS, Shah P, Schneider JG, Wilmes P. From meta-omics to causality: experimental models for human microbiome research. Microbiome. 2013;1:14.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Zoetendal EG, Raes J, van den Bogert B, et al. The human small intestinal microbiota is driven by rapid uptake and conversion of simple carbohydrates. ISME J. 2012;6:1415–26.CrossRefPubMedPubMedCentral Zoetendal EG, Raes J, van den Bogert B, et al. The human small intestinal microbiota is driven by rapid uptake and conversion of simple carbohydrates. ISME J. 2012;6:1415–26.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Verberkmoes NC, Russell AL, Shah M, et al. Shotgun metaproteomics of the human distal gut microbiota. ISME J. 2009;3:179–89.CrossRefPubMed Verberkmoes NC, Russell AL, Shah M, et al. Shotgun metaproteomics of the human distal gut microbiota. ISME J. 2009;3:179–89.CrossRefPubMed
19.
Zurück zum Zitat Franzosa EA, Morgan XC, Segata N, et al. Relating the metatranscriptome and metagenome of the human gut. Proc Natl Acad Sci U S A. 2014;111:E2329–38.CrossRefPubMedPubMedCentral Franzosa EA, Morgan XC, Segata N, et al. Relating the metatranscriptome and metagenome of the human gut. Proc Natl Acad Sci U S A. 2014;111:E2329–38.CrossRefPubMedPubMedCentral
20.
21.
Zurück zum Zitat Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214.CrossRefPubMed Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341:1241214.CrossRefPubMed
23.
Zurück zum Zitat Martin FP, Wang Y, Sprenger N, et al. Top-down systems biology integration of conditional prebiotic modulated transgenomic interactions in a humanized microbiome mouse model. Mol Syst Biol. 2008;4:205.PubMedPubMedCentral Martin FP, Wang Y, Sprenger N, et al. Top-down systems biology integration of conditional prebiotic modulated transgenomic interactions in a humanized microbiome mouse model. Mol Syst Biol. 2008;4:205.PubMedPubMedCentral
24.
Zurück zum Zitat Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915–20.CrossRefPubMed Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915–20.CrossRefPubMed
25.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
26.
Zurück zum Zitat Browne HP, Forster SC, Anonye BO, et al. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature. 2016;533:543–6.CrossRefPubMedPubMedCentral Browne HP, Forster SC, Anonye BO, et al. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature. 2016;533:543–6.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Ciccia F, Guggino G, Rizzo A, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann Rheum Dis. 2017;76:1123–32.CrossRefPubMed Ciccia F, Guggino G, Rizzo A, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann Rheum Dis. 2017;76:1123–32.CrossRefPubMed
28.
Zurück zum Zitat Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7.CrossRefPubMed Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7.CrossRefPubMed
29.
Zurück zum Zitat Backhed F, Roswall J, Peng Y, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703.CrossRefPubMed Backhed F, Roswall J, Peng Y, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703.CrossRefPubMed
30.
Zurück zum Zitat Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Molecular analysis of commensal host-microbial relationships in the intestine. Science. 2001;291:881–4.CrossRefPubMed Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Molecular analysis of commensal host-microbial relationships in the intestine. Science. 2001;291:881–4.CrossRefPubMed
31.
Zurück zum Zitat Leser TD, Molbak L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ Microbiol. 2009;11:2194–206.CrossRefPubMed Leser TD, Molbak L. Better living through microbial action: the benefits of the mammalian gastrointestinal microbiota on the host. Environ Microbiol. 2009;11:2194–206.CrossRefPubMed
32.
Zurück zum Zitat 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 Med. 2017;43:59–68.CrossRefPubMed 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 Med. 2017;43:59–68.CrossRefPubMed
34.
Zurück zum Zitat Wischmeyer PE, McDonald D, Knight R. Role of the microbiome, probiotics, and ‘dysbiosis therapy’in critical illness. Curr Opin Crit Care. 2016;22:347–53.CrossRefPubMedPubMedCentral Wischmeyer PE, McDonald D, Knight R. Role of the microbiome, probiotics, and ‘dysbiosis therapy’in critical illness. Curr Opin Crit Care. 2016;22:347–53.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Zaborin A, Smith D, Garfield K, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio. 2014;5:e1361–14.CrossRef Zaborin A, Smith D, Garfield K, et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. MBio. 2014;5:e1361–14.CrossRef
36.
Zurück zum Zitat Ojima M, Motooka D, Shimizu 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.CrossRefPubMed Ojima M, Motooka D, Shimizu 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.CrossRefPubMed
38.
Zurück zum Zitat Yooseph S, Kirkness EF, Tran TM, et al. Stool microbiota composition is associated with the prospective risk of plasmodium falciparum infection. BMC Genomics. 2015;16:631.CrossRefPubMedPubMedCentral Yooseph S, Kirkness EF, Tran TM, et al. Stool microbiota composition is associated with the prospective risk of plasmodium falciparum infection. BMC Genomics. 2015;16:631.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Schuijt TJ, Lankelma JM, Scicluna BP, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65:575–83.CrossRefPubMed Schuijt TJ, Lankelma JM, Scicluna BP, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65:575–83.CrossRefPubMed
40.
Zurück zum Zitat Lankelma JM, Birnie E, Weehuizen TAF, et al. The gut microbiota as a modulator of innate immunity during melioidosis. PLoS Negl Trop Dis. 2017;11:e5548.CrossRef Lankelma JM, Birnie E, Weehuizen TAF, et al. The gut microbiota as a modulator of innate immunity during melioidosis. PLoS Negl Trop Dis. 2017;11:e5548.CrossRef
41.
Zurück zum Zitat Deshmukh HS, Liu Y, Menkiti OR, et al. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia Coli K1 sepsis in neonatal mice. Nat Med. 2014;20:524–30.CrossRefPubMedPubMedCentral Deshmukh HS, Liu Y, Menkiti OR, et al. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia Coli K1 sepsis in neonatal mice. Nat Med. 2014;20:524–30.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Huffnagle GB, Dickson RP, Lukacs NW. The respiratory tract microbiome and lung inflammation: a two-way street. Mucosal Immunol. 2017;10:299–306.CrossRefPubMed Huffnagle GB, Dickson RP, Lukacs NW. The respiratory tract microbiome and lung inflammation: a two-way street. Mucosal Immunol. 2017;10:299–306.CrossRefPubMed
43.
44.
Zurück zum Zitat Dickson RP, Erb-Downward JR, Huffnagle GB. Towards an ecology of the lung: new conceptual models of pulmonary microbiology and pneumonia pathogenesis. Lancet Respir Med. 2014;2:238–46.CrossRefPubMedPubMedCentral Dickson RP, Erb-Downward JR, Huffnagle GB. Towards an ecology of the lung: new conceptual models of pulmonary microbiology and pneumonia pathogenesis. Lancet Respir Med. 2014;2:238–46.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Budden KF, Gellatly SL, Wood DL. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15:55–63.CrossRefPubMed Budden KF, Gellatly SL, Wood DL. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15:55–63.CrossRefPubMed
46.
Zurück zum Zitat Marsland BJ, Trompette A, Gollwitzer ES. The gut-lung axis in respiratory disease. Ann Am Thorac Soc. 2015;12(Suppl 2):S150–6.PubMed Marsland BJ, Trompette A, Gollwitzer ES. The gut-lung axis in respiratory disease. Ann Am Thorac Soc. 2015;12(Suppl 2):S150–6.PubMed
47.
Zurück zum Zitat Winglee K, Eloe-Fadrosh E, Gupta S, Guo H, Fraser C, Bishai W. Aerosol mycobacterium tuberculosis infection causes rapid loss of diversity in gut microbiota. PLoS One. 2014;9:e97048.CrossRefPubMedPubMedCentral Winglee K, Eloe-Fadrosh E, Gupta S, Guo H, Fraser C, Bishai W. Aerosol mycobacterium tuberculosis infection causes rapid loss of diversity in gut microbiota. PLoS One. 2014;9:e97048.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Deriu E, Boxx GM, He X, et al. Influenza virus affects intestinal microbiota and secondary salmonella infection in the gut through type i interferons. PLoS Pathog. 2016;12:e1005572.CrossRefPubMedPubMedCentral Deriu E, Boxx GM, He X, et al. Influenza virus affects intestinal microbiota and secondary salmonella infection in the gut through type i interferons. PLoS Pathog. 2016;12:e1005572.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Crit Care. 2016;19:262.CrossRefPubMed Manzanares W, Lemieux M, Langlois PL, Wischmeyer PE. Probiotic and synbiotic therapy in critical illness: a systematic review and meta-analysis. Crit Care. 2016;19:262.CrossRefPubMed
Metadaten
Titel
The emerging role of the microbiota in the ICU
verfasst von
Nora Suzanne Wolff
Floor Hugenholtz
Willem Joost Wiersinga
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Critical Care / Ausgabe 1/2018
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-018-1999-8

Weitere Artikel der Ausgabe 1/2018

Critical Care 1/2018 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.