Next Article in Journal
Evolutionary Divergent Suppressor Mutations in Conformational Diseases
Next Article in Special Issue
Bioengineering Strategies for Protein-Based Nanoparticles
Previous Article in Journal
Ensemble Consensus-Guided Unsupervised Feature Selection to Identify Huntington’s Disease-Associated Genes
Previous Article in Special Issue
The Multiplanetary Future of Plant Synthetic Biology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeted Approaches for In Situ Gut Microbiome Manipulation

1
Department of Biochemistry, Yong Loo Lin School of Medicine, 8 Medical Drive, Singapore 117596, Singapore
2
NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
3
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore
4
Khoo Teck Puat-National University Children’s Medical Institute, National University Health System, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore
*
Author to whom correspondence should be addressed.
Contributed equally.
Genes 2018, 9(7), 351; https://doi.org/10.3390/genes9070351
Submission received: 28 May 2018 / Revised: 7 July 2018 / Accepted: 9 July 2018 / Published: 12 July 2018
(This article belongs to the Special Issue Emerging Applications in Synthetic Biology)

Abstract

:
Microbial communities and their collective genomes form the gut microbiome, of which bacteria are the major contributor. Through their secreted metabolites, bacteria interact with the host, influencing human health and physiology. Perturbation of the microbiota and metabolome has been associated with various diseases and metabolic conditions. As knowledge on fundamental host-microbiome interactions and genetic engineering tools becomes readily available, targeted manipulation of the gut microbiome for therapeutic applications gains favourable attention. Manipulation of the gut microbiome can be achieved by altering the microbiota population and composition, or by modifying the functional metabolic activity of the microbiome to promote health and restore the microbiome balance. In this article, we review current works that demonstrate various strategies employed to manipulate the gut microbiome in situ to various degrees of precision.

1. Introduction

The human gastrointestinal tract (GIT) is colonised by trillions of microorganisms—namely bacteria, archaea, bacteriophages, and eukaryotes, which form a massive, ecological community through complex metabolic activities and constant host interactions. This community of microorganisms is defined as the gut microbiota, and its collective genomes are known as the gut microbiome [1,2]. Bacterial species are the most dominant members of the gut microbiota, and their genes account for approximately 99% of the gut microbiome [3]. Though the total bacterial cell mass in a healthy individual is estimated to be only around 200 g [4], the bacterial gut metagenome overwhelms the number of distinct genes found in the human genome by 150 times [5]. Thus, the gut microbiota has an extensive impact on both normal human physiology and disease susceptibilities, including defence against pathogens [6], nutrient utilisation [7], and peripheral education of the immune system [8].
The gut microbiome directs a host of metabolic pathways, interacting dynamically with the host through the metabolites generated. Changes in these metabolic pathways may alter the composition of gut lumen metabolites, perturbing the original balance in the host system. Responses from the host are dynamic and lead to the up- or down-regulation of related metabolic pathways until a new balance is attained. Dysbiosis of the microbiota in the host has been linked to various diseases, such as obesity, colorectal cancer, cardiovascular disease, and inflammatory bowel diseases (IBD) [9]. The restoration of healthy gut microbiota is believed to have positive effects on the treatment of these diseases. Moreover, the regulation of certain microbial groups in healthy individuals can be helpful in disease prevention. Therefore, manipulation of the gut microbiome presents valuable avenues for therapeutic and clinical applications.
To positively impact health through therapeutic modulation of gut dysbiosis, the gut microbiome can be altered by changing the population, composition, and/or the functional output of metabolic pathways. Specifically, direct manipulation of the gut microbiome can be achieved by using targeted prebiotics, probiotics, engineered probiotics, or bacteriophages for targeted effects (Figure 1). Here, we give an overview of the progress in applying these approaches to directly modulate the gut microbiome in situ.

2. Prebiotics-Mediated Modulation of Gut Microbiome

Over the years, studies in the areas of metabolic syndrome, obesity, and prebiotics have significantly advanced knowledge on the gut microbiome and its role in maintaining human health. Modulating gut microbiota composition through diet manipulation, or, more specifically, through prebiotics, has garnered significant attention due to the (1) rising phenomenon of obesity in developed countries and increasing evidence of the gut microbiota’s influence, (2) ease of using prebiotics as a therapeutic approach, and (3) the increasing knowledge of the benefits of known metabolites through bacterial fermentation.
The current definition of prebiotics is proposed as ‘a substrate that is selectively utilised by host microorganisms conferring a health benefit’ [10]. Based on this, prebiotics need to be resistant to gastric digestion in the upper alimentary canal, and undergo fermentation by colonic bacteria to promote the growth of certain bacterial populations that are positively associated with health [10]. Due to these prerequisites, the most commonly known prebiotics used as target modifiers of the gut microbiome are non-digestible carbohydrates (NDC), such as fructo-oligosaccharides, galacto-oligosaccharides, inulin, and oligosaccharides [11]. Non-carbohydrate substrates that encompass similar characteristics, including polyphenols [12,13], phenolic acids [14], minerals, and peptides [15], are also being considered for use as prebiotics. However, the use of these non-carbohydrates is yet to be the focus in recent studies on prebiotics.
Though, seemingly, a general approach to manipulate the gut microbiome, recent studies on prebiotics focus on supplementing NDC to select Bifidobacterium, which is over-represented among the saccharolytic commensals [16], and is known to play important roles in carbohydrate utilisation [17]. Decreased levels of Bifidobacterium have been observed and associated with obesity and Type II diabetes, while supplementation of NDC in murine models [18] and human subjects [19,20] has been found to restore those levels. The major end-products of NDC fermentation are short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate [21,22,23,24]. Acetate and propionate are involved in gluconeogenesis in hepatocytes and act as signalling molecules [25], while butyrate is an important carbon source for epithelial cells in the colon [26]. Interestingly, during NDC supplementation, the growth of butyrate-producing bacteria has been observed to increase, along with Bifidobacterium [18,27,28,29,30,31]. Prebiotic supplementation promotes the growth of Bifidobacterium and their utilisation of oligosaccharides to produce fermentative end products, mainly acetate and lactate. The excess acetate produced is subsequently utilised by butyrate-producing bacteria, such as Faecalibacterium prausnitzii, Roseburia, and Eubacterium, to produce butyrate. This ‘cross-feeding’ effect between Bifidobacterium and butyrate-producing bacteria ultimately leads to an increased butyrate production and augments beneficial effects to the host, such as improvement of the gut barrier integrity and pathogen inhibition (Figure 2) [26]. The health-promoting attributes of these butyrate-producing bacteria are supported in numerous diseased conditions, such as IBD [32], Crohn’s disease [33], and ulcerative colitis [34], where a significant reduction of butyrate-producing bacteria is reported. Though these butyrate-producing bacteria are not directly affected by the supplementation of oligosaccharides, their butyrate production is elevated due to the increased availability of fermentative end products generated by Bifidobacterium. Such cross-feeding behaviour reveals the limitations of prebiotics as targeted modifiers of the gut microbiota for specific effects. Nevertheless, this highlights the role of prebiotics in mediating complex interactions among populations in the gut microbiota, which presents opportunities to delve into more therapeutic approaches.
There are several limitations in the efficacy and efficiency of modulating gut microbiota via prebiotics. Efficacy can be compromised as a result of high inter-individual variability in the gut microbiome profile [35,36], in which individual responses may vary significantly, and results are difficult to reproduce. Efficiency, on the other hand, is diminished due to the transient lifespan of prebiotics as they pass through the GIT. While prebiotics favour and promote the growth of certain bacterial populations, this growth may be transient and may limit the extension of health benefits to the host. Prebiotics, therefore, must be constantly consumed or administered for a significant effect to be observed. Currently, natural prebiotics are most commonly used to selectively boost the growth of existing bacteria, or to modulate metabolic pathways present in the gut, though the precise mechanisms have yet to be elucidated.

3. Cell-Mediated Modulation of Gut Microbiome

3.1. Modulation through Targeted Probiotics

Beside prebiotics, probiotics are being used as targeted modifiers to modulate the gut microbiome. Probiotics are live microorganisms that, when administered in adequate amounts, confer health benefits to the host [37]. Probiotics generally promote gut health through mechanisms, such as regulation of pH levels and colonisation resistance [37]. Supplementation of probiotics is a direct way of manipulating the gut microbiome as it changes the microbiota composition. Most probiotics are, in fact, original gut commensals that have been isolated and characterised. Through various metabolic activities, probiotics can directly modulate the microbiota to exert beneficial effects on the host, as seen commonly during the treatment of gut dysbiosis. Well-known, gram-positive bacteria that confer such health benefits include Bifidobacterium and Lactobacillus. Bifidobacterium adolescentis IM38, for example, is reported to exert anti-inflammatory and anti-colitis effects on mice by modulating the ratio of Proteobacteria and Bacteroidetes populations [38]. Bifidobacterium, which has an innate metabolism for glycan harvesting, has also been shown to regulate and favour the expansion of species of its genus, such as Bifidobacterium breve, Bifidobacterium longum, and Bifidobacterium bifidum, leading to increased SCFAs, particularly acetate and butyrate, in the murine cecum [39]. Moreover, the supplementation of bifidobacterial strains was observed to modulate other populations of bacteria, such as Lachnospiraceae, which has been reported to be correlated with the early onset of diabetes [39,40]. Likewise, Lactobacillus strains, such as Lactobacillus rhamnosus GG and Lactobacillus sakei, have been shown to regulate gut microbiota profiles and lower obesity-related markers in mice, presumably through antagonistic activities against Clostridium [41]. In addition to gram-positive bacteria, gram-negative probiotics, such as Escherichia coli Nissle 1971 (EcN), have also been well-characterised [42]. Particularly, EcN administration has been shown to inhibit gut pathogens through various mechanisms, such as nutrient limitation [43] and colonisation resistance [44,45]. These examples demonstrate that the administration of targeted probiotics can directly modulate the gut microbiota by altering the population and metabolic output of the microbiome to actively re-establish a healthy balance. The beneficial effects of probiotics can be further harnessed through concurrent supplementation of both prebiotics and probiotics, also known as synbiotics, where the growth of the supplemented probiotics is specifically enhanced by the prebiotics [46,47]. In synbiotics, both prebiotics and probiotics are the modifiers of the microbiome. Specifically, the main role of prebiotics in synbiotics is to enhance the growth of probiotics so that the sheer increase in the population would lead to beneficial effects.
Despite these promising proofs-of-concept studies, probiotics have not shown significance in many clinical trials [48,49]. This is likely due to differences in genetic backgrounds and gut microbiome profiles across individuals [50,51]. One of the major drawbacks lies in the difficulty of isolating and culturing intestinal commensal microbes, which are mostly obligate anaerobes [52]. Furthermore, probiotics are known to colonise the intestinal tract for only short durations, which explains the frequent administration required for tangible health effects [53]. To address these drawbacks, researchers are seeking new methods to isolate and culture gut commensals [54,55,56]. The findings from these studies can contribute to discovering potential probiotic strains with distinct health effects and a niche colonisation.

3.2. Targeted Modulation through Engineered Probiotics

Probiotics can directly alter the composition of the gut microbiota for host benefits, yet the precise mechanisms are known for only a few strains [37]. As such, targeted approaches to modulate the gut microbiome are more likely to achieve therapeutic significance for the host.
One targeted approach is through the rational engineering of probiotics to directly alter the functional output of the microbiome; a specific metabolic output is achieved and host benefits are conferred without unnecessarily causing major alterations to the microbiota population and composition. This approach often involves introducing non-native pathways in probiotics through heterologous expression of enzymes. The additional functionalities allow probiotics to wield more targeted effects on the host, rather than confer general positive host benefits. For example, EcN was engineered to produce N-acylphosphatidylethanolamines, which are precursors to a family of anorexigenic lipids [57]. Incorporation of this engineered EcN into mice on a high-fat diet showed a reduction in fat mass and weight gain compared to mice supplemented with wild-type EcN [57]. Significant changes in fatty acid oxidation-related gene expression were observed, demonstrating the targeted effect of an engineered microbe [57]. In another study, EcN was engineered as a chassis to deliver glucagon-like peptide-1 (GLP-1) in diabetic rats to restore the insulin sensitivity of intestinal cells [58]. This approach directly addresses the impaired function of the gut microbiome as it has been postulated that diminished SCFAs levels in the diabetic gut with dysbiosis are linked to reduced GLP-1 secretion [59]. Other studies have harnessed lactic acid bacteria, specifically Lactococcus lactis, for the production and delivery of bioactive molecules, such as interleukin-10, for specific metabolic effects, including anti-inflammatory effects [60,61,62]. Apart from the delivery of therapeutic compounds, probiotics were also engineered for pathogen sensing, suppression, and elimination, specifically in the gut. For example, an improved ‘sense-kill’ system [63] employed in EcN was shown to successfully eradicate and prevent Pseudomonas aeruginosa colonisation in both Caenorhabditis elegans and murine models [64]. In a recent study, Mao et al. [65] engineered lactic-acid producing L. lactis to detect quorum-sensing molecules produced by Vibrio cholera, and express an enzymatic reporter upon detection. The engineered L. lactis was found to specifically detect and report the presence of V. cholerae and significantly improve the survival rate of infected mice [65]. Besides well-known probiotics, there has been recent progress in the engineering of gut commensals, which also presents great potential for bio-medical applications. Specifically, in Bacteroides thetaiotaomicron, components for tunable gene expression were developed and characterised. Expected functional outputs were observed in mice after administration of these engineered B. thetaiotaomicron [66,67]. These present opportunities to harness such engineered commensals for therapeutic purposes. Taken together, these studies demonstrate the feasibility of using engineered bacteria to directly manipulate the functional output of the microbiota without major modulation of the microbiota population and composition.
Precision approaches that integrate prebiotics and engineered probiotics to accentuate the targeted effects have also emerged. EcN has been reported to be engineered to specifically target and kill colorectal cancer cells in vivo [68]. Interestingly, the engineered EcN expresses and secretes a myrosinase that transforms host-ingested glucosinolate, a compound mostly found in cruciferous vegetables, into the anti-carcinogenic compound, sulphoraphane [68]. This study demonstrates how components in the normal diet (possibly emerging prebiotics) and engineered probiotics can be harnessed simultaneously to render a targeted effect on the host through modulating the functional output of the microbiome.

4. Phage-Mediated Modulation of Gut Microbiome

While the use of probiotics to promote or compete with natural resident bacteria of the gut is actively being explored, the use of genetic tools to selectively remove a targeted population is also being investigated. The discovery of bacteriophages in the earlier advent of phage therapy [69], combined with a more recent discovery of the clustered regularly interspaced short palindromic repeats (CRISPR) and the CRISPR-associated nuclease 9 (Cas9) system, has led to an array of strategies to manipulate the gut microbiome with precision [70,71]. For instance, engineered phage (with the CRISPR-Cas9 system) can be employed to target pathogenic bacteria, or remove the population of bacteria that aids pathogenic bacterial growth, thereby fine-tuning and restoring the balance of the gut microbiome. In recent years, several groups have attempted to show the effectiveness of using bacteriophages to eliminate specific populations of bacteria without disrupting other commensals. For example, virulent bacteriophages were tested against adherent invasive Escherichia coli (AIEC) found on the mucosa of the ileum of patients with Crohn’s disease. A considerable reduction of AIEC was observed after phage treatment in vivo, demonstrating a microbiota composition change through pathogen removal [72]. Similarly, the effectiveness of the oral administration of a bacteriophage cocktail showed that phage treatment yielded greater clearance of the pathogen compared to the use of ampicillin in treating Shigella-challenged mice [73]. The treatment was also specific in removing the target population without affecting other commensals [73]. Other studies have demonstrated the specificity of bacteriophages in altering the microbiota composition through pathogen removal and reduction, without perturbation to other commensal populations, include the targeting of Enterococcus faecalis [74], Listeria monocytogenes [75], and Clostridium difficile [76].
While phage therapy is used for the population removal of pathogens, CRISPR/Cas9 can be used to manipulate and differentiate genetically heterogeneous bacteria, even of the same species. Currently, one of the pressing clinical issues is antibiotic resistance and the rise of multi-drug resistant (MDR) bacteria that can result in superinfections [69]. Unlike conventional drugs, the CRISPR/Cas9 system targets specific bacteria at the gene level to selectively remove pathogens, making this tool a potential antimicrobial adjuvant to improve antibiotic treatment. This concept was demonstrated by Citorik et al. [77], where CRISPR/Cas9 was delivered using bacteriophages, targeting the ndm-1 gene, which codes for the broad-spectrum carbapenemase, New-Delhi metallo-β-lactamase. Ndm-1 targeting CRISPR/Cas9 specifically eliminated E. coli harbouring the gene without affecting wild-type, or other, E. coli strains present in a synthetic consortium of microbes [77]. Another notable example is the re-sensitisation of bacteria to antibiotics and immunisation of bacteria to incoming plasmids conferring antibiotic resistance using temperate phages. Specifically, Yosef et al. used CRISPR/Cas9 to target ndm-1 and ctx-M-15, which expresses a broad-spectrum beta-lactamase, and effectively selected the transduced bacteria that were antibiotic-sensitive [78]. Though these studies are still mostly in vitro, they pave the way to advance future work using CRISPR/Cas9 to manipulate the gut microbiome by discriminating at the gene level to change the characteristics and functional output of the gut microbiome for therapeutic applications.
While phage therapy and the CRISPR/Cas9 system hold promise for future applications, there are several concerns for each of them. One of the concerns for phage therapy is the unintended activation of host immune responses. Phage therapy relies on the lysis of the bacterial host, which releases unwanted cellular components, such as endotoxins, into the gut lumen environment that may trigger an immune response [79]. Though the probability of immune activation by phages cannot be eliminated, clinical trials using phage therapy has thus far shown to be well-tolerated without any adverse side effects. Interestingly, a study demonstrated that the use of carbapenems caused more endotoxins than phages [80]; emerging alternatives to circumvent this issue include engineering lysis-deficient phages [81]. For CRISPR/Cas9, some concerns, including off-target gene targeting and escape rates, are summarised in a review, along with recent works [77,82,83]. These concerns can be addressed progressively as areas in CRISPR/Cas9 research, such as optimising gene multiplexing and continuing the development of increased specificity and efficiency of CRISPR/Cas9 in target organisms. As phage therapy and the CRISPR/Cas9 system gain momentum as tools for microbiome manipulation, more in vivo work and clinical experimentation are needed to ascertain the various effects of using such viral-based approaches.

5. Conclusions and Future Outlook

In this review, we focused on recent work showing how the gut microbiome can be directly manipulated in a targeted manner in situ. Population modulation through composition alteration can be achieved though the administration of targeted modifiers, such as prebiotics, probiotics, and phages, with measurable improvements in health markers for various gut disorders and metabolic conditions. Targeted manipulation of known pathways in the gut microbiome can also be achieved to alter the functional metabolic output of the microbiome. Manipulating the gut microbiome fundamentally relies on the extent of knowledge about the microbiota, the multitude of metabolic pathways present in the GIT, and the complex interactions with the host. As the therapeutic applications of gut microbiome manipulation are still in the incipient phase, current efforts to advance the fundamental understanding of the gut microbiome and its host interactions should be fortified and strongly encouraged to actualise its therapeutic and clinical potential.

Acknowledgments

We thank Carol Han Ping for her comments on this manuscript. This work was supported by the Synthetic Biology Initiative of the National University of Singapore (DPRT/943/09/14), Summit Research Program of the National University Health System (NUHSRO/2016/053/SRP/05), the Ministry of Defence of Singapore (MINDEF, RE2016-074), the Singapore Ministry of Education (MOE/2014/T2/2/128), and the U.S. Defense Threat Reduction Agency (HDTRA1-13- 0037).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef] [PubMed]
  2. Peterson, J.; Garges, S.; Giovanni, M.; McInnes, P.; Wang, L.; Schloss, J.A.; Bonazzi, V.; McEwen, J.E.; Wetterstrand, K.A.; Deal, C. The NIH human microbiome project. Genome Res. 2009, 19, 2317–2323. [Google Scholar] [PubMed]
  3. Bielecka, M. Probiotics in Food. In Chemical and Functional Properties of Food Components, 3rd ed.; Sikorski, Z., Ed.; CRC Press: Boca Raton, FL, USA, 2007; pp. 413–426. [Google Scholar] [CrossRef]
  4. Sender, R.; Fuchs, S.; Milo, R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016, 14, 1–14. [Google Scholar] [CrossRef] [PubMed]
  5. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kamada, N.; Chen, G.Y.; Inohara, N.; Núñez, G. Control of pathogens and pathobionts by the gut microbiota. Nat. Immunol. 2013, 14, 685–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. David, L.A.; Materna, A.C.; Friedman, J.; Campos-Baptista, M.I.; Blackburn, M.C.; Perrotta, A.; Erdman, S.E.; Alm, E.J. Host lifestyle affects human microbiota on daily timescales. Genome Biol. 2015, 15, 1–15. [Google Scholar] [CrossRef] [PubMed]
  8. Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the microbiota and the immune system. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [PubMed]
  9. Cho, I.; Blaser, M.J. The human microbiome: At the interface of health and disease. Nat. Rev. Genet. 2012, 13, 260–270. [Google Scholar] [CrossRef] [PubMed]
  10. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed]
  11. Chassard, C.; Lacroix, C. Carbohydrates and the human gut microbiota. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 453–460. [Google Scholar] [CrossRef] [PubMed]
  12. Anhe, F.F.; Roy, D.; Pilon, G.; Dudonne, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E.; et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883. [Google Scholar] [CrossRef] [PubMed]
  13. Larrosa, M.; Yañéz-Gascón, M.J.; Selma, M.V.; González-Sarrías, A.; Toti, S.; Cerón, J.J.; Tomás-Barberán, F.; Dolara, P.; Espín, J.C. Effect of a low dose of dietary resveratrol on colon microbiota, inflammation and tissue damage in a DSS-induced colitis rat model. J. Agric. Food Chem. 2009, 57, 2211–2220. [Google Scholar] [CrossRef] [PubMed]
  14. Chaplin, A.; Parra, P.; Serra, F.; Palou, A. Conjugated linoleic acid supplementation under a high-fat diet modulates stomach protein expression and intestinal microbiota in adult mice. PLoS ONE 2015, 10, e0125091. [Google Scholar] [CrossRef] [PubMed]
  15. Sawin, E.A.; De Wolfe, T.J.; Aktas, B.; Stroup, B.M.; Murali, S.G.; Steele, J.L.; Ney, D.M. Glycomacropeptide is a prebiotic that reduces Desulfovibrio bacteria, increases cecal short-chain fatty acids, and is anti-inflammatory in mice. Am. J. Physiol. Liver Physiol. 2015, 309, G590–G601. [Google Scholar] [CrossRef] [PubMed]
  16. Vieira-Silva, S.; Falony, G.; Darzi, Y.; Lima-Mendez, G.; Garcia Yunta, R.; Okuda, S.; Vandeputte, D.; Valles-Colomer, M.; Hildebrand, F.; Chaffron, S.; et al. Species-function relationships shape ecological properties of the human gut microbiome. Nat. Microbiol. 2016, 1, 1–8. [Google Scholar] [CrossRef] [PubMed]
  17. Pokusaeva, K.; Fitzgerald, G.F.; Van Sinderen, D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr. 2011, 6, 285–306. [Google Scholar] [CrossRef] [PubMed]
  18. Neyrinck, A.M.; Possemiers, S.; Druart, C.; van de Wiele, T.; de Backer, F.; Cani, P.D.; Larondelle, Y.; Delzenne, N.M. Prebiotic effects of wheat Arabinoxylan related to the increase in Bifidobacteria, Roseburia and bacteroides/Prevotella in diet-induced obese mice. PLoS ONE 2011, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Gonai, M.; Shigehisa, A.; Kigawa, I.; Kurasaki, K.; Chonan, O.; Matsuki, T.; Yoshida, Y.; Aida, M.; Hamano, K.; Terauchi, Y. Galacto-oligosaccharides ameliorate dysbiotic Bifidobacteriaceae decline in Japanese patients with type 2 diabetes. Benef. Microbes 2017, 8, 705–716. [Google Scholar] [CrossRef] [PubMed]
  20. Salazar, N.; Dewulf, E.M.; Neyrinck, A.M.; Bindels, L.B.; Cani, P.D.; Mahillon, J.; de Vos, W.M.; Thissen, J.P.; Gueimonde, M.; de los Reyes-Gavilán, C.G.; et al. Inulin-type fructans modulate intestinal Bifidobacterium species populations and decrease fecal short-chain fatty acids in obese women. Clin. Nutr. 2015, 34, 501–507. [Google Scholar] [CrossRef] [PubMed]
  21. Vulevic, J.; Juric, A.; Walton, G.E.; Claus, S.P.; Tzortzis, G.; Toward, R.E.; Gibson, G.R. Influence of galacto-oligosaccharide mixture (B-GOS) on gut microbiota, immune parameters and metabonomics in elderly persons. Br. J. Nutr. 2015, 114, 586–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Krishna, G.; Divyashri, G.; Prapulla, S.G.; Muralidhara. A Combination Supplement of Fructo- and Xylo-Oligosaccharides Significantly Abrogates Oxidative Impairments and Neurotoxicity in Maternal/Fetal Milieu Following Gestational Exposure to Acrylamide in Rat. Neurochem. Res. 2015, 40, 1904–1918. [Google Scholar] [CrossRef] [PubMed]
  23. Autran, C.A.; Schoterman, M.H.C.; Jantscher-Krenn, E.; Kamerling, J.P.; Bode, L. Sialylated galacto-oligosaccharides and 2′-fucosyllactose reduce necrotising enterocolitis in neonatal rats. Br. J. Nutr. 2016, 116, 294–299. [Google Scholar] [CrossRef] [PubMed]
  24. Intanon, M.; Arreola, S.L.; Pham, N.H.; Kneifel, W.; Haltrich, D.; Nguyen, T.H. Nature and biosynthesis of galacto-oligosaccharides related to oligosaccharides in human breast milk. FEMS Microbiol. Lett. 2014, 353, 89–97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Rivière, A.; Selak, M.; Lantin, D.; Leroy, F.; De Vuyst, L. Bifidobacteria and butyrate-producing colon bacteria: Importance and strategies for their stimulation in the human gut. Front. Microbiol. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
  27. Rios-Covian, D.; Gueimonde, M.; Duncan, S.H.; Flint, H.J.; De Los Reyes-Gavilan, C.G. Enhanced butyrate formation by cross-feeding between Faecalibacterium prausnitzii and Bifidobacterium adolescentis. FEMS Microbiol. Lett. 2015, 362, 1–7. [Google Scholar] [CrossRef] [PubMed]
  28. Everard, A.; Lazarevic, V.; Derrien, M.; Girard, M.; Muccioli, G.M.; Neyrinck, A.M.; Possemiers, S.; Van Holle, A.; François, P.; De Vos, W.M.; et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 2011, 60, 2775–2786. [Google Scholar] [CrossRef] [PubMed]
  29. Quévrain, E.; Maubert, M.A.; Michon, C.; Chain, F.; Marquant, R.; Tailhades, J.; Miquel, S.; Carlier, L.; Bermúdez-Humarán, L.G.; Pigneur, B.; et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016, 65, 415–425. [Google Scholar] [CrossRef] [PubMed]
  30. Ramirez-Farias, C.; Slezak, K.; Fuller, Z.; Duncan, A.; Holtrop, G.; Loius, P. Effect of inulin on the human gut microbiota: Stimulation of Bifidobacterium adolenscentis and Faecalibacterium prausnitzii. Br. J. Nutr. 2009, 101, 541–550. [Google Scholar] [CrossRef] [PubMed]
  31. Ohashi, Y.; Sumitani, K.; Tokunaga, M.; Ishihara, N.; Okubo, T.; Fujisawa, T. Consumption of partially hydrolysed guar gum stimulates Bifidobacteria and butyrate-producing bacteria in the human large intestine. Benef. Microbes 2015, 6, 451–455. [Google Scholar] [CrossRef] [PubMed]
  32. Halfvarson, J.; Brislawn, C.J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W.A.; Bramer, L.M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A.; et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol. 2017, 2, 1–7. [Google Scholar] [CrossRef] [PubMed]
  33. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermudez-Humaran, L.G.; Gratadoux, J.-J.; Blugeon, S.; Bridonneau, C.; Furet, J.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. PNAS 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [PubMed]
  34. Machiels, K.; Joossens, M.; Sabino, J.; De Preter, V.; Arijs, I.; Eeckhaut, V.; Ballet, V.; Claes, K.; Van Immerseel, F.; Verbeke, K.; et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014, 63, 1275–1283. [Google Scholar] [CrossRef] [PubMed]
  35. Davis, L.M.G.; Martínez, I.; Walter, J.; Goin, C.; Hutkins, R.W. Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS ONE 2011, 6, e25200. [Google Scholar] [CrossRef] [PubMed]
  36. Lampe, J.W.; Navarro, S.L.; Hullar, M.A.J.; Shojaie, A. Inter-individual differences in response to dietary intervention: Integrating omics platforms towards personalised dietary recommendations. Proc. Nutr. Soc. 2013, 72, 207–218. [Google Scholar] [CrossRef] [PubMed]
  37. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document: The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  38. Lim, S.M.; Kim, D.H. Bifidobacterium adolescentis IM38 ameliorates high-fat diet–induced colitis in mice by inhibiting NF-κB activation and lipopolysaccharide production by gut microbiota. Nutr. Res. 2017, 41, 86–96. [Google Scholar] [CrossRef] [PubMed]
  39. Turroni, F.; Milani, C.; Duranti, S.; Mancabelli, L.; Mangifesta, M.; Viappiani, A.; Lugli, G.A.; Ferrario, C.; Gioiosa, L.; Ferrarini, A.; et al. Deciphering bifidobacterial-mediated metabolic interactions and their impact on gut microbiota by a multi-omics approach. ISME J. 2016, 10, 1656–1668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Krych; Nielsen, D.S.; Hansen, A.K.; Hansen, C.H.F. Gut microbial markers are associated with diabetes onset, regulatory imbalance, and IFN-γ level in NOD Mice. Gut Microbes 2015, 6, 101–109. [Google Scholar] [CrossRef] [Green Version]
  41. Ji, Y.S.; Kim, H.N.; Park, H.J.; Lee, J.E.; Yeo, S.Y.; Yang, J.S.; Park, S.Y.; Yoon, H.S.; Cho, G.S.; Franz, C.M.A.P.; et al. Modulation of the murine microbiome with a concomitant anti-obesity effect by Lactobacillus rhamnosus GG and Lactobacillus sakei NR28. Benef. Microbes 2012, 3, 13–22. [Google Scholar] [CrossRef] [PubMed]
  42. Sonnenborn, U.; Schulze, J. The non-pathogenic Escherichia coli strain Nissle 1917 – features of a versatile probiotic. Microb. Ecol. Health Dis. 2009, 21, 122–158. [Google Scholar] [CrossRef]
  43. Deriu, E.; Liu, J.Z.; Pezeshiki, M.; Edwards, R.A.; Ochoa, R.J.; Contreras, H.; Libby, S.J.; Fang, F.C.; Raffatellu, M. Probiotic bacteria reduce Salmonella typhimurium intestinal colonization by competing for iron. Cell Host Microbe 2014, 14, 26–37. [Google Scholar] [CrossRef] [PubMed]
  44. Rund, S.A.; Rohde, H.; Sonnenborn, U.; Oelschlaeger, T.A. Antagonistic effects of probiotic Escherichia coli Nissle 1917 on EHEC strains of serotype O104: H4 and O157: H7. Int. J. Med. Microbiol. 2013, 303, 1–8. [Google Scholar] [CrossRef] [PubMed]
  45. Schierack, P.; Kleta, S.; Tedin, K.; Babila, J.T.; Oswald, S.; Oelschlaeger, T.A.; Hiemann, R.; Paetzold, S.; Wieler, L.H. E. coli Nissle 1917 affects Salmonella adhesion to porcine intestinal epithelial cells. PLoS ONE 2011, 6, e14712. [Google Scholar] [CrossRef] [PubMed]
  46. Chiu, W.-C.; Huang, Y.-L.; Chen, Y.-L.; Peng, H.-C.; Liao, W.-H.; Chuang, H.-L.; Chen, J.-R.; Yang, S.-C. Synbiotics reduce ethanol-induced hepatic steatosis and inflammation by improving intestinal permeability and microbiota in rats. Food Funct. 2015, 6, 1692–1700. [Google Scholar] [CrossRef] [PubMed]
  47. Ishikawa, H.; Matsumoto, S.; Ohashi, Y.; Imaoka, A.; Setoyama, H.; Umesaki, Y.; Tanaka, R.; Otani, T. Beneficial effects of probiotic Bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: A randomized controlled study. Digestion 2011, 84, 128–133. [Google Scholar] [CrossRef] [PubMed]
  48. Ritchie, M.L.; Romanuk, T.N. A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS ONE 2012, 7, e34938. [Google Scholar] [CrossRef] [PubMed]
  49. McLoughlin, R.F.; Berthon, B.S.; Jensen, M.E.; Baines, K.J.; Wood, L.G. Short-chain fatty acids, prebiotics, synbiotics, and systemic inflammation: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2017, ajcn156265. [Google Scholar] [CrossRef] [PubMed]
  50. Kalliomäki, M.; Antoine, J.-M.; Herz, U.; Rijkers, G.T.; Wells, J.M.; Mercenier, A. Guidance for Substantiating the Evidence for Beneficial Effects of Probiotics: Prevention and Management of Allergic Diseases by Probiotics–3. J. Nutr. 2010, 140, 713S–721S. [Google Scholar] [CrossRef] [PubMed]
  51. Bleich, A.; Sundberg, J.P.; Smoczek, A.; Von Wasielewski, R.; De Buhr, M.F.; Janus, L.M.; Julga, G.; Ukena, S.N.; Hedrich, H.; Gunzer, F. Sensitivity to Escherichia coli Nissle 1917 in mice is dependent on environment and genetic background. Int. J. Exp. Pathol. 2008, 89, 45–54. [Google Scholar] [CrossRef] [PubMed]
  52. Walker, A.W.; Duncan, S.H.; Louis, P.; Flint, H.J. Phylogeny, culturing, and metagenomics of the human gut microbiota. Trends Microbiol. 2014, 22, 267–274. [Google Scholar] [CrossRef] [PubMed]
  53. Jacobsen, C.N.; Nielsen, V.R.; Hayford, A.E.; Møller, P.L.; Michaelsen, K.F.; Paerregaard, A.; Sandström, B.; Tvede, M.; Jakobsen, M. Screening of probiotic activities of forty-seven strains of Lactobacillus spp. by in vitro techniques and evaluation of the colonization ability of five selected strains in humans. Appl. Environ. Microbiol. 1999, 65, 4949–4956. [Google Scholar] [PubMed]
  54. Lagier, J.C.; Armougom, F.; Million, M.; Hugon, P.; Pagnier, I.; Robert, C.; Bittar, F.; Fournous, G.; Gimenez, G.; Maraninchi, M.; et al. Microbial culturomics: Paradigm shift in the human gut microbiome study. Clin. Microbiol. Infect. 2012, 18, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
  55. Goodman, A.L.; Kallstrom, G.; Faith, J.J.; Reyes, A.; Moore, A.; Dantas, G.; Gordon, J.I. Extensive personal human gut microbiota culture collections characterized and manipulated in gnotobiotic mice. Proc. Natl. Acad. Sci. USA 2011, 108, 6252–6257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Martín, R.; Miquel, S.; Benevides, L.; Bridonneau, C.; Robert, V.; Hudault, S.; Chain, F.; Berteau, O.; Azevedo, V.; Chatel, J.M.; et al. Functional characterization of novel Faecalibacterium prausnitzii strains isolated from healthy volunteers: A step forward in the use of F. prausnitzii as a next-generation probiotic. Front. Microbiol. 2017, 8, 1–13. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, Z.; Guo, L.; Zhang, Y.; Walzem, R.L.; Pendergast, J.S.; Printz, R.L.; Morris, L.C.; Matafonova, E.; Stien, X.; Kang, L.; et al. Incorporation of therapeutically modified bacteria into gut microbiota inhibits obesity. J. Clin. Investig. 2014, 124, 3391–3406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Duan, F.F.; Liu, J.H.; March, J.C. Engineered commensal bacteria reprogram intestinal cells into glucose-responsive insulin-secreting cells for the treatment of diabetes. Diabetes 2015, 64, 1794–1803. [Google Scholar] [CrossRef] [PubMed]
  59. Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef] [PubMed]
  60. Shigemori, S.; Watanabe, T.; Kudoh, K.; Ihara, M.; Nigar, S.; Yamamoto, Y.; Suda, Y.; Sato, T.; Kitazawa, H.; Shimosato, T. Oral delivery of Lactococcus lactis that secretes bioactive heme oxygenase-1 alleviates development of acute colitis in mice. Microb. Cell Fact. 2015, 14, 1–12. [Google Scholar] [CrossRef] [PubMed]
  61. Bermúdez-Humarán, L.G.; Motta, J.P.; Aubry, C.; Kharrat, P.; Rous-Martin, L.; Sallenave, J.M.; Deraison, C.; Vergnolle, N.; Langella, P. Serine protease inhibitors protect better than IL-10 and TGF-β anti-inflammatory cytokines against mouse colitis when delivered by recombinant lactococci. Microb. Cell Fact. 2015, 14, 1–11. [Google Scholar] [CrossRef] [PubMed]
  62. Martín, R.; Chain, F.; Miquel, S.; Natividad, J.M.; Sokol, H.; Verdu, E.F.; Langella, P.; Bermúdez-Humarán, L.G. Effects in the use of a genetically engineered strain of Lactococcus lactis delivering in situ IL-10 as a therapy to treat low-grade colon inflammation. Hum. Vaccines Immunother. 2014, 10, 1611–1621. [Google Scholar] [CrossRef] [PubMed]
  63. Saeidi, N.; Wong, C.K.; Lo, T.; Nguyen, H.X.; Ling, H.; Leong, S.S.J.; Poh, C.L.; Chang, M.W. Engineering microbes to sense and eradicate Pseudomonas aeruginosa, a human pathogen. Mol. Syst. Biol. 2011, 7, 521. [Google Scholar] [CrossRef] [PubMed]
  64. Hwang, I.Y.; Koh, E.; Wong, A.; March, J.C.; Bentley, W.E.; Lee, Y.S.; Chang, M.W. Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models. Nat. Commun. 2017, 8, 15028. [Google Scholar] [CrossRef] [PubMed]
  65. Mao, N.; Cubillos-Ruiz, A.; Cameron, D.E.; Collins, J.J. Probiotic strains detect and suppress cholera in mice. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
  66. Mimee, M.; Tucker, A.C.; Voigt, C.A.; Lu, T.K. Programming a human commensal bacterium, Bacteroides thetaiotaomicron, to sense and respond to stimuli in the murine gut microbiota. Cell Syst. 2015, 1, 62–71. [Google Scholar] [CrossRef] [PubMed]
  67. Lim, B.; Zimmermann, M.; Barry, N.A.; Goodman, A.L. Engineered regulatory systems modulate gene expression of human commensals in the gut. Cell 2017, 169, 547–558. [Google Scholar] [CrossRef] [PubMed]
  68. Ho, C.L.; Tan, H.Q.; Chua, K.J.; Kang, A.; Lim, K.H.; Ling, K.L.; Yew, W.S.; Lee, Y.S.; Thiery, J.P.; Chang, M.W. Engineered commensal microbes for diet-mediated colorectal-cancer chemoprevention. Nat. Biomed. Eng. 2018, 2, 27–37. [Google Scholar] [CrossRef]
  69. Abedon, S.T.; Kuhl, S.J.; Blasdel, B.G.; Kutter, E.M. Phage treatment of human infections. Bacteriophage 2011, 1, 66–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Hsu, P.D.; Lander, E.S.; Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 2014, 157, 1262–1278. [Google Scholar] [CrossRef] [PubMed]
  71. Sander, J.D.; Joung, J.K. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat. Biotechnol. 2014, 32, 347–350. [Google Scholar] [CrossRef] [PubMed]
  72. Galtier, M.; De Sordi, L.; Sivignon, A.; de Vallée, A.; Maura, D.; Neut, C.; Rahmouni, O.; Wannerberger, K.; Darfeuille-Michaud, A.; Desreumaux, P.; et al. Bacteriophages targeting adherent invasive Escherichia coli strains as a promising new treatment for Crohn’s disease. J. Crohn’s Colitis 2017, 11, 840–847. [Google Scholar] [CrossRef] [PubMed]
  73. Mai, V.; Ukhanova, M.; Reinhard, M.K.; Li, M.; Sulakvelidze, A. Bacteriophage administration significantly reduces Shigella colonization and shedding by Shigella-challenged mice without deleterious side effects and distortions in the gut microbiota. Bacteriophage 2015, 5, e1088124. [Google Scholar] [CrossRef] [PubMed]
  74. Cheng, M.; Liang, J.; Zhang, Y.; Hu, L.; Gong, P.; Cai, R.; Zhang, L.; Zhang, H.; Ge, J.; Ji, Y.; et al. The bacteriophage EF-P29 efficiently protects against lethal vancomycin-resistant Enterococcus faecalis and alleviates gut microbiota imbalance in a murine bacteremia model. Front. Microbiol. 2017, 8, 837. [Google Scholar] [CrossRef] [PubMed]
  75. Mai, V.; Ukhanova, M.; Visone, L.; Abuladze, T.; Sulakvelidze, A. Bacteriophage administration reduces the concentration of Listeria monocytogenes in the gastrointestinal tract and its translocation to spleen and liver in experimentally infectedmice. Int. J. Microbiol. 2010, 2010. [Google Scholar] [CrossRef] [PubMed]
  76. Meader, E.; Mayer, M.J.; Steverding, D.; Carding, S.R.; Narbad, A. Evaluation of bacteriophage therapy to control Clostridium difficile and toxin production in an invitro human colon model system. Anaerobe 2013, 22, 25–30. [Google Scholar] [CrossRef] [PubMed]
  77. Citorik, R.J.; Mimee, M.; Lu, T.K. Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases. Nat. Biotechnol. 2014, 32, 1141–1145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Yosef, I.; Manor, M.; Kiro, R.; Qimron, U. Temperate and lytic bacteriophages programmed to sensitize and kill antibiotic-resistant bacteria. Proc. Natl. Acad. Sci. USA 2015, 112, 7267–7272. [Google Scholar] [CrossRef] [PubMed]
  79. Cisek, A.A.; Dąbrowska, I.; Gregorczyk, K.P.; Wyżewski, Z. Phage therapy in bacterial infections treatment: One hundred years after the discovery of bacteriophages. Curr. Microbiol. 2017, 74, 277–283. [Google Scholar] [CrossRef] [PubMed]
  80. Dufour, N.; Delattre, R.; Ricard, J.D.; Debarbieux, L. The lysis of pathogenic Escherichia coli by bacteriophages releases less endotoxin than by β-lactams. Clin. Infect. Dis. 2017, 64, 1582–1588. [Google Scholar] [CrossRef] [PubMed]
  81. Paul, V.; Sundarrajan, S.; Rajagopalan, S.; Hariharan, S.; Kempashanaiah, N.; Padmanabhan, S.; Sriram, B.; Ramachandran, J. Lysis-deficient phages as novel therapeutic agents for controlling bacterial infection. BMC Microbiol. 2011, 11, 195. [Google Scholar] [CrossRef] [PubMed]
  82. Bikard, D.; Euler, C.W.; Jiang, W.; Nussenzweig, P.M.; Goldberg, G.W.; Duportet, X.; Fischetti, V.A.; Marraffini, L.A. Exploiting CRISPR-cas nucleases to produce sequence-specific antimicrobials. Nat. Biotechnol. 2014, 32, 1146–1150. [Google Scholar] [CrossRef] [PubMed]
  83. Fagen, J.R.; Collias, D.; Singh, A.K.; Beisel, C.L. Advancing the design and delivery of CRISPR antimicrobials. Curr. Opin. Biomed. Eng. 2017, 4, 57–64. [Google Scholar] [CrossRef]
Figure 1. Overview of targeted methods to manipulate the gut microbiome. (a) Administration of targeted prebiotics to stimulate the growth of beneficial microbes; (b) use of targeted probiotics and engineered probiotics to eliminate pathogens or directly change the functional output of the gut microbiome; and (c) use of bacteriophages to eliminate specific species of pathogens or target pathogens with certain genes.
Figure 1. Overview of targeted methods to manipulate the gut microbiome. (a) Administration of targeted prebiotics to stimulate the growth of beneficial microbes; (b) use of targeted probiotics and engineered probiotics to eliminate pathogens or directly change the functional output of the gut microbiome; and (c) use of bacteriophages to eliminate specific species of pathogens or target pathogens with certain genes.
Genes 09 00351 g001
Figure 2. Cross-feeding effect between Bifidobacterium and butyrate-producing bacteria. Bifidobacterium utilises supplemented prebiotics, which stimulates their growth. Acetate produced by Bifidobacterium becomes a carbon source for butyrate-producing microbes, stimulating their growth and butyrate-producing activities and, in turn, modulating the microbiome function and improving gut health.
Figure 2. Cross-feeding effect between Bifidobacterium and butyrate-producing bacteria. Bifidobacterium utilises supplemented prebiotics, which stimulates their growth. Acetate produced by Bifidobacterium becomes a carbon source for butyrate-producing microbes, stimulating their growth and butyrate-producing activities and, in turn, modulating the microbiome function and improving gut health.
Genes 09 00351 g002

Share and Cite

MDPI and ACS Style

Lee, H.L.; Shen, H.; Hwang, I.Y.; Ling, H.; Yew, W.S.; Lee, Y.S.; Chang, M.W. Targeted Approaches for In Situ Gut Microbiome Manipulation. Genes 2018, 9, 351. https://doi.org/10.3390/genes9070351

AMA Style

Lee HL, Shen H, Hwang IY, Ling H, Yew WS, Lee YS, Chang MW. Targeted Approaches for In Situ Gut Microbiome Manipulation. Genes. 2018; 9(7):351. https://doi.org/10.3390/genes9070351

Chicago/Turabian Style

Lee, Hui Ling, Haosheng Shen, In Young Hwang, Hua Ling, Wen Shan Yew, Yung Seng Lee, and Matthew Wook Chang. 2018. "Targeted Approaches for In Situ Gut Microbiome Manipulation" Genes 9, no. 7: 351. https://doi.org/10.3390/genes9070351

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop