Skip to main content

MINI REVIEW article

Front. Microbiol., 07 August 2019
Sec. Microbial Immunology
This article is part of the Research Topic Understanding and Exploiting Host-Commensal Interactions to Combat Pathogens View all 17 articles

Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis

  • 1Infectious Diseases Department, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
  • 2National Institute of Infectious Diseases “Prof. Dr. Matei Balş”, Bucharest, Romania

The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.

Introduction

The intestinal barrier defines the morpho-functional unit responsible for the defense of the intestinal mucosa and consists of the intestinal microbiota, intestinal epithelial cells (IECs) and mucosal immunity tightly linked through a complex network of cytokines, antimicrobial peptides (AMPs), metabolic products, and numerous regulatory molecules (Meng et al., 2017). Given that the intestinal mucosa is the largest body surface at risk of infectious threats, the anatomic and functional homeostasis of the intestinal barrier is a key step in the anti-infectious defense of the human organism.

The intestinal microbiota represents the first defense line of the intestinal barrier. The microbiota entails millions of microogranisms colonizing the gastrointestinal tract most of which are bacteria. This large number of microorganisms withstand the unfavorable intestinal habitat thanks to their symbiotic relationships with the human organism. These symbiotic host-commensal relationships develop after birth and enable the metabolic, immune and antiinfectious processes through which the microbiota contributes to gut homeostasis (O’Hara and Shanahan, 2006). The structural and functional stability of commensal populations is regulated through numerous signaling molecules (quorum sensing) and cellular regulators (miRNAs) as well as through other physiologic and pathologic factors. Qualitative or quantitative alterations of this microbial community broadly defined as dysbiosis impair the relationships between the host and commensal species, modify the balance between commensals and pathogens, decrease the intestinal barrier protection and favor infectious pathogens (McDonald et al., 2016). Consequently, the microbiota loses its anti-infectious role and becomes the weak link responsible for persistent infections.

The article discusses gut barrier defense mechanisms, the key anti-infectious role of microbiota inside of the gut barrier and the impact of dysbiosis in the life-threatening infections.

The Gut Barrier and Antibacterial Defense Mechanisms

Commensal Flora – The First Line of Defense

The intestinal microbiota encompasses all microbial species populating the gastrointestinal tract. Molecular techniques in healthy individuals have revealed a diverse ecosystem, containing nine bacterial phyla, out of which four are dominant: Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria (Rajilić-Stojanović et al., 2007). The structure of the microbiota is gradually delineated after the age of three through symbiotic relationships with the organism and ensures the intestinal dominance of specific commensals (“symbionts”) belonging to the Firmicutes and Bacteroidetes phylum or Bifidobacteriales order. Symbionts will compete with pathogens or potential pathogenic germs (“pathobionts”) for pre-existent intestinal niches (“niche competition”) as well as for intestinal nutrients (“nutritive competition”). This competition ensures the structural stability of the microbiota and is referred to as “colonization resistance.” Colonization resistance employs a network of specific molecules with a critical anti-infectious roles (Sassone-Corsi and Raffatellu, 2015; Sorbara and Pamer, 2019). Among these are molecules with important metabolic and antimicrobial roles such as short chain fatty acids (SCFAs) and bacteriocins.

Short chain fatty acids are end products of the anaerobic fermentation of intestinal microbiota and the major energy source for colonocytes (Roediger, 1980). SCFAs effects are mediated by G protein-coupled receptors (GPRs) (GPR41, GPR43, and GPR109A) expressed on immune cells and a variety of tissues including gut epithelial cells. The predominant SCFAs are present at high mM levels in the colon (butyrate), entero-hepatic circulation (propionate), and systemic circulation (acetate) and are responsible for epithelial protection and regulation of the inflammatory intestinal response, reviewed in Donohoe et al. (2011) and Fukuda et al. (2011). SCFAs ensure a low antibacterial pH around colonic cells, favor mucus synthesis, and contribute to IECs integrity through the upregulation of tight junction proteins, stabilization of HIF transcription factor, IL-18 release, and NLR pyrin domain 3 (NLRP3) inflammasome modulation (Wang et al., 2012; Kelly et al., 2015; Macia et al., 2015; Feng Y. et al., 2018). At the same time SCFAs contribute to the antibacterial defense against pathogenic species through neutrophil recruitment, the release of cytokines and AMPs while also inducing the intestinal immunotolerant response against commensal species as further discussed.

Gut bacteria also, particularly Firmicutes, Proteobacteria, and Bacteroidetes, synthesize multiple microbicidal molecules (bacteriocins) with a broad spectrum of activity (Todorov et al., 2014; Drissi et al., 2015). Bacteriocin-producing strains such as lactic acid bacteria are commonly used as probiotics in the food industry (Parada et al., 2007). Certain bacteriocins (nissin, pediocin) have been approved for oral or topical use and others are studied as oral alternatives to antibiotics (reviewed in Cavera et al., 2015). Nevertheless, the oral use of bacteriocins is still uncertain due to a lack of data on their action mechanisms as well as on their efficiency, cytotoxicity, stability, and immunogenicity.

Dysbiosis modifies not only the balance between commensals and pathogens but also the release of antimicrobial molecules. In turn this disturbs the process of colonization resistance and allows the invasion of the intestinal epithelium by various pathogens.

The Intestinal Epithelium – The Second Anti-infectious Defense Line

The intestinal epithelium has its own defense mechanisms, both structural and functional. The intestinal epithelial monolayer includes several subsets of IECs united through tight apical junctions, externally covered with a mucus layer. IECs come into direct contact with the lamina propria and immune cells (Goto and Ivanov, 2013). All intestinal epithelial lineages arise from HOPX-quiescent stem cells with immunoregulatory and tumor suppressing properties (Takeda et al., 2011). IECs include entero-absorptive enterocytes, mucus secreting goblet cells, antigen-sampling cells (M cells), and Paneth cells, each exhibiting specific surface receptors.

The IECs express specific receptors namely “pattern recognition receptors” (PRRs) such as Nod-like receptors (NLRs), toll-like receptors (TLRs) and other PRR families located on the cell membrane or in the cytoplasm which recognize specific microbial-associated molecular patterns (MAMP). Once the activation of these receptors induces cytoplasmic signal transduction cascades and further promotes the NF-kappaB (NF-kB) pathway along with other cellular transcription factors, inflammasome activation and pro-inflammatory cytokines (IL-17, IL-18, IL-22) (Hirota et al., 2011; Jones and Neish, 2011; Levy et al., 2015). The IECs and particularly Paneth cells residing at the bottom of the intestinal crypts release AMPs (human-cathelicidin, defensins) with microbicidal, immunomodulatory and wound-healing properties. Thus IECs further contribute along with microbial colonization resistance to the stabilization of the gut barrier. Therefore Paneth cell MyD88 expression is an essential mechanism for the restriction of intestinal translocation and penetration by enteric commensals or pathogens (Vaishnava et al., 2008).

Short chain fatty acids and GPR43/109A stimulation also protects epithelial integrity via inflammasome activation and epigenetic immunomodulation of FoxP3+ regulatory T cells (Treg) proliferation as shown below (Macia et al., 2015).

Dysbiosis induced by infectious or non-infectious causes (ischemia, inflammation, tumors, or various treatments) favors the distruption of the intestinal epitelium (Kitajima et al., 1999; Nagpal and Yadav, 2017; Stewart A. S. et al., 2017). The injured epithelium permits the translocation of various intestinal bacteria and of their toxins including endotoxins (lipopolysaccharide, LPS). In turn these translocations lead to bacteraemia, endotoxemia and life-threatening infections (sepsis, primitive peritonitis, portal encephalopathy in cirrhotic patients, or immune activation in human immunodeficiency virus (HIV) infection.

The Intestinal Immunity – The Third Anti-infectious Defense Line

The intestinal epithelium represents the largest epithelial surface in the human body. It connects with the intestinal ecosystem containing commensal and pathogenic species. Commensal germs need to be immunologically accepted, whereas intestinal pathogens must be eliminated. This differentiation requires the activation of an extremely well-coordinated and efficient immune system. Considering that intestinal commensals are present in the human body ever since birth they represent, in fact, the first contact between the immune system and the exterior environment. As such, this large and diverse microbiota will serve as one of the body’s defense mechanisms against the invasion of pathogenic germs. At the same time, the microbiota develops mutually beneficial relationships with the organism and together with the intestinal epithelium and gut-associated lymphoid tissue (GALT) the microbiota forms a complex intestinal barrier against infectious threats.

These symbiotic relationships are facilitated by the presence of PRRs and by the immunomodulatory capacity of the intestinal microbiota against the antigenic structures exposed by the intestinal microbiota.

The Role of Cellular Receptors

The intestinal epithelium expresses numerous types of receptors able to recognize different MAMPs expressed by microbial species and to convert them into gut signals for inflammatory cascades (Kim et al., 2004). Thus TLR 1,2,4,5,6 (extracellular sensors) and NLR1,2 and TLR9 (cytoplasmic sensors) are expressed on epithelial cells and act complementary, promoting both innate and adaptive immunity (Kim et al., 2004; Abreu et al., 2005). MAMPs are expressed by commensal species since birth and examples of MAMPs include the LPS found in the outer membrane of Gram-negative bacteria, lipoteichoic acid present on the gram positive bacteria wall, peptidoglycan, a component of the bacterial wall, flagellin, a component of intestinal flagellated bacteria, or release outer membrane vesicles that function as PRR ligands (Cañas et al., 2018). PRRs activation in the presence of MAMPs stimulates the NF-kB signal transduction pathway, induces pro-inflammatory interleukins and enables an innate immune response lastly maintaining a state of controlled inflammation (Hayashi et al., 2001; Bambou et al., 2004; Vora et al., 2004; Rhayat et al., 2019). In the absence of MAMPs or of specific receptors (germ free animals or genetic mutations of receptors) the organism fails to detect microbial antigens and does not mount adequate defense mechanisms (Abreu et al., 2005). Hence, it is considered that the rich and diverse commensal flora plays an active role in the proper development of the immune system since birth.

Antigen-presenting cells (APC) belonging to GALT such as macrophages and dendritic cells (DCs) also exhibit cell receptors which regulate numerous genes and modulate the release of NF-κB transcription factor, immunomodulatory cytokines and AMP. Depending on the type of activated receptor DCs may generate a Th1/Th17 pro-inflammatory response. Thus, to exemplify, the NLR2 induced by commensal bacteria appears to play a central role in the downregulation of the GALT inflammatory activity and the ability of DCs to induce a polarized CD4+Th1 response in mice and human experiments (Butler et al., 2007; Barreau et al., 2010).

By activating the IECs or APCs receptors, the commensal flora induces a tolerogenic response in DCs, characterized by the release of immunosuppressive cytokines (IL10, IL4, TGF-β), promotion of Treg cells and of CD4+Th2 phenotype in the periphery (Iwasaki and Kelsall, 1999; Stagg et al., 2003; Atarashi et al., 2013; Maharshak et al., 2013; Martin-Gallausiaux et al., 2018). The induction of mucosal immune tolerance protects the epithelia from a detrimental inflammatory immune response and contributes to the immune system’s maturation (Jung et al., 2019). Consequently any disturbance involving this process plays an essential role in intestinal inflammatory processes.

The activation of cellular receptors is intricately linked to the pro-inflammatory response of mesenteric DCs and is particularly involved in the maintenance of the Th17/Th1 response directed against pathogens (reviewed in Sorini et al., 2018). What is interesting is that DCs may activate Treg cells to certain antigens such as LPS of the commensal flora but not LPS of the pathogenic flora (Shirai et al., 2004). Thus the stimulation of mice with E. coli-derived LPS activates inflammatory mechanisms (IL-12 production, Th1 response) whereas the stimulation with LPS derived from Poprhyromonas gingivalis induces IL-4 production and a Th2 response (Pulendran et al., 2001). Similarly TLR recognize flagellin antigens of pathogenic Salmonella species and trigger an inflammatory response against them, while commensal species lacking this antigen do not induce an inflammatory response (Gewirtz et al., 2001).

All these aspects evince that cell receptors represent a filter for intestinal signals and play a deciding role in the induction of specific defense mechanisms adapted to the commensal flora. This role of cell receptors is indeed significant considering the density and variety of the intestinal bacteria. Dysbsiosis eliminates commensal species that trigger Treg-cell polarization and therefore leads to an excessive Th1 or Th2 response, further promoting an inflammatory or autoimmune response.

The Immunomodulatory Role of GALT

Gut-associated lymphoid tissue is the most important lymphatic network in humans and involves isolated or aggregated lymphoid follicles (Peyer’s Patches), intra-epithelial lymphocytes, macrophages, DCs, mesenteric ganglia, secretory IgA (sIgA) cells and lymphatics.

Peyer’s patches localized in the mucosa and submucosa of the small intestine are covered by a “follicle-associated epithelium” containing specialized “M cells”. These cells engulf and transport antigens from the intestinal lumen to intestinal DCs where T cell lymphocytes (LT) are primed (Alpan et al., 2001). LT subsequently return to the intestinal lymphatic compartment for their effector function. At the same time, M cells initiate mucosal sIgA production and humoral responses (Rios et al., 2016). Thus, Peyer’s patches directly mediate the interaction between the intestinal flora and the humoral or cellular immune response. Enteric pathogens are adapted to invade and destroy follicle-associated epithelium, especially M cells, further interfering with T-cell differentiation and the immune response; these germs adhere to the intestinal epithelium through different mechanisms (Phillips et al., 2000), invade the enterocytes (Kühbacher et al., 2018), or invade and destroy M cells (Clark et al., 1994; Autenrieth and Firsching, 1996; Penheiter et al., 1997; Corr et al., 2006).

Gut-associated lymphoid tissue also harbors a tolerogenic DCs population namely CD103+DCs involved in Foxp3 expression and intestinal conversion of naïve CD4+T cells into Treg cells. Commensal flora stimulates CD103+ DCs through the activation of specific receptors or through its own metabolic products such as SCFAs (Furusawa et al., 2013; Nastasi et al., 2015; Kaisar et al., 2017). Thus, butyrate stimulates intestinal DCs, ensures intestinal immune tolerance through IL-10 release (Liu et al., 2012) and T cell polarization toward Treg cells while maintaining the balance between the immunosuppressive IL-10-secreting CD4+T cells, IL-17-secreting Th17 cells, and the CD4+Th1 effector cells (Arpaia et al., 2013; Furusawa et al., 2013). Additionally, SCFAs attenuate the excessive inflammatory response induced by LPS-producing Gram negative bacteria (Cox et al., 2009).

The dysregulation of the Th1/Th17 immune response against commensal species is associated with intestinal inflammatory diseases. On the other hand the excessive polarization toward a Treg response attenuates the immune response to infections (Brenchley et al., 2004). Another example of GALT dysregulation occurs in HIV due to the destruction of the intestinal gut barrier and the ensuing chronic inflammatory response. GALT depends on the microbiota and it is worth noting that the development of GALT and the activation of T cell lymphocytes and B cell lymphocytes cannot occur in the absence of signals released by the intestinal flora.

Experiments on commensals belonging to Clostridium and Bacteroides species further highlighted the ability of the intestinal microbiota to maintain the intestinal homeostasis and to orchestrate an adequate T-cell response through specific MAMPs during bacterial invasion (Ivanov et al., 2008; Lécuyer et al., 2014). Thus, Bacteroides fragilis strains expressing polysaccharide A binding TLR2 on CD4+T cells and gut-indigenous Clostridium belonging to XIVa and IV clusters favor Tregs, suppress Th-17 cells and facilitate mucosal tolerance toward the colon microbiota (Mazmanian et al., 2008; Round et al., 2011; Atarashi et al., 2013). On the other hand segmented filamentous bacteria, a genetic relative of the genus Clostridium promote both effector Th17 CD4+T cells (Ivanov et al., 2009; Farkas et al., 2015; Schnupf et al., 2017) and sIgA antibodies during intestinal invasion (Lécuyer et al., 2014). Other microbial species also modulate the Treg/Th17 axis, potentially controlling the intestinal inflammation and tolerance (Pandiyan et al., 2019). This aspect also explains the disequilibrium between these species during dysbiosis as well as the expansion of pathogens and of mucous lesions.

Epigenetic Regulatory Implications

Modulation of gene expression by epigenetic mechanisms in the intestinal environment was studied especially for neoplasms and inflammatory processes (Vdovikova et al., 2018). Epigenetic mechanisms modulate gene transcription by various processes: DNA methylation, histone modifications and modulation of long non-coding RNA and microRNA expression. However the mechanisms through which bacteria are either affected or induce gut epigenetic changes are not well understood.

Histone modifications are correlated with the activation or repression of genetic transcription through the modulation of two antagonistic enzymes, namely histone acetyl transferases (HATs) inducing histone acetylation, and histone deacetylases (HDACs) inducing histone deacetylation. During acetylation, the chromatin structure loosens and can be accessed by transcription factors. HDAC inhibitors increase histone acetylation and subsequently regulate gene expression in numerous immune cells such as epithelial cells, neutrophils, APCs and T cells. On the contrary, deacetylation of histones by HDACs prevents gene transcription. Thus studies performed in vitro showed that HDAC inhibitors promote the release of several transcription factors such is NFκB, MyoD, p53, or HSP90 (Vinolo et al., 2011; Kumar S. A. et al., 2018; Banik et al., 2019). The NF-kB imbalance has been correlated with numerous inflammatory and antiapoptotic mechanisms that interfere with viral evasion (Le Negrate, 2012; Carrasco Pro et al., 2018), septic shock (Liu et al., 1999; Han et al., 2002), or inflammatory diseases (Makarov, 2001; Lawrence, 2009). Likewise, p53 regulates the cell cycle and apoptosis, functioning as a tumor suppressor (Aubrey et al., 2018). P53-mediated apoptosis was associated with the spread of viral infections (Lazo and Santos, 2011; Aloni-Grinstein et al., 2018), while HSP90 reverse transcriptase mediated activity was, most likely, associated with extensive hepatitis B virus (HBV) infection (Hu and Seeger, 1996). Several viruses among which HIV, HBV, hepatitis C virus, Epstein Barr virus, cytomegalovirus, herpes simplex virus and human T-lymphotropic virus have NF-kB activation strategies and cell apoptosis blockage (Santoro et al., 2003) while others, especially the oncogenic ones have p53 suppression mechanisms (Sato and Tsurumi, 2013). It has been demonstrated that HDAC inhibitors could block stellate cell activation thus hindering liver fibrosis in experimental animal models (Park et al., 2014; Ding et al., 2018).

By inhibiting HDAC, SCFAs could therefore control numerous infectious and immune processes (Vinolo et al., 2011; Zhou et al., 2017b; Sun et al., 2018). Although the role of HDAC inhibitors hasn’t been completely elucidated yet, most experimental studies evinced an anti-inflammatory dose dependent effect (Yin et al., 2001; Le Poul et al., 2003; Weber and Kerr, 2006; Asarat et al., 2016; Kaisar et al., 2017; Li et al., 2018) and an immunosuppressive role of SCFAs in tumors (Villagra et al., 2010; Tang et al., 2011). Still, while other HDAC inhibitors are already used in oncologic therapies, the exact role of SCFAs in the immune and tumor processes remains purely theoretical (Meijer et al., 2010; Matthews et al., 2012; Ulven, 2012).

MicroRNAs (miRNAs) are small, evolutionary conserved non-coding RNAs of approximately 19-23 nucleotides involved in the post-transcriptional regulation of cellular mRNAs. The biogenesis of mature miRNA includes a two-step cleavage process from primary miRNAs (pri-miRNA). The mature miRNA is then loaded into the effector complex RNA-induced complex (RISC). RISC interacts with target mRNA and induce mRNA cleavage or translational repression hence controlling diverse metabolic or cellular pathways including cell cycle progression, differentiation, apoptosis, immune regulation or oncogenesis (Dalmasso et al., 2011; Singh et al., 2012; Nakata et al., 2017). miRNAs are released extracellularly by most eukaryotic cells and various types of small non-coding RNAs (sRNAs) around 50–200 nt in length have been observed within extracellular vesicles released by gram negative bacteria (“outer membrane vesicles”) (Gong et al., 2011). The general conception is that sRNA, as well as microRNA-size small RNAs (msRNAs) of 15–25 nucleotides in length such as msRNA observed in the model bacterium E. coli (Kang et al., 2013) act as post-transcriptional regulators and function as signaling molecules for bacterial growth and virulence under experimental conditions (Ortega et al., 2012; Zhao et al., 2017) and virulence mechanisms, at least under experimental conditions (Padalon-Brauch et al., 2008; Choi et al., 2017). The role of these structures in intestinal homeostasis, has been, for now, scarcely covered, most information having been acquired from studies on conventional or germ-free mice.

Intestinal miRNAs are released by IECs (Liu et al., 2016) and are regulated by intestinal microbiota through TLR/MyD88-dependent pathway (Dalmasso et al., 2011; Singh et al., 2012; reviewed in Eulalio et al., 2012; Williams et al., 2017). Liu and Weiner (2016) conducted an extensive study identifying numerous types of extracellular miRNAs circulating in exosomes in the gut lumen and feces of mice and humans. The study proved that IECs and Hopx-expressing cells are the main sources of exosomal intestinal miRNAs and also highlighted their uptake by intestinal bacteria and potential role in post-transcriptional regulation of bacterial genes.

Studies on mice indicate a certain pattern of miRNA gut compartimentalization after bacterial colonization (Dalmasso et al., 2011) and reported the dominance of miR-143,-145 in the jejunum and cecum and of miR-200b in the large intestine and caecum (Singh et al., 2012). On the other hand the human cells or animal experimental models with pathogenic species (Helicobacter pylori, Citrobacter rodentium, Listeria monocytogenes, Francisella tularensis, and Salmonella enterica) induce a different miRNA panel mainly represented by miR-155 and miR-146 (Eulalio et al., 2012; Archambaud et al., 2013; Staedel and Darfeuille, 2013).

On a molecular level, there is a growing interest for the intestinal role of miRNAs and their regulatory implications in the gut-barrier functionality but data on this topic is divergent, scarce and fragmented. Cellular and extracellular miRNAs from the intestinal lumen modulate the epithelial integrity, inflammatory response, and probably bacterial gene mRNAs through insufficiently known mechanisms (reviewed in Belcheva, 2017).

Studies on human intestinal cells have documented the miRNA importance in the protection of epithelial tight junctions (e.g., miRNA-122) (Ye et al., 2011), epithelial regeneration (e.g., miR-143, miR-145) (Chivukula et al., 2014), and proliferation (miR-30 family members) (Peck et al., 2016), the modulation of epithelial integrity (e.g., miR-122) (Ye et al., 2011), mucin gene expression (Mo et al., 2016), and epithelial permeability (e.g., miR-21-5p) (Nakata et al., 2017).

Intestinal miRNAs are also key regulators of the immune response against infections. Host miRNAs orchestrate the immune response through PRR families and TLR signaling pathways (miR-146a) (Xue et al., 2011, 2014), while the microbiota downregulates the expression of miR-10a and miR-107 in host DCs, decreases the release of proinflammatory cytokines in mice and controls the excessive inflammatory response in human and mice (e.g., miRNA-146a and miR-193a-3p) (Taganov et al., 2006; Nahid et al., 2011; Singh et al., 2012; Dai et al., 2015). MiRNAs could also potentially influence the microbiota and alleviate colonic inflammation as was suggested by a negative correlation on ulcerative colitis between the release of miR-193a-3p and colonic inflammation (Dai et al., 2015). This concept was further explored in a study in which the oral ingestion of endotoxin in mice led to the upregulation of epithelial miR-146 and promoted innate immune tolerance and epithelial protection in the postnatal period (Chassin et al., 2010). On a similar note mi-RNA-155 defective mice failed to develop a protective immunity toward H. pylori or Salmonella Typhimurium (Rodriguez et al., 2007).

Pathogenic bacteria also release membrane or outer-membrane vesicles containing sRNAs that modulate the host miRNA profile and gene expression (Gu et al., 2017). Some studies on intestinal inflammation due to enteropathogens revealed the upregulation of miR-16,-21,-223,-594 and miRNA-31 or downregulation of miR-124 within the human intestinal lumen (Wu et al., 2010; Koukos et al., 2013; Lin et al., 2014). Their biological relevance is under study.

Various miRNAs have been correlated with intestinal oncogenesis according to human studies. In this respect miR-182,-503 and miR-17∼92 clusters modulate glycan production and correlate with the growth of certain bacteria which could potentially initiate the microenvironmental changes in colorectal cancer (Yuan et al., 2018). Gut miRNAs could therefore induce and perpetuate dysbiosis favoring various infections or intestinal cancers (Liu et al., 2016; Williams et al., 2017). Furthermore, single nucleotide polymorphisms of miRNA-146a,-27a genes promote various infections associated with inflammatory and neoplasic intestinal changes (Song et al., 2013; Shao et al., 2014). Butyrate was also shown to affect miRNA-106b expression in IECs and to modulate carcinogenesis (Hu et al., 2011). Therefore, it is probable that epigenetic regulators such as miRNAs could play a key role in the interaction between host and microbiota and miRNA dysregulations.

Dysbiosis, the Trojan Horse Inside the Gut Barrier

Dysbiosis and Enteral Infections

Enteric pathogens alter the intestinal barrier, antagonize the intestinal microbiota, and trigger enteral infections through various mechanisms including increased intestinal inflammation, the release of bacteriocins and upregulation of AMPs and toxin delivery secretory systems (T6SS, T3SS) as well as the exploitation of nutrients or intestinal niches (reviewed in Rolhion and Chassaing, 2016; Sorbara and Pamer, 2018).

Dysbiosis occurs as a consequence of different enteroinvasive or entorotoxinogenic species which distrupt colonization resistance, reduce the protective species belonging to the Bacteroidetes and Firmicutes phylum (especially the Clostridiales incertae sedis XI or the IV/XIVa cluster) and allow the overgrowth of species belonging to the Proteobacteria phylum (especially Enterococcaceae and Enterobacteriaceae families) (Vincent et al., 2013; Livanos et al., 2018). Antibiotic treatment of intestinal infections additionally modifies the structure of the microbiota and drastically disturbs the process of “colonization resistance” (Panda et al., 2014).

As a result, the first alterations due to dysbiosis include the loss of commensals synthesizing bacteriocins and SCFAs such as Bacteroides thuringiensis and the Lachnospiraceae and Ruminococcaceae families (Rea et al., 2010) along with the loss of Clostridia commensals competing with pathobionts for the same intestinal niche (Sailhamer et al., 2009; Buffie et al., 2015; Geerlings et al., 2018) and of Bacteroides thetaiotaomicron and Akkermansia muciniphila which protect the gut barrier (Hooper et al., 2001; Donato et al., 2010; Martín et al., 2019). In these cases, the normally scarce pathobionts eventually become the dominant species and display various virulence mechanisms. Consequently, pathobionts or their endotoxins trigger intestinal infections, gut sepsis or postantibiotic colitis (Brown et al., 2013; Pérez-Cobas et al., 2013). Moreover, antibiotic treatment favors the development of multidrug-resistance species (MDR) and in turn these become invasive and pathogenic after extraluminal translocation (Hirakata et al., 2002; Ayres et al., 2012). Hence, Pseudomonas MDR species with efflux pump systems like MexAB-OprM develop quorum sensing machinery to sense host stress and express multiple virulence determinants (Hirakata et al., 2002). The wide distribution of efflux pump systems in MDR intestinal species after antibiotic treatment increases the virulence of these species and their associated risk of extraluminal translocation (Nishino et al., 2006; also reviewed in Nikaido, 1996). Therefore dysbiosis may be both the cause and the consequence of enteral infection.

The restoration of the microbiota is particularly difficult and is best ensured through fecal microbiota transplantation (FMT) from a healthy donor. The effectiveness of FMT in the management of C. difficile enterocolitis or sepsis dysbiosis further underlines the critical role of the microbiota for gut health (Gough et al., 2011; Rao and Young, 2015; Wei et al., 2016).

Dysbiosis and Sepsis

Sepsis is a life-threatening organ dysfunction caused by a dysregulated inflammatory response to infectious agents or their proinflamatory products. Sepsis has a progressive and fatal course to generalized microvascular injury, cellular hypoxia, and shock (Singer et al., 2016). Under certain conditions enteric bacteria could elicit a dysregulated systemic inflammatory response causing sepsis (Deitch et al., 1994; Mainous et al., 1995). The germs most commonly encountered in gut-derived sepsis are Gram negative bacilli producing either endotoxins or pore-forming exotoxins (Wallace et al., 2000) and probably one of the most common species involved in these cases is E. coli, a common gut resident (MacFie et al., 1999). The presence of pore-forming exotoxins is followed by the efflux of cellular potassium and activation of NLRP3 inflammasome. In turn these contribute to cellular apoptosis and loss of epithelial integrity and permit the access of toxins and pathogens to the portal and systemic circulation. This further stimulates the releases of various pro-inflammatory and blood coagulation mediators and a cytokine storm followed by vasoconstriction, ischemia and peripheral necrosis and finally organ dysfunction and shock (reviewed in Los et al., 2013; Sonnen and Henneke, 2013). The excessive activation of the CD14/TLR4/MD2 complex by bacterial endotoxins also induces the inflammasome signaling pathway, along with caspase activation (Hotchkiss et al., 1999), cells lysis and disproportionate release of pro-inflammatory cytokines, further eliciting and exacerbating sepsis, reviewed in Lamkanfi (2011), Gao et al. (2018), and Skirecki and Cavaillon (2019).

Sepsis alters the intestinal barrier through multiple factors which ultimately promote dysbiosis including intestinal ischemia and inflammatory lesions, aggressive care, antibiotic treatments, intestinal comorbidities, parenteral nutrition, etc. (reviewed in Hassoun et al., 2001; Alverdy and Luo, 2017; Fay et al., 2017). Gut vascular dysfunctions in sepsis and particularly intestinal ischemia stimulates pro-inflammatory cytokines, activates HIF-1 alpha factor, disrupts the epithelial tight junction and finally induces colonic cell apoptosis (Hassoun et al., 2001; Li et al., 2009; Yoseph et al., 2016). All of these events increase the permeability of intestinal epithelia and promote intestinal translocations (Diebel et al., 2003). The intestinal translocation of bacteria or microbial products such as endotoxin-LPS or MAMPs perpetuates and aggravates the systemic pro-inflammatory response further speeding the progression to organ failure and death (Shimizu et al., 2006, 2011; also reviewed in Deitch and Berg, 1987; Doig et al., 1998; Meng et al., 2017).

Dysbiosis entails the decrease of anaerobes Bifidobacterium and Lactobacillus (the main producers of SCFAs) accompanied by a detrimental increase of intestinal pathobionts such as E. coli or Staphylococcus and Pseudomonas species (Shimizu et al., 2006, 2011; Hayakawa et al., 2011; Ayres et al., 2012) replacing the protective microbiome. Additionally, the reduction of SCFAs favor the cascade release of LPS-induced pro-inflammatory mediators, LPS-triggered macrophage migration and massive release of nitric oxide by neutrophils (PMN), thus promoting intestinal inflammation and destabilizing the gut-barrier (Maa et al., 2010; Wang et al., 2017). Certain strains or specific phylogenetic groups of intestinal Gram negative bacilli (E. coli and Pseudomonas aeruginosa) acquire additional virulence genes during colonization or even change their morphotype after translocation to express virulence genes and to avoid host defense mechanisms giving rise to gut-derived sepsis (Johnson et al., 2001; Zaborina et al., 2007; Hickey et al., 2018). Thus, dysbiosis induces and supports sepsis associated mechanisms. In favor of this hypothesis, Souza et al. (2004) administered LPS to germ free mice and reported the absence of a severe inflammatory response and low mortality. Literature data on sepsis dysbiosis in newborns has confirmed significant changes of the microbiome, a decreased bacterial diversity, pre-sepsis gut colonization with invasive species (Carl et al., 2014), intestinal translocations (Madan et al., 2012; Mai et al., 2013; Taft et al., 2015; Stewart C. J. et al., 2017) or the activation of virulence and antibiotic resistance factors in intestinal germs (Mittal and Coopersmith, 2014). Of note, these alterations continue to occur throughout the progression of sepsis.

The administration of probiotics in sepsis (Lactobacillus rhamnosus and Bifidobacterium longum) decreases epithelial apoptosis as well as the release of cytokines and bacterial translocations in experimental mice models (Khailova et al., 2013; Panpetch et al., 2017). Probiotics can also restore the lactic acid-producing flora and ensure colonization resistance toward pathogenic flora. According to Haak and Wiersinga (2017), probiotics further influence the prognosis in sepsis by down-regulating colonic TLR-2/TLR-4 via MyD88 and by mitigating the systemic pro-inflammatory response. However, their benefit to reduce the probability of sepsis in critically ill patients is questionable (Jain et al., 2004; Kotzampassi et al., 2006; Jacobi et al., 2011; Panigrahi et al., 2017). On the other hand, data on prebiotic supplementation in infections is discordant and clear recommendations are missing (Srinivasjois et al., 2013; Chi et al., 2019).

Dysbiosis and Liver Infections

The liver is connected to the intestine through the portal vein, biliary tract, and numerous signaling molecules, together building the “gut-liver axis.” As a result, the liver is continuously exposed to microbial and metabolic molecules produced by intestinal bacteria (SCFAs, MAMPs) and further releases its own antibacterial products (bile salts) involved in the intestinal homeostasis (reviewed in Macpherson et al., 2016). Thus intestinal MAMPs reach the portal system and hepatic sinusoids and are further processed by Kuppfer cells and liver-resident T cells. SCFAs released by the microbiota are absorbed in the colon and filtered intrahepatically where most modulate the immune response, oncogenesis and are involved in the epigenetic control of liver pathogens through HDAC inhibition and transcriptional changes (Candido et al., 1978). As HDAC inhibitors, SCFAs decrease HBV replication (Pollicino et al., 2006), mediate hepatitis C virus replication (Taguwa et al., 2008, 2009), impede liver necrosis and prevent hepatocellular carcinoma, reviewed in Koumbi and Karayiannis (2015). Recently, butyrate was shown to regulate the expression of tumor-suppresive miRNAs (miRNA-26a,-26a-1,-192, etc.) through the bile acid nuclear receptor, farnesoid X receptor (FXR) while also promoting hepatocyte apoptosis through miR-22 up-regulation in hepatic cells (Pant et al., 2017; reviewed in Feng Q. et al., 2018).

Host liver miRNAs are able to regulate the replication of viral hepatitis yet the precise roles of intestinal miRNAs in the regulation of hepatotropic viruses remain unclear (Bandiera et al., 2016; Feng Q. et al., 2018; reviewed in Fan and Tang, 2014; Li et al., 2016). Hepatocytes also release immunoregulatory molecules such as primary bile acids (BA). A small proportion of primary BA are dehydroxylated in the intestine through bile salt hydrolases and converted to secondary BA entering the entero-hepatic circulation. The latter exert a bacteriostatic effect on the intestinal barrier and an anti-fibrotic and anti-inflammatory intrahepatic activity (Wu et al., 2012).

Dysbiosis is closely related to liver pathology. The liver provides an immunotolerogenic environment with an attenuated immune responsiveness and an increased risk of persistent infections (Crispe, 2014). Chronic liver diseases and their neurologic complications (hepatic encephalopathy and coma) are induced and aggravated by changes of the gut microbiota such as the overgrowth of pathobionts (Enterobacteriaceae, Bacteroidaceae, and Enterococcus species) and the dramatic decline of commensals such as Roseburia, Bifidobacterium, and Lactobacillales (Wu et al., 2012; Bajaj et al., 2014; Schnabl and Brenner, 2014). On the other hand cirrhosis modifies the composition of the microbiota, namely it decreases commensal Clostridia species (the clusters XI and XIVab), the Bacteroidales-Prevotella group and butyrate-producing Roseburia genus (Lu et al., 2011; Bajaj et al., 2012; also reviewed in Schnabl, 2013; Kang and Cai, 2017). HBV associated cirrhosis significantly reduces the Bifidobacterium/Enterobacteriaceae ratio and increases the bacterial virulence through adhesins and toxins (Lu et al., 2011). The ensuing dysbiosis compromises the integrity of the intestinal barrier and favors bacterial translocations, bacteraemia, or endotoxemia (Mainous et al., 1991; Lin et al., 1995; Bajaj et al., 2014). Given the reduced phagocytosis and canalization of porto-systemic shunts in this setting, bacterial translocations increase the risk of primitive peritonitis and septic shock.

Translocated bacteria and their derived products enter the portal circulation and liver sinusoids, promote liver inflammation (Seki et al., 2007) and aggravate the fibrogenic and oncogenic potential of liver diseases, reviewed in Yang and Seki (2012).

The mechanisms through which intestinal dysbiosis promotes liver injury employ the activation of TLR signaling pathways, reviewed in Seki and Schnabl (2012). Hence, LPS binding to TLR4 receptors of Kuppfer cells triggers a cascade of inflammatory cytokines (Su et al., 2000) and plays a key role in hepatic injury. Bacterial DNA activates TLR9 signaling pathways in Kuppfer and stellate cells which further induces liver inflammation and fibrogenesis in animal models (Miura et al., 2010). NLRP3 and NLRP6 inflammasome deficiency due to intestinal dysbiosis activate TLR4 and TLR9 pathways and exacerbate hepatic inflammation (Henao-Mejia et al., 2012). Therefore dysbiosis ensures a pro-inflammatory intrahepatic environment (Richer et al., 2013; Miura and Ohnishi, 2014; Carrasco Pro et al., 2018) and supports the progression to liver cirrhosis. The imbalance of BA synthesis in dysbiosis contributes to liver inflammation, fibrosis and carcinogenesis as well as to small intestine bacterial overgrowth, which in turn prompts an intestinal inflammatory response (Gunnarsdottir et al., 2003; Jun et al., 2010; Ridlon et al., 2014).

Considering the pathogenic role of microbiota in liver diseases various authors investigated therapeutic and prophylactic interventions. Studies on the latter explored the therapeutic use of probiotics (mostly Lactobacillus and Bifidobacterium species), antibiotics (Rifaximine), gut-derived hormones and even FMT for the manipulation of the gut-liver axis (Ponziani et al., 2015; Zhou et al., 2017a; Román et al., 2019; also reviewed in Wiest et al., 2017). Concurrently, the administration of conjugated BA in rats could normalize bile secretion and reduce intestinal bacterial overgrowth and translocations (Lorenzo-Zúñiga et al., 2003).

Dysbiosis and HIV Infection

Human immunodeficiency virus is a retrovirus with CD4+T cell tropism leading to a severe immunodeficiency and AIDS, followed by secondary infections, reactivations of opportunistic germs and HIV-associated cancers. HIV induces systemic immune activation, alters the intestinal barrier and promotes irreversible metabolic, cardiovascular and neurologic changes (Steele et al., 2014). As most experiments on non-human primate model of acquired immunodeficiency have shown, HIV/simian immunodeficiency virus (SIV) induces an ongoing activation of the mucosal immune response and a rapid and massive destruction of CD4+T lymphocytes belonging to the GALT system. The cells most affected are the Th17/22 CD4+T intestinal subsets involved in the maintenance of the gut barrier (Veazey et al., 1998; Guadalupe et al., 2003; Brenchley et al., 2004, 2006; McGowan et al., 2004; Kim et al., 2013). Thus, HIV reconfigures the intestinal barrier early during the infection through immune mechanisms which promote gut alterations, chronic inflammation, and immunodeficiency.

Human immunodeficiency virus-associated intestinal lesions (“HIV enteropathy”) initially arise due to the imbalance of the epithelial barrier turnover (Sankaran et al., 2008). In time, the increasing inflammation and intestinal permeability contribute to HIV enteropathy and persist despite antiretroviral therapy (ART) (Olsson et al., 2000; Guadalupe et al., 2003; Neff et al., 2018). The histopathological lesions described in this setting involve intestinal inflammation and mucosal atrophy that nevertheless differ from changes recorded in other intestinal immune-mediated diseases (Magro et al., 2013). On a molecular level, animal studies showed that during HIV enteropathy, IECs display a downregulation of host genes and of miRNAs associated with the homeostasis of the epithelial barrier (Sankaran et al., 2005), epithelial permeability (miRNA-21,-130a,-212) (Gaulke et al., 2014; Zhang et al., 2015; Kumar V. et al., 2018), inflammatory signaling (miR-150) (Kumar et al., 2016), and immune activation syndrome (miR-34a) (Mohan et al., 2015). Some of these miRNAs also regulate the gut microbiota (Johnston et al., 2018) or inflammatory pathways associated with irritable bowel syndrome (Zhou et al., 2010, 2015; Fourie et al., 2014). A study performed on the intestinal lamina propria and leukocytes of SIV-infected macaques indicated that the host miRNA gut profile displays a bitemporal variation initially dictated by the rapid reduction of SIV replication and T-cell activation and afterwards correlated with the consequent boost of the inflammatory response (Kumar et al., 2016). Under these conditions gut epithelia gradually lose their protective role and permit the translocation of bacterial immunogenic molecules (Chege et al., 2011; Marchetti et al., 2013) or enteropathogens (Zeitz et al., 1998; Nazli et al., 2010). In the presence of an impaired immune response, these translocations recur and re-enforce the systemic inflammatory response activating CD4+T cells and HIV replication in a vicious circle (Kotler et al., 1993; Brenchley et al., 2006; Cassol et al., 2010; Dillon et al., 2014). Thus dysbiosis perpetuates enteric and opportunistic infections and aggravates the course of HIV.

Studies on HIV associated dysbiosis are discordant, hindering comparisons, yet available data shows a major depletion of Bacteroides in favor of Proteobacteria (Dillon et al., 2014), the enrichment of pathobionts (Enterococcus, Streptococcus, Staphylococcus, Salmonella, and Escherichia species) and the significant reduction of symbionts throughout the entire intestinal tract (Nishitsuji et al., 2017). Whereas the higher population of Enterobacteriaceae increases the risk of translocations, epithelial cells exposed to E. coli could increase the susceptibility of CD4+Th17 and Th-1 lymphocytes to HIV and viral replication (Dillon et al., 2012).

Intestinal dysbiosis occurs early during HIV and is aggravated by ART itself (Ling et al., 2016; Pinto-Cardoso et al., 2017; also reviewed in Pinto-Cardoso et al., 2018). Hence, patients with immunologic failure on ART, predominantly host species of Enterobacteriaceae instead of Lactobacillus known for its immunomodulatory and anti-inflammatory activity (Merlini et al., 2011). Otherwise, butyrate synthesized by dominant species in HIV patients (Fusobacterium nucleatum, Clostridium cochlearium, and Eubacterium multiforme) could reactivate both unintegrated HIV-1 genomes and latent HIV proviruses through HDAC inhibition along with other transcription factors and concurrently explains HIV immunosuppression during ART (Kantor et al., 2009; Imai et al., 2012; Lee et al., 2018). The role of SCFAs in the regulation of mucosal immunity during HIV is controversial due to their implication in the reactivation of various infections as well as in the attenuation of mucosal inflammation and microbial translocation (Das et al., 2015; Ye and Karn, 2015; Dillon et al., 2017). Consequently, certain authors proposed the use of chemical HDAC inhibitors but not of SCFAs along with ART for a successful antiretroviral treatment of HIV reservoirs as well as for the attenuation of the LPS-induced inflammatory response (Archin et al., 2009, 2012; also reviewed in Shirakawa et al., 2013; McManamy et al., 2014).

In conclusion, HIV pathogenesis involves the loss of the immunomodulatory gastrointestinal activity, bacterial translocations and intestinal dysbiosis, independent of ART. Current therapeutic strategies are modest and based on few studies in humans or macaques: the administration of probiotics (Hummelen et al., 2011; Ortiz et al., 2016; also reviewed in Hummelen et al., 2010; D’Angelo et al., 2017; Kazemi et al., 2018), anti-inflammatory drugs (reviewed in Deeks et al., 2013), FMT (Hensley-McBain et al., 2016; Vujkovic-Cvijin et al., 2017) stimulating the immune response with a T lymphocyte-adjuvated-DNA vaccine (Fuller et al., 2012), or using an oral recombinant vaccine with Cl. perfringens expressing HIV-1 Gag protein (Pegu et al., 2011).

Conclusion

The microbiota, the intestinal epithelia and mucosal immunity form an anti-infectious barrier rigorously regulated through complex mechanisms. The host-microbiota-pathogen interactions employ numerous cell receptors and molecules with antibacterial and anti-inflammatory roles, which also modulate the epigenetic and immune response. Together with epithelial and immune cells, these signaling molecules form a network that is essential for intestinal homeostasis and anti-infectious defense. Dysbiosis renders these defense mechanisms non-functional. It consequently aggravates gastrointestinal infections, favors bacterial and LPS translocations in sepsis, unbalances the immune defense in hepatitis viruses and HIV replications and controls the progression of infectious diseases to an unknown extent. A better knowledge on the interactions driving the antimicrobial response of the intestinal barrier is therefore crucial to improve the current anti-infectious armamentarium.

Author Contributions

Both authors contributed equally to the acquisition, analysis, and critical revision of the manuscript, gave their consent for the publication, and agreed to be responsible for the accuracy and integrity of the manuscript.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Abreu, M. T., Fukata, M., and Arditi, M. (2005). TLR signaling in the gut in health and disease. J. Immunol. 174, 4453–4460. doi: 10.4049/jimmunol.174.8.4453

PubMed Abstract | CrossRef Full Text | Google Scholar

Aloni-Grinstein, R., Charni-Natan, M., Solomon, H., and Rotter, V. (2018). P53 and the viral connection: back into the future ‡. Cancers 10:E178. doi: 10.3390/cancers10060178

PubMed Abstract | CrossRef Full Text | Google Scholar

Alpan, O., Rudomen, G., Matzinger, P., and Kapsenberg, M. L. (2001). The role of dendritic cells, B cells, and M cells in gut-oriented immune responses. J. Immunol. 166, 4843–4852. doi: 10.4049/jimmunol.166.8.4843

PubMed Abstract | CrossRef Full Text | Google Scholar

Alverdy, J. C., and Luo, J. N. (2017). The Influence of host stress on the mechanism of infection: lost microbiomes, emergent pathobiomes, and the role of interkingdom signaling. Front. Microbiol. 8:322. doi: 10.3389/fmicb.2017.00322

PubMed Abstract | CrossRef Full Text | Google Scholar

Archambaud, C., Sismeiro, O., Toedling, J., Soubigou, G., Bécavin, C., Lechat, P., et al. (2013). The intestinal microbiota interferes with the microRNA response upon oral Listeria infection. MBio. 4:e00707-13. doi: 10.1128/mBio.00707-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Archin, N. M., Keedy, K. S., Espeseth, A., Dang, H., Hazuda, D. J., and Margolis, D. M. (2009). Expression of latent human immunodeficiency type 1 is induced by novel and selective histone deacetylase inhibitors. AIDS 23, 1799–1806. doi: 10.1097/QAD.0b013e32832ec1dc

PubMed Abstract | CrossRef Full Text | Google Scholar

Archin, N. M., Liberty, A. L., Kashuba, A. D., Choudhary, S. K., Kuruc, J. D., Crooks, A. M., et al. (2012). Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487, 482–485. doi: 10.1038/nature11286

PubMed Abstract | CrossRef Full Text | Google Scholar

Arpaia, N., Campbell, C., Fan, X., Dikiy, S., van der Veeken, J., deRoos, P., et al. (2013). Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455. doi: 10.1038/nature12726

PubMed Abstract | CrossRef Full Text | Google Scholar

Asarat, M., Apostolopoulos, V., Vasiljevic, T., and Donkor, O. (2016). Short-chain fatty acids regulate cytokines and Th17/Treg cells in human peripheral blood mononuclear cells in vitro. Immunol. Invest. 45, 205–222. doi: 10.3109/08820139.2015.1122613

PubMed Abstract | CrossRef Full Text | Google Scholar

Atarashi, K., Tanoue, T., Oshima, K., Suda, W., Nagano, Y., Nishikawa, H., et al. (2013). Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 500, 232–236. doi: 10.1038/nature12331

PubMed Abstract | CrossRef Full Text | Google Scholar

Aubrey, B. J., Kelly, G. L., Janic, A., Herold, M. J., and Strasser, A. (2018). How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 25, 104–113. doi: 10.1038/cdd.2017.169

PubMed Abstract | CrossRef Full Text | Google Scholar

Autenrieth, I. B., and Firsching, R. (1996). Penetration of M cells and destruction of Peyer’s patches by Yersinia enterocolitica: an ultrastructural and histological study. J. Med. Microbiol. 44, 285–294. doi: 10.1099/00222615-44-4-285

PubMed Abstract | CrossRef Full Text | Google Scholar

Ayres, J. S., Trinidad, N. J., and Vance, R. E. (2012). Lethal inflammasome activation by a multidrug-resistant pathobiont upon antibiotic disruption of the microbiota. Nat. Med. 18, 799–806. doi: 10.1038/nm.2729

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajaj, J. S., Heuman, D. M., Hylemon, P. B., Sanyal, A. J., White, M. B., Monteith, P., et al. (2014). Altered profile of human gut microbiome is associated with cirrhosis and its complications. J. Hepatol. 60, 940–947. doi: 10.1016/j.jhep.2013.12.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajaj, J. S., Hylemon, P. B., Ridlon, J. M., Heuman, D. M., Daita, K., White, M. B., et al. (2012). Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 303, G675–G685. doi: 10.1152/ajpgi.00152.2012

PubMed Abstract | CrossRef Full Text | Google Scholar

Bambou, J.-C., Giraud, A., Menard, S., Begue, B., Rakotobe, S., Heyman, M., et al. (2004). In vitro and ex vivo activation of the TLR5 signaling pathway in intestinal epithelial Ccells by a commensal Escherichia coli strain. J. Biol. Chem. 279, 42984–42992.

PubMed Abstract | Google Scholar

Bandiera, S., Pernot, S., El Saghire, H., Durand, S. C., Thumann, C., Crouchet, E., et al. (2016). Hepatitis C virus-induced upregulation of microRNA miR-146a-5p in hepatocytes promotes viral infection and deregulates metabolic pathways associated with liver disease pathogenesis. J. Virol. 90, 6387–6400. doi: 10.1128/JVI.00619-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Banik, D., Moufarrij, S., and Villagra, A. (2019). Immunoepigenetics combination therapies: an overview of the role of HDACs in cancer immunotherapy. Int. J. Mol. Sci. 20:E2241. doi: 10.3390/ijms20092241

PubMed Abstract | CrossRef Full Text | Google Scholar

Barreau, F., Madre, C., Meinzer, U., Berrebi, D., Dussaillant, M., Merlin, F., et al. (2010). Nod2 regulates the host response towards microflora by modulating T cell function and epithelial permeability in mouse Peyer’s patches. Gut 59, 207–217. doi: 10.1136/gut.2008.171546

PubMed Abstract | CrossRef Full Text | Google Scholar

Belcheva, A. (2017). MicroRNAs at the epicenter of intestinal homeostasis. BioEssays 39:1600200. doi: 10.1002/bies.201600200

PubMed Abstract | CrossRef Full Text | Google Scholar

Brenchley, J. M., Price, D. A., Schacker, T. W., Asher, T. E., Silvestri, G., Rao, S., et al. (2006). Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 12, 1365–1371.

PubMed Abstract | Google Scholar

Brenchley, J. M., Schacker, T. W., Ruff, L. E., Price, D. A., Taylor, J. H., Beilman, G. J., et al. (2004). CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J. Exp. Med. 200, 749–759. doi: 10.1084/jem.20040874

PubMed Abstract | CrossRef Full Text | Google Scholar

Brown, K. A., Khanafer, N., Daneman, N., and Fisman, D. N. (2013). Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob. Agents Chemother. 57, 2326–2332. doi: 10.1128/AAC.02176-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Buffie, C. G., Bucci, V., Stein, R. R., McKenney, P. T., Ling, L., Gobourne, A., et al. (2015). Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517, 205–208. doi: 10.1038/nature13828

PubMed Abstract | CrossRef Full Text | Google Scholar

Butler, M., Chaudhary, R., van Heel, D. A., Playford, R. J., and Ghosh, S. (2007). NOD2 activity modulates the phenotype of LPS-stimulated dendritic cells to promote the development of T-helper type 2-like lymphocytes — Possible implications for NOD2-associated Crohn’s disease. J. Crohn’s Colitis 1, 106–115. doi: 10.1016/j.crohns.2007.08.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Cañas, M.-A., Fábrega, M.-J., Giménez, R., Badia, J., and Baldomà, L. (2018). Outer membrane vesicles from probiotic and commensal Escherichia coli activate NOD1-mediated immune responses in intestinal epithelial cells. Front. Microbiol. 9:498. doi: 10.3389/fmicb.2018.00498

PubMed Abstract | CrossRef Full Text | Google Scholar

Candido, E. P., Reeves, R., and Davie, J. R. (1978). Sodium butyrate inhibits histone deacetylation in cultured cells. Cell 14, 105–113. doi: 10.1016/0092-8674(78)90305-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Carl, M. A., Ndao, I. M., Springman, A. C., Manning, S. D., Johnson, J. R., Johnston, B. D., et al. (2014). Sepsis from the gut: the enteric habitat of bacteria that cause late-onset neonatal bloodstream infections. Clin. Infect. Dis. 58, 1211–1218. doi: 10.1093/cid/ciu084

PubMed Abstract | CrossRef Full Text | Google Scholar

Carrasco Pro, S., Lindestam Arlehamn, C. S., Dhanda, S. K., Carpenter, C., Lindvall, M., Faruqi, A. A., et al. (2018). Microbiota epitope similarity either dampens or enhances the immunogenicity of disease-associated antigenic epitopes. PLoS One 13:e0196551. doi: 10.1371/journal.pone.0196551

PubMed Abstract | CrossRef Full Text | Google Scholar

Cassol, E., Malfeld, S., Mahasha, P., van der Merwe, S., Cassol, S., Seebregts, C., et al. (2010). Persistent microbial translocation and immune activation in HIV-1–infected south africans receiving combination cntiretroviral therapy. J. Infect. Dis. 202, 723–733. doi: 10.1086/655229

PubMed Abstract | CrossRef Full Text | Google Scholar

Cavera, V. L., Arthur, T. D., Kashtanov, D., and Chikindas, M. L. (2015). Bacteriocins and their position in the next wave of conventional antibiotics. Int. J. Antimicrob. Agents 46, 494–501. doi: 10.1016/j.ijantimicag.2015.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Chassin, C., Kocur, M., Pott, J., Duerr, C. U., Gütle, D., Lotz, M., et al. (2010). miR-146a mediates protective innate immune tolerance in the neonate Intestine. Cell Host Microbe. 8, 358–368. doi: 10.1016/j.chom.2010.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Chege, D., Sheth, P. M., Kain, T., Kim, C. J., Kovacs, C., Loutfy, M., et al. (2011). Sigmoid Th17 populations, the HIV latent reservoir, and microbial translocation in men on long-term antiretroviral therapy. AIDS 25, 741–749. doi: 10.1097/QAD.0b013e328344cefb

PubMed Abstract | CrossRef Full Text | Google Scholar

Chi, C., Buys, N., Li, C., Sun, J., and Yin, C. (2019). Effects of prebiotics on sepsis, necrotizing enterocolitis, mortality, feeding intolerance, time to full enteral feeding, length of hospital stay, and stool frequency in preterm infants: a meta-analysis. Eur. J. Clin. Nutr. 73, 657–670. doi: 10.1038/s41430-018-0377-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Chivukula, R. R., Shi, G., Acharya, A., Mills, E. W., Zeitels, L. R., Anandam, J. L., et al. (2014). An essential mesenchymal function for miR-143/145 in intestinal epithelial regeneration. Cell 157, 1104–1116. doi: 10.1016/j.cell.2014.03.055

PubMed Abstract | CrossRef Full Text | Google Scholar

Choi, J.-W., Um, J.-H., Cho, J.-H., and Lee, H.-J. (2017). Tiny RNAs and their voyage via extracellular vesicles: secretion of bacterial small RNA and eukaryotic microRNA. Exp. Biol. Med. 242, 1475–1481. doi: 10.1177/1535370217723166

PubMed Abstract | CrossRef Full Text | Google Scholar

Clark, M. A., Jepson, M. A., Simmons, N. L., and Hirst, B. H. (1994). Preferential interaction of Salmonella typhimurium with mouse Peyer’s patch M cells. Res. Microbiol. 145, 543–552. doi: 10.1016/0923-2508(94)90031-0

CrossRef Full Text | Google Scholar

Corr, S., Hill, C., and Gahan, C. G. M. (2006). An in vitro cell-culture model demonstrates internalin- and hemolysin-independent translocation of Listeria monocytogenes across M cells. Microb. Pathog. 41, 241–250. doi: 10.1016/j.micpath.2006.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Cox, M. A., Jackson, J., Stanton, M., Rojas-Triana, A., Bober, L., Laverty, M., et al. (2009). Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E(2) and cytokines. World J. Gastroenterol. 15, 5549–5557.

PubMed Abstract | Google Scholar

Crispe, I. N. (2014). Immune tolerance in liver disease. Hepatology 60, 2109–2117. doi: 10.1002/hep.27254

PubMed Abstract | CrossRef Full Text | Google Scholar

Dai, X., Chen, X., Chen, Q., Shi, L., Liang, H., Zhou, Z., et al. (2015). MicroRNA-193a-3p reduces intestinal inflammation in response to microbiota via down-regulation of colonic PepT1. J. Biol. Chem. 290, 16099–16115. doi: 10.1074/jbc.M115.659318

PubMed Abstract | CrossRef Full Text | Google Scholar

Dalmasso, G., Nguyen, H. T. T., Yan, Y., Laroui, H., Charania, M. A., Ayyadurai, S., et al. (2011). Microbiota modulate host gene expression via microRNAs. PLoS One 6:e19293. doi: 10.1371/journal.pone.0019293

PubMed Abstract | CrossRef Full Text | Google Scholar

D’Angelo, C., Reale, M., and Costantini, E. (2017). Microbiota and probiotics in health and HIV Infection. Nutrients 9:E615. doi: 10.3390/nu9060615

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, B., Dobrowolski, C., Shahir, A.-M., Feng, Z., Yu, X., Sha, J., et al. (2015). Short chain fatty acids potently induce latent HIV-1 in T-cells by activating P-TEFb and multiple histone modifications. Virology 474, 65–81. doi: 10.1016/j.virol.2014.10.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Deeks, S. G., Tracy, R., and Douek, D. C. (2013). Systemic effects of inflammation on health during chronic HIV infection. Immunity 39, 633–645. doi: 10.1016/j.immuni.2013.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Deitch, E. A., and Berg, R. (1987). Bacterial translocation from the gut: a mechanism of infection. J. Burn Care Rehabil. 8, 475–482. doi: 10.1097/00004630-198711000-00005

CrossRef Full Text | Google Scholar

Deitch, E. A., Xu, D., Franko, L., Ayala, A., and Chaudry, I. H. (1994). Evidence favoring the role of the gut as a cytokine-generating organ in rats subjected to hemorrhagic shock. Shock 1, 141–145. doi: 10.1097/00024382-199402000-00010

PubMed Abstract | CrossRef Full Text | Google Scholar

Diebel, L. N., Liberati, D. M., Dulchavsky, S. A., Diglio, C. A., and Brown, W. J. (2003). Enterocyte apoptosis and barrier function are modulated by SIgA after exposure to bacteria and hypoxia/reoxygenation. Surgery 134, 574–580. discussion 580-1, doi: 10.1016/s0039-6060(03)00302-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Dillon, S. M., Kibbie, J., Lee, E. J., Guo, K., Santiago, M. L., Austin, G. L., et al. (2017). Low abundance of colonic butyrate-producing bacteria in HIV infection is associated with microbial translocation and immune activation. AIDS 31, 511–521. doi: 10.1097/QAD.0000000000001366

PubMed Abstract | CrossRef Full Text | Google Scholar

Dillon, S. M., Lee, E. J., Kotter, C. V., Austin, G. L., Dong, Z., Hecht, D. K., et al. (2014). An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal Immunol. 7, 983–994. doi: 10.1038/mi.2013.116

PubMed Abstract | CrossRef Full Text | Google Scholar

Dillon, S. M., Manuzak, J. A., Leone, A. K., Lee, E. J., Rogers, L. M., McCarter, M. D., et al. (2012). HIV-1 infection of human intestinal lamina propria CD4+ T cells in vitro is enhanced by exposure to commensal Escherichia coli. J. Immunol. 189, 885–896. doi: 10.4049/jimmunol.1200681

PubMed Abstract | CrossRef Full Text | Google Scholar

Ding, D., Chen, L.-L., Zhai, Y.-Z., Hou, C.-J., Tao, L.-L., Lu, S.-H., et al. (2018). Trichostatin A inhibits the activation of Hepatic stellate cells by increasing C/EBP-α acetylation in vivo and in vitro. Sci. Rep. 8:4395. doi: 10.1038/s41598-018-22662-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Doig, C. J., Sutherland, L. R., Dean Sandham, J., Fick, G. H., Verhoef, M., and Meddings, J. B. (1998). Increased intestinal permeability is associated with the development of multiple organ dysfunction syndrome in critically Ill ICU patients. Am. J. Respir. Crit. Care Med. 158, 444–451. doi: 10.1164/ajrccm.158.2.9710092

PubMed Abstract | CrossRef Full Text | Google Scholar

Donato, K. A., Gareau, M. G., Wang, Y. J. J., and Sherman, P. M. (2010). Lactobacillus rhamnosus GG attenuates interferon- and tumour necrosis factor- -induced barrier dysfunction and pro-inflammatory signalling. Microbiology 156, 3288–3297. doi: 10.1099/mic.0.040139-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Donohoe, D. R., Garge, N., Zhang, X., Sun, W., O’Connell, T. M., Bunger, M. K., et al. (2011). The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 13, 517–526. doi: 10.1016/j.cmet.2011.02.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Drissi, F., Buffet, S., Raoult, D., and Merhej, V. (2015). Common occurrence of antibacterial agents in human intestinal microbiota. Front. Microbiol. 6:441. doi: 10.3389/fmicb.2015.00441

PubMed Abstract | CrossRef Full Text | Google Scholar

Eulalio, A., Schulte, L., and Vogel, J. (2012). The mammalian microRNA response to bacterial infections. RNA Biol. 9, 742–750. doi: 10.4161/rna.20018

PubMed Abstract | CrossRef Full Text | Google Scholar

Fan, H.-X., and Tang, H. (2014). Complex interactions between microRNAs and hepatitis B/C viruses. World J. Gastroenterol. 20, 13477–13492. doi: 10.3748/wjg.v20.i37.13477

PubMed Abstract | CrossRef Full Text | Google Scholar

Farkas, A. M., Panea, C., Goto, Y., Nakato, G., Galan-Diez, M., Narushima, S., et al. (2015). Induction of Th17 cells by segmented filamentous bacteria in the murine intestine. J. Immunol. Methods 421, 104–111. doi: 10.1016/j.jim.2015.03.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Fay, K. T., Ford, M. L., and Coopersmith, C. M. (2017). The intestinal microenvironment in sepsis. Biochim. Biophys. Acta. - Mol. Basis Dis. 1863, 2574–2583. doi: 10.1016/j.bbadis.2017.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, Q., Chen, W.-D., and Wang, Y.-D. (2018). Gut microbiota: an integral moderator in health and disease. Front. Microbiol. 9:151. doi: 10.3389/fmicb.2018.00151

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, Y., Wang, Y., Wang, P., Huang, Y., and Wang, F. (2018). Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell Physiol. Biochem. 49, 190–205. doi: 10.1159/000492853

PubMed Abstract | CrossRef Full Text | Google Scholar

Fourie, N. H., Peace, R. M., Abey, S. K., Sherwin, L. B., Rahim-Williams, B., Smyser, P. A., et al. (2014). Elevated circulating miR-150 and miR-342-3p in patients with irritable bowel syndrome. Exp. Mol. Pathol. 96, 422–425. doi: 10.1016/j.yexmp.2014.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Fukuda, S., Toh, H., Hase, K., Oshima, K., Nakanishi, Y., Yoshimura, K., et al. (2011). Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 469, 543–547. doi: 10.1038/nature09646

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuller, D. H., Rajakumar, P., Che, J. W., Narendran, A., Nyaundi, J., Michael, H., et al. (2012). Therapeutic DNA vaccine induces broad T cell responses in the gut and sustained protection from viral rebound and AIDS in SIV-infected rhesus macaques. PLoS One 7:e33715. doi: 10.1371/journal.pone.0033715

PubMed Abstract | CrossRef Full Text | Google Scholar

Furusawa, Y., Obata, Y., Fukuda, S., Endo, T. A., Nakato, G., Takahashi, D., et al. (2013). Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504, 446–450. doi: 10.1038/nature12721

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, Y.-L., Zhai, J.-H., and Chai, Y.-F. (2018). Recent advances in the molecular mechanisms underlying pyroptosis in sepsis. Mediators Inflamm. 2018:5823823. doi: 10.1155/2018/5823823

PubMed Abstract | CrossRef Full Text | Google Scholar

Gaulke, C. A., Porter, M., Han, Y.-H., Sankaran-Walters, S., Grishina, I., George, M. D., et al. (2014). Intestinal epithelial barrier disruption through altered mucosal microRNA expression in human immunodeficiency virus and simian immunodeficiency virus infections. J. Virol. 88, 6268–6280. doi: 10.1128/JVI.00097-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Geerlings, S. Y., Kostopoulos, I., de Vos, W. M., and Belzer, C. (2018). Akkermansia muciniphila in the human gastrointestinal tract: when, where, and how? Microorganisms 6:E75. doi: 10.3390/microorganisms6030075

PubMed Abstract | CrossRef Full Text | Google Scholar

Gewirtz, A. T., Navas, T. A., Lyons, S., Godowski, P. J., and Madara, J. L. (2001). Cutting edge: bacterial flagellin activates basolaterally expressed TLR5 to induce epithelial proinflammatory gene expression. J. Immunol. 167, 1882–1885. doi: 10.4049/jimmunol.167.4.1882

PubMed Abstract | CrossRef Full Text | Google Scholar

Gong, H., Vu, G.-P., Bai, Y., Chan, E., Wu, R., Yang, E., et al. (2011). A Salmonella small non-coding RNA facilitates bacterial invasion and intracellular replication by modulating the expression of virulence factors. PLoS Pathog. 7:e1002120. doi: 10.1371/journal.ppat.1002120

PubMed Abstract | CrossRef Full Text | Google Scholar

Goto, Y., and Ivanov, I. I. (2013). Intestinal epithelial cells as mediators of the commensal-host immune crosstalk. Immunol. Cell Biol. 91, 204–214. doi: 10.1038/icb.2012.80

PubMed Abstract | CrossRef Full Text | Google Scholar

Gough, E., Shaikh, H., and Manges, A. R. (2011). Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin. Infect. Dis. 53, 994–1002. doi: 10.1093/cid/cir632

PubMed Abstract | CrossRef Full Text | Google Scholar

Gu, H., Zhao, C., Zhang, T., Liang, H., Wang, X.-M., Pan, Y., et al. (2017). Salmonella produce microRNA-like RNA fragment Sal-1 in the infected cells to facilitate intracellular survival. Sci. Rep. 7:2392. doi: 10.1038/s41598-017-02669-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Guadalupe, M., Reay, E., Sankaran, S., Prindiville, T., Flamm, J., McNeil, A., et al. (2003). Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J. Virol. 77, 11708–11717. doi: 10.1128/jvi.77.21.11708-11717.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Gunnarsdottir, S. A., Sadik, R., Shev, S., Simren, M., Sjovall, H., Stotzer, P.-O., et al. (2003). Small intestinal motility disturbances and bacterial overgrowth in patients with liver cirrhosis and portal hypertension. Am. J. Gastroenterol. 98, 1362–1370. doi: 10.1016/s0002-9270(03)00250-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Haak, B. W., and Wiersinga, W. J. (2017). The role of the gut microbiota in sepsis. Lancet Gastroenterol. Hepatol. 2, 135–143. doi: 10.1016/S2468-1253(16)30119-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, S.-J., Ko, H.-M., Choi, J.-H., Seo, K. H., Lee, H.-S., Choi, E.-K., et al. (2002). Molecular mechanisms for lipopolysaccharide-induced biphasic Aactivation of nuclear factor-κB (NF-κB). J. Biol. Chem. 277, 44715–44721. doi: 10.1074/jbc.m202524200

PubMed Abstract | CrossRef Full Text | Google Scholar

Hassoun, H. T., Kone, B. C., Mercer, D. W., Moody, F. G., Weisbrodt, N. W., and Moore, F. A. (2001). Post-injury multiple organ multiple organ failure:the role of the gut: shock. Shock 15, 1–10. doi: 10.1097/00024382-200115010-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

Hayakawa, M., Asahara, T., Henzan, N., Murakami, H., Yamamoto, H., Mukai, N., et al. (2011). Dramatic changes of the gut flora immediately after severe and sudden Insults. Dig. Dis. Sci. 56, 2361–2365. doi: 10.1007/s10620-011-1649-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Hayashi, F., Smith, K. D., Ozinsky, A., Hawn, T. R., Yi, E. C., Goodlett, D. R., et al. (2001). The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 410, 1099–1103. doi: 10.1038/35074106

PubMed Abstract | CrossRef Full Text | Google Scholar

Henao-Mejia, J., Elinav, E., Jin, C., Hao, L., Mehal, W. Z., Strowig, T., et al. (2012). Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 482, 179–185. doi: 10.1038/nature10809

PubMed Abstract | CrossRef Full Text | Google Scholar

Hensley-McBain, T., Zevin, A. S., Manuzak, J., Smith, E., Gile, J., Miller, C., et al. (2016). Effects of fecal microbial transplantation on microbiome and immunity in simian immunodeficiency virus-infected macaques. J. Virol. 90, 4981–4989. doi: 10.1128/JVI.00099-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Hickey, C., Schaible, B., Nguyen, S., Hurley, D., Srikumar, S., Fanning, S., et al. (2018). Increased virulence of bloodstream over peripheral isolates of P. aeruginosa identified through post-transcriptional regulation of virulence factors. Front. Cell Infect. Microbiol. 8:357. doi: 10.3389/fcimb.2018.00357

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirakata, Y., Srikumar, R., Poole, K., Gotoh, N., Suematsu, T., Kohno, S., et al. (2002). Multidrug efflux systems play an important role in the invasiveness of Pseudomonas aeruginosa. J. Exp. Med. 196, 109–118. doi: 10.1084/jem.20020005

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirota, S. A., Ng, J., Lueng, A., Khajah, M., Parhar, K., Li, Y., et al. (2011). NLRP3 inflammasome plays a key role in the regulation of intestinal homeostasis. Inflamm. Bowel Dis. 17, 1359–1372. doi: 10.1002/ibd.21478

PubMed Abstract | CrossRef Full Text | Google Scholar

Hooper, L. V., Wong, M. H., Thelin, A., Hansson, L., Falk, P. G., and Gordon, J. I. (2001). Molecular analysis of commensal host-microbial relationships in the intestine. Science 291, 881–884. doi: 10.1126/science.291.5505.881

PubMed Abstract | CrossRef Full Text | Google Scholar

Hotchkiss, R. S., Swanson, P. E., Freeman, B. D., Tinsley, K. W., Cobb, J. P., Matuschak, G. M., et al. (1999). Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Crit. Care Med. 27, 1230–1251. doi: 10.1097/00003246-199907000-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, J., and Seeger, C. (1996). Hsp90 is required for the activity of a hepatitis B virus reverse transcriptase. Proc. Natl. Acad. Sci. U.S.A. 93, 1060–1064. doi: 10.1073/pnas.93.3.1060

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, S., Dong, T. S., Dalal, S. R., Wu, F., Bissonnette, M., Kwon, J. H., et al. (2011). The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLoS One 6:e16221. doi: 10.1371/journal.pone.0016221

PubMed Abstract | CrossRef Full Text | Google Scholar

Hummelen, R., Changalucha, J., Butamanya, N. L., Koyama, T. E., Cook, A., Habbema, J. D. F., et al. (2011). Effect of 25 weeks probiotic supplementation on immune function of HIV patients. Gut. Microbes. 2, 80–85. doi: 10.4161/gmic.2.2.15787

PubMed Abstract | CrossRef Full Text | Google Scholar

Hummelen, R., Vos, A. P., Land, B., Norren, K., and Reid, G. (2010). Altered host-microbe interaction in HIV: a target for intervention with pro- and prebiotics. Int. Rev. Immunol. 29, 485–513. doi: 10.3109/08830185.2010.505310

PubMed Abstract | CrossRef Full Text | Google Scholar

Imai, K., Yamada, K., Tamura, M., Ochiai, K., and Okamoto, T. (2012). Reactivation of latent HIV-1 by a wide variety of butyric acid-producing bacteria. Cell Mol. Life Sci. 69, 2583–2592. doi: 10.1007/s00018-012-0936-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ivanov, I. I., Atarashi, K., Manel, N., Brodie, E. L., Shima, T., Karaoz, U., et al. (2009). Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498. doi: 10.1016/j.cell.2009.09.033

PubMed Abstract | CrossRef Full Text | Google Scholar

Ivanov, I. I., Frutos, R., de, L., Manel, N., Yoshinaga, K., Rifkin, D. B., et al. (2008). Specific microbiota direct the differentiation of Th17 cells in the mucosa of the small intestine. Cell Host Microbe 4, 337–349. doi: 10.1016/j.chom.2008.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Iwasaki, A., and Kelsall, B. L. (1999). Freshly isolated Peyer’s Patch, but not spleen, dendritic cells produce interleukin 10 and induce the differentiation of T helper type 2 cells. J. Exp. Med. 190, 229–240. doi: 10.1084/jem.190.2.229

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacobi, C. A., Schulz, C., and Malfertheiner, P. (2011). Treating critically ill patients with probiotics: beneficial or dangerous? Gut. Pathog. 3:2. doi: 10.1186/1757-4749-3-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Jain, P. K., McNaught, C. E., Anderson, A. D., MacFie, J., and Mitchell, C. J. (2004). Influence of synbiotic containing Lactobacillus acidophilus La5, Bifidobacterium lactis Bb 12, Streptococcus thermophilus, Lactobacillus bulgaricus and oligofructose on gut barrier function and sepsis in critically ill patients: a randomised controlled trial. Clin. Nutr. 23, 467–475. doi: 10.1016/j.clnu.2003.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, J. R., O’Bryan, T. T., Kuskowski, M., and Maslow, J. N. (2001). Ongoing horizontal and vertical transmission of virulence genes and papA alleles among Escherichia coli blood isolates from patients with diverse-source bacteremia. Infect. Immun. 69, 5363–5374. doi: 10.1128/iai.69.9.5363-5374.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnston, D. G. W., Williams, M. A., Thaiss, C. A., Cabrera-Rubio, R., Raverdeau, M., McEntee, C., et al. (2018). Loss of microRNA-21 influences the gut microbiota, causing reduced susceptibility in a murine model of colitis. J. Crohn’s Colitis 12, 835–848. doi: 10.1093/ecco-jcc/jjy038

PubMed Abstract | CrossRef Full Text | Google Scholar

Jones, R. M., and Neish, A. S. (2011). Recognition of bacterial pathogens and mucosal immunity. Cell Microbiol. 13, 670–676. doi: 10.1111/j.1462-5822.2011.01579.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Jun, D. W., Kim, K. T., Lee, O. Y., Chae, J. D., Son, B. K., Kim, S. H., et al. (2010). Association between small intestinal bacterial overgrowth and peripheral bacterial DNA in cirrhotic patients. Dig. Dis. Sci. 55, 1465–1471. doi: 10.1007/s10620-009-0870-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Jung, J., Surh, C. D., and Lee, Y. J. (2019). Microbial colonization at early life promotes the development of diet-induced CD8αβ intraepithelial T cells. Mol. Cells. 42, 313–320. doi: 10.14348/molcells.2019.2431

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaisar, M. M. M., Pelgrom, L. R., van der Ham, A. J., Yazdanbakhsh, M., and Everts, B. (2017). Butyrate Conditions Human Dendritic Cells to Prime Type 1 Regulatory T Cells via both Histone Deacetylase Inhibition and G Protein-Coupled Receptor 109A Signaling. Front. Immunol. 8:1429. doi: 10.3389/fimmu.2017.01429

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, S.-M., Choi, J.-W., Lee, Y., Hong, S.-H., and Lee, H.-J. (2013). Identification of microRNA-Size, Small RNAs in Escherichia coli. Curr. Microbiol. 67, 609–613. doi: 10.1007/s00284-013-0411-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, Y., and Cai, Y. (2017). Gut microbiota and hepatitis-B-virus-induced chronic liver disease: implications for faecal microbiota transplantation therapy. J. Hosp. Infect. 96, 342–348. doi: 10.1016/j.jhin.2017.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Kantor, B., Ma, H., Webster-Cyriaque, J., Monahan, P. E., and Kafri, T. (2009). Epigenetic activation of unintegrated HIV-1 genomes by gut-associated short chain fatty acids and its implications for HIV infection. Proc. Natl. Acad. Sci. U.S.A. 106, 18786–18791. doi: 10.1073/pnas.0905859106

PubMed Abstract | CrossRef Full Text | Google Scholar

Kazemi, A., Djafarian, K., Speakman, J. R., Sabour, P., Soltani, S., and Shab-Bidar, S. (2018). Effect of probiotic supplementation on CD4 cell count in HIV-infected patients: a systematic review and meta-analysis. J. Diet Suppl. 15, 776–788. doi: 10.1080/19390211.2017.1380103

PubMed Abstract | CrossRef Full Text | Google Scholar

Kelly, C. J., Zheng, L., Campbell, E. L., Saeedi, B., Scholz, C. C., Bayless, A. J., et al. (2015). Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 17, 662–671. doi: 10.1016/j.chom.2015.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Khailova, L., Frank, D. N., Dominguez, J. A., and Wischmeyer, P. E. (2013). Probiotic administration reduces mortality and improves intestinal epithelial homeostasis in experimental sepsis. Anesthesiology 119, 166–177. doi: 10.1097/ALN.0b013e318291c2fc

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, C. J., McKinnon, L. R., Kovacs, C., Kandel, G., Huibner, S., Chege, D., et al. (2013). Mucosal Th17 cell function is altered during HIV infection and is an independent predictor of systemic immune activation. J. Immunol. 191, 2164–2173. doi: 10.4049/jimmunol.1300829

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J. G., Lee, S. J., and Kagnoff, M. F. (2004). Nod1 is an essential signal transducer in intestinal epithelial cells infected with bacteria that avoid recognition by toll-like receptors. Infect. Immun. 72, 1487–1495. doi: 10.1128/iai.72.3.1487-1495.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Kitajima, S., Takuma, S., and Morimoto, M. (1999). Changes in colonic mucosal permeability in mouse colitis induced with dextran sulfate sodium. Exp. Anim. 48, 137–143. doi: 10.1538/expanim.48.137

PubMed Abstract | CrossRef Full Text | Google Scholar

Kotler, D. P., Reka, S., and Clayton, F. (1993). Intestinal mucosal inflammation associated with human immunodeficiency virus infection. Dig. Dis. Sci. 38, 1119–1127. doi: 10.1007/bf01295730

PubMed Abstract | CrossRef Full Text | Google Scholar

Kotzampassi, K., Giamarellos-Bourboulis, E. J., Voudouris, A., Kazamias, P., and Eleftheriadis, E. (2006). Benefits of a synbiotic formula (Synbiotic 2000Forte®) in critically Ill trauma patients: early results of a randomized controlled trial. World J. Surg. 30, 1848–1855. doi: 10.1007/s00268-005-0653-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Koukos, G., Polytarchou, C., Kaplan, J. L., Morley–Fletcher, A., Gras–Miralles, B., Kokkotou, E., et al. (2013). MicroRNA-124 regulates STAT3 expression and is down-regulated in colon tissues of pediatric patients with ulcerative colitis. Gastroenterology 145, 842.e2–852.e2. doi: 10.1053/j.gastro.2013.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Koumbi, L., and Karayiannis, P. (2015). The epigenetic control of hepatitis B virus modulates the outcome of infection. Front. Microbiol. 6:1491. doi: 10.3389/fmicb.2015.01491

PubMed Abstract | CrossRef Full Text | Google Scholar

Kühbacher, A., Novy, K., Quereda, J. J., Sachse, M., Moya-Nilges, M., Wollscheid, B., et al. (2018). Listeriolysin O-dependent host surfaceome remodeling modulates Listeria monocytogenes invasion. Pathog. Dis. 76:fty082. doi: 10.1093/femspd/fty082

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, S. A., Bishayee, A., and Pandey, A. K. (2018). Targeting histone deacetylases with natural and synthetic agents: an emerging anticancer strategy. Nutrients 10:E731. doi: 10.3390/nu10060731

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, V., Mansfield, J., Fan, R., MacLean, A., Li, J., and Mohan, M. (2018). miR-130a and miR-212 disrupt the intestinal epithelial barrier through modulation of PPARγ and occludin expression in chronic simian immunodeficiency virus-infected rhesus macaques. J. Immunol. 200, 2677–2689. doi: 10.4049/jimmunol.1701148

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, V., Torben, W., Kenway, C. S., Schiro, F. R., and Mohan, M. (2016). Longitudinal examination of the intestinal lamina propria cellular compartment of simian immunodeficiency virus-infected Rhesus macaques provides broader and deeper insights into the link between aberrant microRNA expression and persistent immune activati. J. Virol. 90, 5003–5019. doi: 10.1128/JVI.00189-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Lamkanfi, M. (2011). Emerging inflammasome effector mechanisms. Nat. Rev. Immunol. 11, 213–220. doi: 10.1038/nri2936

PubMed Abstract | CrossRef Full Text | Google Scholar

Lawrence, T. (2009). The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect. Biol. 1:a001651. doi: 10.1101/cshperspect.a001651

PubMed Abstract | CrossRef Full Text | Google Scholar

Lazo, P. A., and Santos, C. R. (2011). Interference with p53 functions in human viral infections, a target for novel antiviral strategies? Rev. Med. Virol. 21, 285–300. doi: 10.1002/rmv.696

PubMed Abstract | CrossRef Full Text | Google Scholar

Le Negrate, G. (2012). Viral interference with innate immunity bypreventing NF-kB activity. Cell Microbiol. 14, 168–181. doi: 10.1111/j.1462-5822.2011.01720.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Le Poul, E., Loison, C., Struyf, S., Springael, J.-Y., Lannoy, V., Decobecq, M.-E., et al. (2003). Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J. Biol. Chem. 278, 25481–25489. doi: 10.1074/jbc.m301403200

PubMed Abstract | CrossRef Full Text | Google Scholar

Lécuyer, E., Rakotobe, S., Lengliné-Garnier, H., Lebreton, C., Picard, M., Juste, C., et al. (2014). Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses. Immunity 40, 608–620. doi: 10.1016/j.immuni.2014.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. C., Chua, L. L., Yap, S. H., Khang, T. F., Leng, C. Y., Raja Azwa, R. I., et al. (2018). Enrichment of gut-derived Fusobacterium is associated with suboptimal immune recovery in HIV-infected individuals. Sci. Rep. 8:14277. doi: 10.1038/s41598-018-32585-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Levy, M., Thaiss, C. A., Katz, M. N., Suez, J., and Elinav, E. (2015). Inflammasomes and the microbiota—partners in the preservation of mucosal homeostasis. Semin. Immunopathol. 37, 39–46. doi: 10.1007/s00281-014-0451-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., Jiang, J.-D., and Peng, Z.-G. (2016). MicroRNA-mediated interactions between host and hepatitis C virus. World J. Gastroenterol. 22, 1487–1496. doi: 10.3748/wjg.v22.i4.1487

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, M., van Esch, B. C. A. M., Henricks, P. A. J., Garssen, J., and Folkerts, G. (2018). Time and concentration dependent effects of short chain fatty acids on lipopolysaccharide- or tumor necrosis factor α-induced endothelial activation. Front. Pharmacol. 9:233. doi: 10.3389/fphar.2018.00233

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Q., Zhang, Q., Wang, C., Liu, X., Li, N., and Li, J. (2009). Disruption of tight junctions during polymicrobial sepsis in vivo. J. Pathol. 218, 210–221. doi: 10.1002/path.2525

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, J., Welker, N. C., Zhao, Z., Li, Y., Zhang, J., Reuss, S. A., et al. (2014). Novel specific microRNA biomarkers in idiopathic inflammatory bowel disease unrelated to disease activity. Mod. Pathol. 27, 602–608. doi: 10.1038/modpathol.2013.152

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, R. S., Lee, F. Y., Lee, S. D., Tsai, Y. T., Lin, H. C., Lu, R. H., et al. (1995). Endotoxemia in patients with chronic liver diseases: relationship to severity of liver diseases, presence of esophageal varices, and hyperdynamic circulation. J. Hepatol. 22, 165–172. doi: 10.1016/0168-8278(95)80424-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ling, Z., Jin, C., Xie, T., Cheng, Y., Li, L., and Wu, N. (2016). Alterations in the fecal microbiota of patients with HIV-1 infection: an observational study in a chinese population. Sci. Rep. 6:30673. doi: 10.1038/srep30673

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Li, L., Min, J., Wang, J., Wu, H., Zeng, Y., et al. (2012). Butyrate interferes with the differentiation and function of human monocyte-derived dendritic cells. Cell Immunol. 277, 66–73. doi: 10.1016/j.cellimm.2012.05.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S., da Cunha, A. P., Rezende, R. M., Cialic, R., Wei, Z., Bry, L., et al. (2016). The host shapes the gut microbiota via fecal microRNA. Cell Host Microbe 19, 32–43. doi: 10.1016/j.chom.2015.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S., and Weiner, H. L. (2016). Control of the gut microbiome by fecal microRNA. Microb. Cell. 3, 176–177. doi: 10.15698/mic2016.04.492

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, S. F., Ye, X., and Malik, A. B. (1999). Inhibition of NF-kappaB activation by pyrrolidine dithiocarbamate prevents In vivo expression of proinflammatory genes. Circulation 100, 1330–1337. doi: 10.1161/01.cir.100.12.1330

PubMed Abstract | CrossRef Full Text | Google Scholar

Livanos, A. E., Snider, E. J., Whittier, S., Chong, D. H., Wang, T. C., Abrams, J. A., et al. (2018). Rapid gastrointestinal loss of Clostridial Clusters IV and XIVa in the ICU associates with an expansion of gut pathogens. PLoS One. 13:e0200322. doi: 10.1371/journal.pone.0200322

PubMed Abstract | CrossRef Full Text | Google Scholar

Lorenzo-Zúñiga, V., Bartolí, R., Planas, R., Hofmann, A. F., Viñado, B., Hagey, L. R., et al. (2003). Oral bile acids reduce bacterial overgrowth, bacterial translocation, and endotoxemia in cirrhotic rats. Hepatology 37, 551–557. doi: 10.1053/jhep.2003.50116

PubMed Abstract | CrossRef Full Text | Google Scholar

Los, F. C. O., Randis, T. M., Aroian, R. V., and Ratner, A. J. (2013). Role of pore-forming toxins in bacterial infectious diseases. Microbiol. Mol. Biol. Rev. 77, 173–207. doi: 10.1128/MMBR.00052-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, H., Wu, Z., Xu, W., Yang, J., Chen, Y., and Li, L. (2011). Intestinal microbiota was assessed in cirrhotic patients with hepatitis B virus infection. Microb. Ecol. 61, 693–703. doi: 10.1007/s00248-010-9801-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Maa, M.-C., Chang, M. Y., Hsieh, M.-Y., Chen, Y.-J., Yang, C.-J., Chen, Z.-C., et al. (2010). Butyrate reduced lipopolysaccharide-mediated macrophage migration by suppression of Src enhancement and focal adhesion kinase activity. J. Nutr. Biochem. 21, 1186–1192. doi: 10.1016/j.jnutbio.2009.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

MacFie, J., O’Boyle, C., Mitchell, C. J., Buckley, P. M., Johnstone, D., and Sudworth, P. (1999). Gut origin of sepsis: a prospective study investigating associations between bacterial translocation, gastric microflora, and septic morbidity. Gut 45, 223–228. doi: 10.1136/gut.45.2.223

PubMed Abstract | CrossRef Full Text | Google Scholar

Macia, L., Tan, J., Vieira, A. T., Leach, K., Stanley, D., Luong, S., et al. (2015). Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 6:6734. doi: 10.1038/ncomms7734

PubMed Abstract | CrossRef Full Text | Google Scholar

Macpherson, A. J., Heikenwalder, M., and Ganal-Vonarburg, S. C. (2016). The liver at the nexus of host-microbial interactions. Cell Host Microbe 20, 561–571. doi: 10.1016/j.chom.2016.10.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Madan, J. C., Salari, R. C., Saxena, D., Davidson, L., O’Toole, G. A., Moore, J. H., et al. (2012). Gut microbial colonisation in premature neonates predicts neonatal sepsis. Arch. Dis. Child Fetal. Neonatal. Ed. 97, F456–F462. doi: 10.1136/fetalneonatal-2011-301373

PubMed Abstract | CrossRef Full Text | Google Scholar

Magro, F., Langner, C., Driessen, A., Ensari, A., Geboes, K., Mantzaris, G. J., et al. (2013). European consensus on the histopathology of inflammatory bowel disease. J. Crohn’s Colitis 7, 827–851. doi: 10.1016/j.crohns.2013.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Maharshak, N., Packey, C. D., Ellermann, M., Manick, S., Siddle, J. P., Huh, E. Y., et al. (2013). Altered enteric microbiota ecology in interleukin 10-deficient mice during development and progression of intestinal inflammation. Gut Microbes 4, 316–324. doi: 10.4161/gmic.25486

PubMed Abstract | CrossRef Full Text | Google Scholar

Mai, V., Torrazza, R. M., Ukhanova, M., Wang, X., Sun, Y., Li, N., et al. (2013). Distortions in development of intestinal microbiota associated with late onset sepsis in preterm infants. PLoS One 8:e52876. doi: 10.1371/journal.pone.0052876

PubMed Abstract | CrossRef Full Text | Google Scholar

Mainous, M. R., Ertel, W., Chaudry, I. H., and Deitch, E. A. (1995). The gut: a cytokine-generating organ in systemic inflammation? Shock 4, 193–199. doi: 10.1097/00024382-199509000-00007

PubMed Abstract | CrossRef Full Text | Google Scholar

Mainous, M. R., Tso, P., Berg, R. D., and Deitch, E. A. (1991). Studies of the route, magnitude, and time course of bacterial translocation in a model of systemic inflammation. Arch. Surg. 126, 33–37.

PubMed Abstract | Google Scholar

Makarov, S. S. (2001). NF-kappa B in rheumatoid arthritis: a pivotal regulator of inflammation, hyperplasia, and tissue destruction. Arthritis Res. 3, 200–206.

PubMed Abstract | Google Scholar

Marchetti, G., Tincati, C., and Silvestri, G. (2013). Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin. Microbiol. Rev. 26, 2–18. doi: 10.1128/CMR.00050-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Martín, R., Chamignon, C., Mhedbi-Hajri, N., Chain, F., Derrien, M., Escribano-Vázquez, U., et al. (2019). The potential probiotic Lactobacillus rhamnosus CNCM I-3690 strain protects the intestinal barrier by stimulating both mucus production and cytoprotective response. Sci. Rep. 9:5398. doi: 10.1038/s41598-019-41738-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Martin-Gallausiaux, C., Béguet-Crespel, F., Marinelli, L., Jamet, A., Ledue, F., Blottière, H. M., et al. (2018). Butyrate produced by gut commensal bacteria activates TGF-beta1 expression through the transcription factor SP1 in human intestinal epithelial cells. Sci. Rep. 8:9742. doi: 10.1038/s41598-018-28048-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Matthews, G. M., Howarth, G. S., and Butler, R. N. (2012). Short-chain fatty acids induce apoptosis in colon cancer cells associated with changes to intracellular redox state and glucose metabolism. Chemotherapy 58, 102–109. doi: 10.1159/000335672

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazmanian, S. K., Round, J. L., and Kasper, D. L. (2008). A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453, 620–625. doi: 10.1038/nature07008

PubMed Abstract | CrossRef Full Text | Google Scholar

McDonald, D., Ackermann, G., Khailova, L., Baird, C., Heyland, D., Kozar, R., et al. (2016). Extreme dysbiosis of the microbiome in critical illness. mSphere 1:e00199-16. doi: 10.1128/mSphere.00199-16

PubMed Abstract | CrossRef Full Text | Google Scholar

McGowan, I., Elliott, J., Fuerst, M., Taing, P., Boscardin, J., Poles, M., et al. (2004). Increased HIV-1 mucosal replication is associated with generalized mucosal cytokine activation. J. Acquir. Immune. Defic. Syndr. 37, 1228–1236. doi: 10.1097/01.qai.0000131846.12453.29

PubMed Abstract | CrossRef Full Text | Google Scholar

McManamy, M. E. M., Hakre, S., Verdin, E. M., and Margolis, D. M. (2014). Therapy for latent HIV-1 infection: the role of histone deacetylase inhibitors. Antivir. Chem. Chemother. 23, 145–149. doi: 10.3851/IMP2551

PubMed Abstract | CrossRef Full Text | Google Scholar

Meijer, K., De Vos, P., and Priebe, M. G. (2010). Butyrate and other short-chain fatty acids as modulators of immunity: what relevance for health? Curr. Opin. Clin. Nutr. Metab. Care. 13, 715–721. doi: 10.1097/MCO.0b013e32833eebe5

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng, M., Klingensmith, N. J., and Coopersmith, C. M. (2017). New insights into the gut as the driver of critical illness and organ failure. Curr. Opin. Crit. Care. 23, 143–148. doi: 10.1097/MCC.0000000000000386

PubMed Abstract | CrossRef Full Text | Google Scholar

Merlini, E., Bai, F., Bellistrì, G. M., Tincati, C., d’Arminio Monforte, A., and Marchetti, G. (2011). Evidence for polymicrobic flora translocating in peripheral blood of HIV-infected patients with poor immune response to antiretroviral therapy. PLoS One 6:e18580. doi: 10.1371/journal.pone.0018580

PubMed Abstract | CrossRef Full Text | Google Scholar

Mittal, R., and Coopersmith, C. M. (2014). Redefining the gut as the motor of critical illness. Trends Mol. Med. 20, 214–223. doi: 10.1016/j.molmed.2013.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Miura, K., Kodama, Y., Inokuchi, S., Schnabl, B., Aoyama, T., Ohnishi, H., et al. (2010). Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology 139, 323.e7–334.e7. doi: 10.1053/j.gastro.2010.03.052

PubMed Abstract | CrossRef Full Text | Google Scholar

Miura, K., and Ohnishi, H. (2014). Role of gut microbiota and Toll-like receptors in nonalcoholic fatty liver disease. World J. Gastroenterol. 20:7381. doi: 10.3748/wjg.v20.i23.7381

PubMed Abstract | CrossRef Full Text | Google Scholar

Mo, J.-S., Alam, K. J., Kim, H.-S., Lee, Y.-M., Yun, K.-J., and Chae, S.-C. (2016). MicroRNA 429 regulates mucin gene expression and secretion in murine model of colitis. J. Crohn’s Colitis 10, 837–849. doi: 10.1093/ecco-jcc/jjw033

PubMed Abstract | CrossRef Full Text | Google Scholar

Mohan, M., Kumar, V., Lackner, A. A., and Alvarez, X. (2015). Dysregulated miR-34a–SIRT1–acetyl p65 axis is a potential mediator of immune activation in the colon during chronic simian immunodeficiency virus infection of Rhesus macaques. J. Immunol. 194, 291–306. doi: 10.4049/jimmunol.1401447

CrossRef Full Text | Google Scholar

Nagpal, R., and Yadav, H. (2017). Bacterial translocation from the gut to the distant organs: an overview. Ann. Nutr. Metab. 71, 11–16. doi: 10.1159/000479918

PubMed Abstract | CrossRef Full Text | Google Scholar

Nahid, M. A., Satoh, M., and Chan, E. K. L. (2011). Mechanistic role of microRNA-146a in endotoxin-induced differential cross-regulation of TLR signaling. J. Immunol. 186, 1723–1734. doi: 10.4049/jimmunol.1002311

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakata, K., Sugi, Y., Narabayashi, H., Kobayakawa, T., Nakanishi, Y., Tsuda, M., et al. (2017). Commensal microbiota-induced microRNA modulates intestinal epithelial permeability through the small GTPase ARF4. J. Biol. Chem. 292, 15426–15433. doi: 10.1074/jbc.M117.788596

PubMed Abstract | CrossRef Full Text | Google Scholar

Nastasi, C., Candela, M., Bonefeld, C. M., Geisler, C., Hansen, M., Krejsgaard, T., et al. (2015). The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci. Rep. 5:16148. doi: 10.1038/srep16148

PubMed Abstract | CrossRef Full Text | Google Scholar

Nazli, A., Chan, O., Dobson-Belaire, W. N., Ouellet, M., Tremblay, M. J., Gray-Owen, S. D., et al. (2010). Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Pathog. 6:e1000852. doi: 10.1371/journal.ppat.1000852

PubMed Abstract | CrossRef Full Text | Google Scholar

Neff, C. P., Krueger, O., Xiong, K., Arif, S., Nusbacher, N., Schneider, J. M., et al. (2018). Fecal microbiota composition drives immune activation in HIV-infected individuals. EBioMedicine 30, 192–202. doi: 10.1016/j.ebiom.2018.03.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Nikaido, H. (1996). Multidrug Efflux Pumps of Gram-Negative Bacteria [Internet]. J. Bacteriol. 178, 5853–5859. doi: 10.1128/jb.178.20.5853-5859.1996

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishino, K., Latifi, T., and Groisman, E. A. (2006). Virulence and drug resistance roles of multidrug efflux systems of Salmonella enterica serovar Typhimurium. Mol. Microbiol. 59, 126–141. doi: 10.1111/j.1365-2958.2005.04940.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishitsuji, K., Xiao, J., Nagatomo, R., Umemoto, H., Morimoto, Y., Akatsu, H., et al. (2017). Analysis of the gut microbiome and plasma short-chain fatty acid profiles in a spontaneous mouse model of metabolic syndrome. Sci. Rep. 7:15876. doi: 10.1038/s41598-017-16189-5

PubMed Abstract | CrossRef Full Text | Google Scholar

O’Hara, A. M., and Shanahan, F. (2006). The gut flora as a forgotten organ. EMBO Rep. 7, 688–693. doi: 10.1038/sj.embor.7400731

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsson, J., Poles, M., Spetz, A., Elliott, J., Hultin, L., Giorgi, J., et al. (2000). Human immunodeficiency virus type 1 infection is associated with significant mucosal inflammation characterized by increased expression of CCR5, CXCR4, and β-chemokines. J. Infect Dis. 182, 1625–1635. doi: 10.1086/317625

PubMed Abstract | CrossRef Full Text | Google Scholar

Ortega, ÁD., Gonzalo-Asensio, J., and García-del Portillo, F. (2012). Dynamics of Salmonella small RNA expression in non-growing bacteria located inside eukaryotic cells. RNA Biol. 9, 469–488. doi: 10.4161/rna.19317

PubMed Abstract | CrossRef Full Text | Google Scholar

Ortiz, A. M., Klase, Z. A., DiNapoli, S. R., Vujkovic-Cvijin, I., Carmack, K., Perkins, M. R., et al. (2016). IL-21 and probiotic therapy improve Th17 frequencies, microbial translocation and microbiome in ARV-treated, SIV-infected macaques. Mucosal Immunol. 9, 458–467. doi: 10.1038/mi.2015.75

PubMed Abstract | CrossRef Full Text | Google Scholar

Padalon-Brauch, G., Hershberg, R., Elgrably-Weiss, M., Baruch, K., Rosenshine, I., Margalit, H., et al. (2008). Small RNAs encoded within genetic islands of Salmonella typhimurium show host-induced expression and role in virulence. Nucleic Acids Res. 36, 1913–1927. doi: 10.1093/nar/gkn050

PubMed Abstract | CrossRef Full Text | Google Scholar

Panda, S., khader, I., Casellas, F., López Vivancos, J., Cors, M., Santiago, A., et al. (2014). Short-term effect of antibiotics on human gut microbiota. PLoS One 9:e95476. doi: 10.1371/journal.pone.0095476

PubMed Abstract | CrossRef Full Text | Google Scholar

Pandiyan, P., Bhaskaran, N., Zou, M., Schneider, E., Jayaraman, S., and Huehn, J. (2019). Microbiome dependent regulation of Tregs and Th17 cells in mucosa. Front. Immunol. 10:426. doi: 10.3389/fimmu.2019.00426

PubMed Abstract | CrossRef Full Text | Google Scholar

Panigrahi, P., Parida, S., Nanda, N. C., Satpathy, R., Pradhan, L., Chandel, D. S., et al. (2017). A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature 548, 407–412. doi: 10.1038/nature23480

PubMed Abstract | CrossRef Full Text | Google Scholar

Panpetch, W., Chancharoenthana, W., Bootdee, K., Nilgate, S., Finkelman, M., Tumwasorn, S., et al. (2017). Lactobacillus rhamnosus L34 attenuates gut translocation-induced bacterial sepsis in murine models of leaky gut. Infect Immun. 86:e00700-17. doi: 10.1128/IAI.00700-17

PubMed Abstract | CrossRef Full Text | Google Scholar

Pant, K., Yadav, A. K., Gupta, P., Islam, R., Saraya, A., and Venugopal, S. K. (2017). Butyrate induces ROS-mediated apoptosis by modulating miR-22/SIRT-1 pathway in hepatic cancer cells. Redox Biol. 12, 340–349. doi: 10.1016/j.redox.2017.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Parada, J. L., Caron, C. R., Medeiros, A. B. P., and Soccol, C. R. (2007). Bacteriocins from lactic acid bacteria: purification, properties and use as biopreservatives. Brazilian Arch. Biol. Technol. 50, 512–542. doi: 10.1590/s1516-89132007000300018

CrossRef Full Text | Google Scholar

Park, K. C., Park, J. H., Jeon, J. Y., Kim, S. Y., Kim, J. M., Lim, C. Y., et al. (2014). A new histone deacetylase inhibitor improves liver fibrosis in BDL rats through suppression of hepatic stellate cells. Br. J. Pharmacol. 171, 4820–4830. doi: 10.1111/bph.12590

PubMed Abstract | CrossRef Full Text | Google Scholar

Peck, B. C. E., Sincavage, J., Feinstein, S., Mah, A. T., Simmons, J. G., Lund, P. K., et al. (2016). miR-30 Family Controls Proliferation and Differentiation of Intestinal Epithelial Cell Models by Directing a Broad Gene Expression Program That Includes SOX9 and the Ubiquitin Ligase Pathway. J. Biol. Chem. 291, 15975–15984. doi: 10.1074/jbc.M116.733733

PubMed Abstract | CrossRef Full Text | Google Scholar

Pegu, P., Helmus, R., Gupta, P., Tarwater, P., Caruso, L., Shen, C., et al. (2011). Induction of strong anti-HIV cellular immunity by a combination of Clostridium perfringens expressing HIV gag and virus like particles. Curr. HIV Res. 9, 613–622. doi: 10.2174/157016211798998808

PubMed Abstract | CrossRef Full Text | Google Scholar

Penheiter, K. L., Mathur, N., Giles, D., Fahlen, T., and Jones, B. D. (1997). Non-invasive Salmonella typhimurium mutants are avirulent because of an inability to enter and destroy M cells of ileal Peyer’s patches. Mol. Microbiol. 24, 697–709. doi: 10.1046/j.1365-2958.1997.3741745.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Pérez-Cobas, A. E., Gosalbes, M. J., Friedrichs, A., Knecht, H., Artacho, A., Eismann, K., et al. (2013). Gut microbiota disturbance during antibiotic therapy: a multi-omic approach. Gut 62, 1591–1601. doi: 10.1136/gutjnl-2012-303184

PubMed Abstract | CrossRef Full Text | Google Scholar

Phillips, A. D., Navabpour, S., Hicks, S., Dougan, G., Wallis, T., and Frankel, G. (2000). Enterohaemorrhagic Escherichia coli O157:H7 target Peyer’s patches in humans and cause attaching/effacing lesions in both human and bovine intestine. Gut 47, 377–381. doi: 10.1136/gut.47.3.377

PubMed Abstract | CrossRef Full Text | Google Scholar

Pinto-Cardoso, S., Klatt, N. R., and Reyes-Terán, G. (2018). Impact of antiretroviral drugs on the microbiome: unknown answers to important questions. Curr. Opin. HIV AIDS 13, 53–60. doi: 10.1097/COH.0000000000000428

PubMed Abstract | CrossRef Full Text | Google Scholar

Pinto-Cardoso, S., Lozupone, C., Briceño, O., Alva-Hernández, S., Téllez, N., Adriana, A., et al. (2017). Fecal bacterial communities in treated HIV infected individuals on two antiretroviral regimens. Sci. Rep. 7:43741. doi: 10.1038/srep43741

PubMed Abstract | CrossRef Full Text | Google Scholar

Pollicino, T., Belloni, L., Raffa, G., Pediconi, N., Squadrito, G., Raimondo, G., et al. (2006). Hepatitis B virus replication is regulated by the acetylation status of hepatitis B virus cccDNA-bound H3 and H4 histones. Gastroenterology 130, 823–837. doi: 10.1053/j.gastro.2006.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Ponziani, F. R., Gerardi, V., Pecere, S., D’Aversa, F., Lopetuso, L., Zocco, M. A., et al. (2015). Effect of rifaximin on gut microbiota composition in advanced liver disease and its complications. World J. Gastroenterol. 21:12322. doi: 10.3748/wjg.v21.i43.12322

PubMed Abstract | CrossRef Full Text | Google Scholar

Pulendran, B., Kumar, P., Cutler, C. W., Mohamadzadeh, M., Van Dyke, T., Banchereau, J., et al. (2001). Lipopolysaccharides from distinct pathogens induce different classes of immune responses in vivo. J. Immunol. 167, 5067–5076. doi: 10.4049/jimmunol.167.9.5067

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajilić-Stojanović, M., Smidt, H., and de Vos, W. M. (2007). Diversity of the human gastrointestinal tract microbiota revisited. Environ. Microbiol. 9, 2125–2136. doi: 10.1111/j.1462-2920.2007.01369.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Rao, K., and Young, V. B. (2015). Fecal microbiota transplantation for the management of Clostridium difficile infection. Infect Dis. Clin. North Am. 29, 109–122. doi: 10.1016/j.idc.2014.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Rea, M. C., Sit, C. S., Clayton, E., O’Connor, P. M., Whittal, R. M., Zheng, J., et al. (2010). Thuricin CD, a posttranslationally modified bacteriocin with a narrow spectrum of activity against Clostridium difficile. Proc. Natl. Acad. Sci. 107, 9352–9357. doi: 10.1073/pnas.0913554107

PubMed Abstract | CrossRef Full Text | Google Scholar

Rhayat, L., Maresca, M., Nicoletti, C., Perrier, J., Brinch, K. S., Christian, S., et al. (2019). Effect of Bacillus subtilis strains on intestinal barrier function and inflammatory response. Front. Immunol. 10:564. doi: 10.3389/fimmu.2019.00564

PubMed Abstract | CrossRef Full Text | Google Scholar

Richer, M. J., Nolz, J. C., and Harty, J. T. (2013). Pathogen-specific inflammatory milieux tune the antigen sensitivity of CD8+ T cells by enhancing T cell receptor signaling. Immunity 38, 140–152. doi: 10.1016/j.immuni.2012.09.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Ridlon, J. M., Kang, D. J., Hylemon, P. B., and Bajaj, J. S. (2014). Bile acids and the gut microbiome. Curr. Opin. Gastroenterol. 30, 332–338. doi: 10.1097/mog.0000000000000057

PubMed Abstract | CrossRef Full Text | Google Scholar

Rios, D., Wood, M. B., Li, J., Chassaing, B., Gewirtz, A. T., and Williams, I. R. (2016). Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria. Mucosal Immunol. 9, 907–916. doi: 10.1038/mi.2015.121

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez, A., Vigorito, E., Clare, S., Warren, M. V., Couttet, P., Soond, D. R., et al. (2007). Requirement of bic/microRNA-155 for normal immune function. Science 316, 608–611. doi: 10.1126/science.1139253

PubMed Abstract | CrossRef Full Text | Google Scholar

Roediger, W. E. (1980). Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 21, 793–798. doi: 10.1136/gut.21.9.793

PubMed Abstract | CrossRef Full Text | Google Scholar

Rolhion, N., and Chassaing, B. (2016). When pathogenic bacteria meet the intestinal microbiota. Philos. Trans. R. Soc. B Biol. Sci. 371:20150504. doi: 10.1098/rstb.2015.0504

PubMed Abstract | CrossRef Full Text | Google Scholar

Román, E., Nieto, J. C., Gely, C., Vidal, S., Pozuelo, M., Poca, M., et al. (2019). Effect of a multistrain probiotic on cognitive function and risk of falls in patients with cirrhosis: a randomized trial. Hepatol. Commun. 3, 632–645. doi: 10.1002/hep4.1325

PubMed Abstract | CrossRef Full Text | Google Scholar

Round, J. L., Lee, S. M., Li, J., Tran, G., Jabri, B., Chatila, T. A., et al. (2011). The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332, 974–977. doi: 10.1126/science.1206095

PubMed Abstract | CrossRef Full Text | Google Scholar

Sailhamer, E. A., Carson, K., Chang, Y., Zacharias, N., Spaniolas, K., Tabbara, M., et al. (2009). Fulminant Clostridium difficile colitis. Arch. Surg. 144, 433–439. doi: 10.1001/archsurg.2009.51

PubMed Abstract | CrossRef Full Text | Google Scholar

Sankaran, S., George, M. D., Reay, E., Guadalupe, M., Flamm, J., Prindiville, T., et al. (2008). Rapid onset of intestinal epithelial barrier dysfunction in primary human immunodeficiency virus infection is driven by an imbalance between immune response and mucosal repair and regeneration. J. Virol. 82, 538–545. doi: 10.1128/jvi.01449-07

PubMed Abstract | CrossRef Full Text | Google Scholar

Sankaran, S., Guadalupe, M., Reay, E., George, M. D., Flamm, J., Prindiville, T., et al. (2005). Gut mucosal T cell responses and gene expression correlate with protection against disease in long-term HIV-1-infected nonprogressors. Proc. Natl. Acad. Sci. U.S.A. 102, 9860–9865. doi: 10.1073/pnas.0503463102

PubMed Abstract | CrossRef Full Text | Google Scholar

Santoro, M. G., Rossi, A., Amici, C., and Schneider, R. J. (2003). NF-kappaB and virus infection: who controls whom. EMBO J. 22, 2552–2560. doi: 10.1093/emboj/cdg267

PubMed Abstract | CrossRef Full Text | Google Scholar

Sassone-Corsi, M., and Raffatellu, M. (2015). No vacancy: How beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. J. Immunol. 194, 4081–4087. doi: 10.4049/jimmunol.1403169

PubMed Abstract | CrossRef Full Text | Google Scholar

Sato, Y., and Tsurumi, T. (2013). Genome guardian p53 and viral infections. Rev. Med. Virol. 23, 213–220. doi: 10.1002/rmv.1738

PubMed Abstract | CrossRef Full Text | Google Scholar

Schnabl, B. (2013). Linking intestinal homeostasis and liver disease. Curr. Opin. Gastroenterol. 29, 264–270. doi: 10.1097/mog.0b013e32835ff948

PubMed Abstract | CrossRef Full Text | Google Scholar

Schnabl, B., and Brenner, D. A. (2014). Interactions between the intestinal microbiome and liver diseases. Gastroenterology 146, 1513–1524. doi: 10.1053/j.gastro.2014.01.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Schnupf, P., Gaboriau-Routhiau, V., Sansonetti, P. J., and Cerf-Bensussan, N. (2017). Segmented filamentous bacteria, Th17 inducers and helpers in a hostile world. Curr. Opin. Microbiol. 35, 100–109. doi: 10.1016/j.mib.2017.03.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Seki, E., De Minicis, S., Österreicher, C. H., Kluwe, J., Osawa, Y., Brenner, D. A., et al. (2007). TLR4 enhances TGF-β signaling and hepatic fibrosis. Nat. Med. 13, 1324–1332. doi: 10.1038/nm1663

PubMed Abstract | CrossRef Full Text | Google Scholar

Seki, E., and Schnabl, B. (2012). Role of innate immunity and the microbiota in liver fibrosis: crosstalk between the liver and gut. J. Physiol. 590, 447–458. doi: 10.1113/jphysiol.2011.219691

PubMed Abstract | CrossRef Full Text | Google Scholar

Shao, Y., Li, J., Cai, Y., Xie, Y., Ma, G., Li, Y., et al. (2014). The functional polymorphisms of miR-146a are associated with susceptibility to severe sepsis in the Chinese population. Mediators Inflamm. 2014:916202. doi: 10.1155/2014/916202

PubMed Abstract | CrossRef Full Text | Google Scholar

Shimizu, K., Ogura, H., Goto, M., Asahara, T., Nomoto, K., Morotomi, M., et al. (2006). Altered gut flora and environment in patients with severe SIRS. J. Trauma 60, 126–133. doi: 10.1097/01.ta.0000197374.99755.fe

PubMed Abstract | CrossRef Full Text | Google Scholar

Shimizu, K., Ogura, H., Hamasaki, T., Goto, M., Tasaki, O., Asahara, T., et al. (2011). Altered gut flora are associated with septic complications and death in critically ill patients with systemic inflammatory response syndrome. Dig. Dis. Sci. 56, 1171–1177. doi: 10.1007/s10620-010-1418-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Shirai, Y., Hashimoto, M., Kato, R., Kawamura, Y. I., Kirikae, T., Yano, H., et al. (2004). Lipopolysaccharide induces CD25-Positive, IL-10-producing lymphocytes without secretion of proinflammatory cytokines in the human colon: low MD-2 mRNA expression in colonic macrophages. J. Clin. Immunol. 24, 42–52. doi: 10.1023/b:joci.0000018062.01980.ba

PubMed Abstract | CrossRef Full Text | Google Scholar

Shirakawa, K., Chavez, L., Hakre, S., Calvanese, V., and Verdin, E. (2013). Reactivation of latent HIV by histone deacetylase inhibitors. Trends Microbiol. 21, 277–285. doi: 10.1016/j.tim.2013.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Singer, M., Deutschman, C. S., Seymour, C. W., Shankar-Hari, M., Annane, D., Bauer, M., et al. (2016). The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA 315, 801–810.

Google Scholar

Singh, N., Shirdel, E. A., Waldron, L., Zhang, R.-H., Jurisica, I., and Comelli, E. M. (2012). The murine caecal microRNA signature depends on the presence of the endogenous microbiota. Int. J. Biol. Sci. 8, 171–186. doi: 10.7150/ijbs.8.171

PubMed Abstract | CrossRef Full Text | Google Scholar

Skirecki, T., and Cavaillon, J.-M. (2019). Inner sensors of endotoxin – implications for sepsis research and therapy. FEMS Microbiol. Rev. 43, 239–256. doi: 10.1093/femsre/fuz004

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, M., Su, H., Zhang, L., Ma, J., Li, J., Pan, K., et al. (2013). Genetic polymorphisms of miR-146a and miR-27a, H. pylori infection, and risk of gastric lesions in a chinese population. PLoS One 8:e61250. doi: 10.1371/journal.pone.0061250

PubMed Abstract | CrossRef Full Text | Google Scholar

Sonnen, A. F.-P., and Henneke, P. (2013). Role of pore-forming toxins in neonatal sepsis. Clin. Dev. Immunol. 2013:608456. doi: 10.1155/2013/608456

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorbara, M. T., and Pamer, E. G. (2018). Interbacterial mechanisms of colonization resistance and the strategies pathogens use to overcome them. Mucosal Immunol. 12, 1–9. doi: 10.1038/s41385-018-0053-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorbara, M. T., and Pamer, E. G. (2019). Interbacterial mechanisms of colonization resistance and the strategies pathogens use to overcome them. Mucosal Immunol. 12, 1–9. doi: 10.1038/s41385-018-0053-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorini, C., Cardoso, R. F., Gagliani, N., and Villablanca, E. J. (2018). Commensal bacteria-specific CD4+ T cell responses in health and disease. Front. Immunol. 9:2667. doi: 10.3389/fimmu.2018.02667

PubMed Abstract | CrossRef Full Text | Google Scholar

Souza, D. G., Vieira, A. T., Soares, A. C., Pinho, V., Nicoli, J. R., Vieira, L. Q., et al. (2004). The essential role of the intestinal microbiota in facilitating acute inflammatory responses. J. Immunol. 173, 4137–4146. doi: 10.4049/jimmunol.173.6.4137

PubMed Abstract | CrossRef Full Text | Google Scholar

Srinivasjois, R., Rao, S., and Patole, S. (2013). Prebiotic supplementation in preterm neonates: updated systematic review and meta-analysis of randomised controlled trials. Clin. Nutr. 32, 958–965. doi: 10.1016/j.clnu.2013.05.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Staedel, C., and Darfeuille, F. (2013). MicroRNAs and bacterial infection. Cell Microbiol. 15, 1496–1507. doi: 10.1111/cmi.12159

PubMed Abstract | CrossRef Full Text | Google Scholar

Stagg, A. J., Hart, A. L., Knight, S. C., and Kamm, M. A. (2003). The dendritic cell: its role in intestinal inflammation and relationship with gut bacteria. Gut 52, 1522–1529. doi: 10.1136/gut.52.10.1522

PubMed Abstract | CrossRef Full Text | Google Scholar

Steele, A. K., Lee, E. J., Vestal, B., Hecht, D., Dong, Z., Rapaport, E., et al. (2014). Contribution of intestinal barrier damage, microbial translocation and HIV-1 infection status to an inflammaging signature. PLoS One 9:e97171. doi: 10.1371/journal.pone.0097171

PubMed Abstract | CrossRef Full Text | Google Scholar

Stewart, A. S., Pratt-Phillips, S., and Gonzalez, L. M. (2017). Alterations in intestinal permeability: the role of the “Leaky Gut” in health and disease. J. Equine Vet. Sci. 52, 10–22. doi: 10.1016/j.jevs.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Stewart, C. J., Embleton, N. D., Marrs, E. C. L., Smith, D. P., Fofanova, T., Nelson, A., et al. (2017). Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls. Microbiome 5:75. doi: 10.1186/s40168-017-0295-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, G. L., Klein, R. D., Aminlari, A., Zhang, H. Y., Steinstraesser, L., Alarcon, W. H., et al. (2000). Kupffer cell activation by lipopolysaccharide in rats: role for lipopolysaccharide binding protein and toll-like receptor 4. Hepatology 31, 932–936. doi: 10.1053/he.2000.5634

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, B., Jia, Y., Hong, J., Sun, Q., Gao, S., Hu, Y., et al. (2018). Sodium butyrate ameliorates high-fat-diet-induced non-alcoholic fatty liver disease through peroxisome proliferator-activated receptor α-mediated activation of β oxidation and suppression of inflammation. J. Agric. Food Chem. 66, 7633–7642. doi: 10.1021/acs.jafc.8b01189

PubMed Abstract | CrossRef Full Text | Google Scholar

Taft, D. H., Ambalavanan, N., Schibler, K. R., Yu, Z., Newburg, D. S., Deshmukh, H., et al. (2015). Center variation in intestinal microbiota prior to late-onset sepsis in preterm infants. PLoS One 10:e0130604. doi: 10.1371/journal.pone.0130604

PubMed Abstract | CrossRef Full Text | Google Scholar

Taganov, K. D., Boldin, M. P., Chang, K.-J., and Baltimore, D. (2006). NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. U.S.A 103, 12481–12486. doi: 10.1073/pnas.0605298103

PubMed Abstract | CrossRef Full Text | Google Scholar

Taguwa, S., Kambara, H., Omori, H., Tani, H., Abe, T., Mori, Y., et al. (2009). Cochaperone activity of human butyrate-induced transcript 1 facilitates hepatitis C virus replication through an Hsp90-dependent pathway. J. Virol. 83, 10427–10436. doi: 10.1128/JVI.01035-09

PubMed Abstract | CrossRef Full Text | Google Scholar

Taguwa, S., Okamoto, T., Abe, T., Mori, Y., Suzuki, T., Moriishi, K., et al. (2008). Human butyrate-induced transcript 1 interacts with hepatitis C virus NS5A and regulates viral replication. J. Virol. 82, 2631–2641. doi: 10.1128/jvi.02153-07

PubMed Abstract | CrossRef Full Text | Google Scholar

Takeda, N., Jain, R., LeBoeuf, M. R., Wang, Q., Lu, M. M., and Epstein, J. A. (2011). Interconversion between intestinal stem cell populations in distinct niches. Science 334, 1420–1424. doi: 10.1126/science.1213214

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, Y., Chen, Y., Jiang, H., Robbins, G. T., and Nie, D. (2011). G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int. J. Cancer 128, 847–856. doi: 10.1002/ijc.25638

PubMed Abstract | CrossRef Full Text | Google Scholar

Todorov, S. D., Franco, B. D. G. M., and Wiid, I. J. (2014). In vitro study of beneficial properties and safety of lactic acid bacteria isolated from Portuguese fermented meat products. Benef. Microbes 5, 351–366. doi: 10.3920/BM2013.0030

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulven, T. (2012). Short-chain free fatty acid receptors FFA2/GPR43 and FFA3/GPR41 as new potential therapeutic targets. Front. Endocrinol. 3:111. doi: 10.3389/fendo.2012.00111

PubMed Abstract | CrossRef Full Text | Google Scholar

Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L., and Hooper, L. V. (2008). Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc. Natl. Acad. Sci. U.S.A. 105, 20858–20863. doi: 10.1073/pnas.0808723105

PubMed Abstract | CrossRef Full Text | Google Scholar

Vdovikova, S., Gilfillan, S., Wang, S., Dongre, M., Wai, S. N., and Hurtado, A. (2018). Modulation of gene transcription and epigenetics of colon carcinoma cells by bacterial membrane vesicles. Sci. Rep. 8, 7434. doi: 10.1038/s41598-018-25308-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Veazey, R. S., DeMaria, M., Chalifoux, L. V., Shvetz, D. E., Pauley, D. R., Knight, H. L., et al. (1998). Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science 280, 427–431. doi: 10.1126/science.280.5362.427

PubMed Abstract | CrossRef Full Text | Google Scholar

Villagra, A., Sotomayor, E. M., and Seto, E. (2010). Histone deacetylases and the immunological network: implications in cancer and inflammation. Oncogene 29, 157–173. doi: 10.1038/onc.2009.334

PubMed Abstract | CrossRef Full Text | Google Scholar

Vincent, C., Stephens, D. A., Loo, V. G., Edens, T. J., Behr, M. A., Dewar, K., et al. (2013). Reductions in intestinal Clostridiales precede the development of nosocomial Clostridium difficile infection. Microbiome 1:18. doi: 10.1186/2049-2618-1-18

PubMed Abstract | CrossRef Full Text | Google Scholar

Vinolo, M. A. R., Rodrigues, H. G., Hatanaka, E., Sato, F. T., Sampaio, S. C., and Curi, R. (2011). Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J. Nutr. Biochem. 22, 849–855. doi: 10.1016/j.jnutbio.2010.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Vora, P., Youdim, A., Thomas, L. S., Fukata, M., Tesfay, S. Y., Lukasek, K., et al. (2004). Beta-defensin-2 expression is regulated by TLR signaling in intestinal epithelial cells. J. Immunol. 173, 5398–5405. doi: 10.4049/jimmunol.173.9.5398

PubMed Abstract | CrossRef Full Text | Google Scholar

Vujkovic-Cvijin, I., Rutishauser, R. L., Pao, M., Hunt, P. W., Lynch, S. V., McCune, J. M., et al. (2017). Limited engraftment of donor microbiome via one-time fecal microbial transplantation in treated HIV-infected individuals. Gut Microbes 8, 440–450. doi: 10.1080/19490976.2017.1334034

PubMed Abstract | CrossRef Full Text | Google Scholar

Wallace, A. J., Stillman, T. J., Atkins, A., Jamieson, S. J., Bullough, P. A., Green, J., et al. (2000). E. coli hemolysin E (HlyE, ClyA, SheA): X-ray crystal structure of the toxin and observation of membrane pores by electron microscopy. Cell 100, 265–276. doi: 10.1016/s0092-8674(00)81564-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, F., Liu, J., Weng, T., Shen, K., Chen, Z., Yu, Y., et al. (2017). The inflammation induced by lipopolysaccharide can be mitigated by short-chain fatty acid, butyrate, through upregulation of IL-10 in septic shock. Scand. J. Immunol. 85, 258–263. doi: 10.1111/sji.12515

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H.-B., Wang, P.-Y., Wang, X., Wan, Y.-L., and Liu, Y.-C. (2012). Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein claudin-1 transcription. Dig. Dis. Sci. 57, 3126–3135. doi: 10.1007/s10620-012-2259-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Weber, T. E., and Kerr, B. J. (2006). Butyrate differentially regulates cytokines and proliferation in porcine peripheral blood mononuclear cells. Vet. Immunol. Immunopathol. 113, 139–147. doi: 10.1016/j.vetimm.2006.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, Y., Yang, J., Wang, J., Yang, Y., Huang, J., Gong, H., et al. (2016). Successful treatment with fecal microbiota transplantation in patients with multiple organ dysfunction syndrome and diarrhea following severe sepsis. Crit. Care 20:332.

PubMed Abstract | Google Scholar

Wiest, R., Albillos, A., Trauner, M., Bajaj, J. S., and Jalan, R. (2017). Targeting the gut-liver axis in liver disease. J. Hepatol. 67, 1084–1103. doi: 10.1016/j.jhep.2017.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Williams, M. R., Stedtfeld, R. D., Tiedje, J. M., and Hashsham, S. A. (2017). MicroRNAs-Based Inter-Domain Communication between the Host and Members of the Gut Microbiome. Front. Microbiol. 8:1896. doi: 10.3389/fmicb.2017.01896

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, F., Zhang, S., Dassopoulos, T., Harris, M. L., Bayless, T. M., Meltzer, S. J., et al. (2010). Identification of microRNAs associated with ileal and colonic Crohn’s disease. Inflamm. Bowel Dis. 16, 1729–1738. doi: 10.1002/ibd.21267

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Z.-W., Lu, H.-F., Wu, J., Zuo, J., Chen, P., Sheng, J.-F., et al. (2012). Assessment of the fecal lactobacilli population in patients with hepatitis B virus-related decompensated cirrhosis and hepatitis B cirrhosis treated with liver transplant, Microbial Ecology. New York, NY: Springer, 929–937.

Google Scholar

Xue, X., Cao, A. T., Cao, X., Yao, S., Carlsen, E. D., Soong, L., et al. (2014). Downregulation of microRNA-107 in intestinal CD11c + myeloid cells in response to microbiota and proinflammatory cytokines increases IL-23p19 expression. Eur. J. Immunol. 44, 673–682. doi: 10.1002/eji.201343717

PubMed Abstract | CrossRef Full Text | Google Scholar

Xue, X., Feng, T., Yao, S., Wolf, K. J., Liu, C.-G., Liu, X., et al. (2011). Microbiota downregulates dendritic cell expression of miR-10a, which targets IL-12/IL-23p40. J. Immunol. 187, 5879–5886. doi: 10.4049/jimmunol.1100535

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, L., and Seki, E. (2012). Toll-like receptors in liver fibrosis: cellular crosstalk and mechanisms. Front. Physiol. 3:138. doi: 10.3389/fphys.2012.00138

PubMed Abstract | CrossRef Full Text | Google Scholar

Ye, D., Guo, S., Al-Sadi, R., and Ma, T. Y. (2011). MicroRNA regulation of intestinal epithelial tight junction permeability. Gastroenterology 141, 1323–1333. doi: 10.1053/j.gastro.2011.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Ye, F., and Karn, J. (2015). Bacterial short chain fatty acids push all the buttons needed to reactivate latent viruses. Stem Cell Epigenet. 2:e532.

PubMed Abstract | Google Scholar

Yin, L., Laevsky, G., and Giardina, C. (2001). Butyrate suppression of colonocyte NF-kappa B activation and cellular proteasome activity. J. Biol. Chem. 276, 44641–44646. doi: 10.1074/jbc.m105170200

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoseph, B. P., Klingensmith, N. J., Liang, Z., Breed, E. R., Burd, E. M., Mittal, R., et al. (2016). Mechanisms of intestinal barrier dysfunction in sepsis. Shock 46, 52–59. doi: 10.1097/SHK.0000000000000565

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, C., Burns, M. B., Subramanian, S., and Blekhman, R. (2018). Interaction between host microRNAs and the gut microbiota in colorectal cancer. mSystems 3:e00205-17. doi: 10.1128/mSystems.00205-17

PubMed Abstract | CrossRef Full Text | Google Scholar

Zaborina, O., Lepine, F., Xiao, G., Valuckaite, V., Chen, Y., Li, T., et al. (2007). Dynorphin activates quorum sensing quinolone signaling in Pseudomonas aeruginosa. PLoS Pathog. 3:e35. doi: 10.1371/journal.ppat.0030035

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeitz, M., Ullrich, R., Schneider, T., Kewenig, S., Hohloch, K., and Riecken, E. O. (1998). HIV/SIV enteropathy. Ann. N.Y. Acad. Sci. 859, 139–148. doi: 10.1111/j.1749-6632.1998.tb11118.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, L., Shen, J., Cheng, J., and Fan, X. (2015). MicroRNA-21 regulates intestinal epithelial tight junction permeability. Cell Biochem. Funct. 33, 235–240. doi: 10.1002/cbf.3109

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, C., Zhou, Z., Zhang, T., Liu, F., Zhang, C.-Y., Zen, K., et al. (2017). Salmonella small RNA fragment Sal-1 facilitates bacterial survival in infected cells via suppressing iNOS induction in a microRNA manner. Sci. Rep. 7:16979. doi: 10.1038/s41598-017-17205-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, D., Pan, Q., Shen, F., Cao, H., Ding, W., Chen, Y., et al. (2017a). Total fecal microbiota transplantation alleviates high-fat diet-induced steatohepatitis in mice via beneficial regulation of gut microbiota. Sci. Rep. 7:1529. doi: 10.1038/s41598-017-01751-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, D., Pan, Q., Xin, F.-Z., Zhang, R.-N., He, C.-X., Chen, G.-Y., et al. (2017b). Sodium butyrate attenuates high-fat diet-induced steatohepatitis in mice by improving gut microbiota and gastrointestinal barrier. World J. Gastroenterol. 23:60. doi: 10.3748/wjg.v23.i1.60

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q., Costinean, S., Croce, C. M., Brasier, A. R., Merwat, S., Larson, S. A., et al. (2015). MicroRNA 29 targets nuclear Factor-κB–repressing factor and claudin 1 to increase intestinal permeability. Gastroenterology 148, 158.e–169.e.

Google Scholar

Zhou, Q., Souba, W. W., Croce, C. M., and Verne, G. N. (2010). MicroRNA-29a regulates intestinal membrane permeability in patients with irritable bowel syndrome. Gut 59, 775–784. doi: 10.1136/gut.2009.181834

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: dysbiosis, intestinal microbiota, epigenetic modulation, short chain fatty acids, immunity, sepsis, HIV infection, liver infections

Citation: Iacob S and Iacob DG (2019) Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front. Microbiol. 10:1676. doi: 10.3389/fmicb.2019.01676

Received: 14 January 2019; Accepted: 08 July 2019;
Published: 07 August 2019.

Edited by:

Sudhanshu Shekhar, University of Oslo, Norway

Reviewed by:

Elisabeth Billard, Université Clermont Auvergne, France
Eliana Mariño, Monash University, Australia

Copyright © 2019 Iacob and Iacob. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Diana Gabriela Iacob, dianagiacob@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.