Next Article in Journal
Nitrate and Nitrite Metabolism in Aging Rats: A Comparative Study
Previous Article in Journal
Lactobacillus casei and Its Supplement Alleviate Stress-Induced Depression and Anxiety in Mice by the Regulation of BDNF Expression and NF-κB Activation
Previous Article in Special Issue
Is Autologous Fecal Microbiota Transfer after Exclusive Enteral Nutrition in Pediatric Crohn’s Disease Patients Rational and Feasible? Data from a Feasibility Test
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Dietary Fibre in Enteral Nutrition in Sepsis Prevention and Therapy: A Narrative Review

by
Valentina V. Huwiler
1,2,†,
Melanie Scalise
3,4,†,
Katja A. Schönenberger
1,2,
Stefan Mühlebach
2,
Zeno Stanga
1 and
Maria L. Balmer
1,3,4,*
1
Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism (UDEM), Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
2
Division of Clinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, 4031 Basel, Switzerland
3
Department of Biomedical Research, University Clinic of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
4
Diabetes Center Berne (DCB), 3010 Bern, Switzerland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2023, 15(11), 2489; https://doi.org/10.3390/nu15112489
Submission received: 15 May 2023 / Revised: 22 May 2023 / Accepted: 23 May 2023 / Published: 26 May 2023
(This article belongs to the Special Issue Current State of the Art and Practical Aspects of Enteral Nutrition)

Abstract

:
Objective: This narrative review summarises the current evidence on the role of dietary fibre in enteral nutrition in the prevention and therapy of sepsis, with a focus on critically ill patients. The aim is to discuss the implications for clinical practice and identify future directions for policy and research. Resources: We searched MEDLINE and Google Scholar for records on sepsis, critically ill, enteral nutrition, and dietary fibre. We included all types of articles such as meta-analyses, reviews, clinical trials, preclinical studies, and in vitro studies. Data were evaluated for significance and clinical relevance. Synopsis of Review: Despite the ongoing debate, enteral nutrition containing dietary fibres showed great potential in attenuating sepsis-related outcomes and preventing the incidence of sepsis in critically ill patients on enteral nutrition. Dietary fibres target different underlying mechanisms such as microbiota, mucosal barrier integrity, local cellular immune response, and systemic inflammation. We discuss the clinical potential and concerns that currently exist with the standard implementation of dietary fibre in enterally fed intensive care patients. Additionally, we identified research gaps that should be addressed to determine effectiveness and the role of dietary fibres in sepsis itself and its associated outcomes.

1. Introduction

Sepsis is a complex and “life-threatening organ dysfunction caused by a dysregulated host response to infection” that can lead to septic shock, multiple organ failure, and death [1]. It is the leading cause of mortality in intensive care units (ICUs), with nearly one in four cases being fatal, and is responsible for nearly 20% of deaths worldwide. In 2017, 48.9 million cases of sepsis were recorded worldwide, making it a significant global health burden. The most common causes of sepsis are infections, accounting for 68.9% of the cases, followed by non-communicable diseases (27.5%) and injuries (3.6%) [2].
Sepsis prevention and treatment strategies differ depending on the stage and course of the occurrence. The primary goal of sepsis prevention is to avoid infections. This can be achieved through strategies such as safe food preparation, improving sanitation and water quality, and adequate nutrition. In case of infections, early detection and prompt treatment (e.g., with antibiotics) are essential. This includes the prevention of microbial translocation through disrupted physiological barriers such as the mucosal membrane of the gastrointestinal tract. In the case of translocation, it is essential to halt the course of sepsis by ensuring an appropriate host response. Signs and symptoms of sepsis may vary depending on the patient and the course of the disease and include fever or low temperature, altered mental status, difficulty breathing, increased heart rate, and weak pulse/low blood pressure [3].
Critically ill patients are prone to sepsis and show complex and fluctuating immune and inflammatory changes. Malnutrition occurs frequently because of increased catabolism in the early period and augmented anabolism in the late period. In such cases, enteral nutrition (EN), which provides nutrients through the gastrointestinal tract, can be an effective way to cover the nutritional requirements of these patients [4]. Dietary fibres (DFs) are a type of indigestible carbohydrate that can be fermented by the gut microbiota to produce short-chain fatty acids (SCFAs), among others, which have pleiotropic beneficial effects on the body [5]. Due to the stimulating effect on protective microbial populations, some DFs can be classified as prebiotics. Prebiotics are often administered together with beneficial bacteria, i.e., probiotics, which are summarised under the term synbiotics [6]. Evidence is accumulating that DFs, as a component of EN, may have protective and therapeutic effects in sepsis. However, their effectiveness, safety, and generalisability remain controversial. This narrative review aims to explore the existing evidence on the role of DFs in EN and its potential to prevent and treat sepsis.

2. Methods

This narrative review was based on a literature search of the MEDLINE and Google Scholar databases to identify relevant studies on sepsis, critically ill, EN, and DFs from inception to September 2022. The search terms included the keywords sepsis, critically ill, bacteraemia, fungaemia, viraemia, bloodstream infection, enteral nutrition, tube feeding, dietary fibre, prebiotic, dietary carbohydrate, wheat bran, roughage, resistant starch, inulin, arabinoxylan, xylan, pectin, beta-glucan, guar gum, arabica gum, oligosaccharide, oligofructose, inulin, non-starch polysaccharides, soy polysaccharides, lignin, cellulose, pea fibre, and inulin-type fructans (Appendix A). The types of articles reviewed in this paper covered clinical trials, animal studies, in vitro examinations, reviews, and meta-analyses. We favoured the most clinically accurate literature addressing our research question and up-to-date articles. We included preclinical studies in case no human clinical evidence was available or to underline mechanistic aspects. Studies with a mixed intervention that included a potential confounder in addition to DFs were excluded if a study was found that examined the isolated effect of DFs. Similarly, studies that included other forms of nutrition, such as oral or parenteral nutrition, were excluded if we found a study that examined the effect of enteral nutrition on the same research question. Case studies and clinical trials were excluded if a recent meta-analysis was published on the same research question. Literature for the mode of action was not restricted to search terms to prevent the exclusion of relevant studies.

3. Impact of Dietary Fibres on Intestinal Homeostasis

A cascade of negative effects including infection, intestinal microbiome dysbiosis, microbial translocation, and dysregulated host immune response may result in sepsis. The role of DFs as part of EN during sepsis could be due to several mechanisms, which are discussed in this section (Figure 1, Table 1). DFs can perform the following functions:
  • Shaping the microbiome composition, diversity, and function [7];
  • Supporting the intestinal barrier integrity [8,9,10];
  • Regulating immune responses in intestinal tissue [10,11];
  • Mediating systemic inflammation [10,11].
Table 1. Overview of studies and meta-analyses on dietary fibre supplementation in enteral nutrition and intestinal homeostasis.
Table 1. Overview of studies and meta-analyses on dietary fibre supplementation in enteral nutrition and intestinal homeostasis.
MechanismStudy (First Author, Year)Study Population and SizeDietary Fibre TypeResults
MicrobiotaFreedberg, 2020 [12]ICU patients with broad-spectrum
antibiotics
(n = 20, I 10/C 10)
Soy- and oat-derived fibre↑ SCFA stool
↑ SCFA producer
Majid, 2013 [13]ICU patients
(n = 22, I 12/C 10)
FOS/Inulin= Faecal Bifidobacterium
Simakachorn, 2011 [14]ICU paediatric patients
(n = 80, I 41/C 39)
FOS, Inulin, Acacia gum↑ Faecal Bifidobacterium
↑ Total Lactobacillus
= Enterobacteriaceae
Intestinal barrier integrityLiu, 2022 [15]MA, critically ill patients
(ON, EN and PN, n = 115, I 63/C 52)
Various↓ Intestinal permeability
Spindler-Vesel, 2007 [16]Trauma patients
(n = 81, F 29/C 26/S 26)
Guar gum, β-glucan, inulin, pectin, resistant starch (+probiotics)↓ Intestinal permeability
Lopez, 2014 [17]Patients with multi-organ failure
(n = 89, I 46/C 43)
Dietary fibre
(+probiotics)
↓ Mucosa colonisation by Candida
↓ Lactate levels
Wang, 2020 [18]Mice with sepsis 1
(n = 48, I 24/C 24)
GOS, stachyose, MOS↑ Intestinal barrier function
Aydogan, 2007 [19]Operated rats
(n = 24, I 12/C 12)
Cellulose= BTR
= Ileal changes
Sanchez, 1994 [20]Rats with enterocolitis
(n = 72, I 36/C 36)
Pectin↑ Mucous parameters
Hou, 2010 [21]Rats with trauma
(n = 12, I 6/C 6)
Dietary fibre↓ Endotoxins in portal vein
Local cellular immune responseDe Luis, 2002 [22]Oral and laryngeal cancer patients
(n = 47, I 23/C 24)
Dietary fibre (+arginine)= Lymphocytes
Lee, 2016 [23]ICU patients
(n = 15, I 8/C 7)
β-glucan↑ NK cell activity
Mao, 2022 [24]Stroke ICU patients
(n = 60, I 30/C 30)
Pectin (+probiotics)= Lymphocytes
Abe, 2018 [25]Patients with oesophageal cancer surgery
(n = 326 (I 137/C 189)
Guar gum, FOS
(+glutamine)
↓ L/N ratio
Hou, 2010 [21]Rats with trauma
(n = 12, I 6/C 6)
Dietary fibre= TNF-α
= IL-6
↑ sIgA
Systemic
inflammation
Liu, 2022 [15]MA, critically ill patients
(ON, EN and PN; n = 104, I 53/C 51)
Various↓ CRP
De Luis, 2002 [22]Oral and laryngeal cancer patients
(n = 47, I 23/C 24)
Dietary fibre
(+arginine)
= Albumin
= Prealbumin
= Transferrin
Mao, 2022 [24]Stroke ICU patients
(n = 60, I 30/C 30)
Pectin↑ Prealbumin
Abe, 2018 [25]Patients with oesophageal cancer surgery
(n = 326, I 137/C 189)
Guar gum, FOS
(+glutamine)
↓ SIRS
↓ CRP
Olah, 2007 [26]Patients with severe acute pancreatitisDietary fibre (+probiotics)↓ SIRS
1 Oral nutrition of mice, ↑ significant increase, = no significant effect, ↓ significant decrease, BTR = bacterial translocation rates, C = control group, CRP = C-reactive protein, FOS = fructooligosaccharide, GOS = galactooligosaccharide, I = intervention/dietary fibre group, ICU = intensive care unit, IL = interleukin, L/N ratio = lymphocyte/neutrophil ratio, MA = meta-analysis, MOS = mannose oligosaccharide, NK = natural killer, sIgA = secretory immunoglobulin A, SIRS = systemic inflammatory response syndrome, TNF-a = tumor necrosis factor alpha.

3.1. Shaping the Composition and Function of the Microbiota through Dietary Fibres (1)

In response to sepsis and critical illness, a sudden and drastic collapse of the commensal microbiota and its replacement by pathobionts has been demonstrated, in line with a decline in microbial diversity [27,28,29] (Figure 1).

3.1.1. Immunological Role of the Microbiota in Health and Disease

The gut microbiota plays an important role in shaping and modulating the host immune system, and commensal bacteria are crucial for protection against infections. The microbial composition and complexity within the gut ensure the ability of the microbiota to generate a consistent immunomodulatory effect within the host and maintain the intestinal gut barrier integrity [30,31]. An alteration of predominant commensal gut microbes and their reduced stability is called dysbiosis and can be associated with the development of several diseases [31,32,33]. It has been shown that microbial composition and complexity vary between healthy and diseased individuals [34,35,36,37,38]. Not only endogenous but also exogenous factors such as host physiology and immunity, diet, and drugs such as antibiotics influence microbial composition [39,40]. Several studies showed that the use of antibiotics early in life leads to alterations of the intestinal microbiota in a murine model and that these alterations recovered after cessation of antibiotics, while the altered metabolic phenotype caused by the application of antibiotics persisted during adulthood [41,42,43,44]. Others showed how the use of antibiotics strongly affected the gut microbial composition and promoted a pro-inflammatory phenotype in the long term by changing the frequency and function of invariant natural killer T cells even after reconstitution of the gut microbiome [42,45,46].

3.1.2. Beneficial Effect of Dietary Fibre Fermentation through the Gut Microbiota

Diet is an important exogenous factor reshaping the intestinal microbiota [47]. A westernised diet has been characterised as consisting of a high fat content but a low DF content [10,31]. There are several studies implementing the effect of diet, especially a low-DF westernised diet, on gut microbial composition contributing to an altered host immune response in murine models and in humans [10,31,48,49,50]. During DF fermentation by commensal gut bacteria, SCFAs such as acetate, propionate, and butyrate are produced [51,52]. Dietary changes such as high-DF diets have been associated with an increase in SCFA production within the gut [53]. SCFAs have been shown to have a beneficial effect on the intestinal gut barrier integrity and regulatory T cell (Treg) differentiation which will be discussed later in more detail.

3.1.3. Impact of Dietary Fibre Supplementation on the Intestinal Microbiota during Enteral Nutrition in Sepsis

A preclinical study showed that the diversity of the faecal microbiota in mice with sepsis increased after oral DF supplementation compared to the standard diet [18]. Two clinical studies in enterally fed ICU patients showed an increase in SCFA-producing bacteria, Bifidobacterium, and Lactobacillus, as well as increased levels of SCFAs in the stool, with no effect on Enterobacteriaceae populations upon DF supplementation [12,14]. Another study with a similar DF found no significant effect on faecal Bifidobacterium abundance [13] (Table 1).

3.2. Supporting the Intestinal Barrier Integrity through Dietary Fibres (2)

Maintaining the intestinal gut barrier integrity is fundamental to prevent the translocation of microorganisms and endotoxins to the systemic circulation, which could eventually lead to sepsis [54] (Figure 1).

3.2.1. Structure and Function of Intestinal Barrier

The intestinal barrier consists of a single layer of cells, forming an interface between luminal intestinal microorganisms and the host immune system, and represents the first line of defence against intestinal microorganisms and other environmental factors [10,45,55]. The intestinal epithelium earns its barrier function through tight junctions (TJs) which are intercellular adhesion molecules controlling paracellular permeability [56]. A second key player contributing to the intestinal barrier function is the mucus layer produced by so-called goblet cells (GCs) which are located within the intestinal epithelium [55]. Several studies showed that mice lacking the Mucin 2 gene (MUC2), a glycoprotein produced by GCs that strengthens the gut barrier integrity, were more susceptible to colitis since pathobionts could easily translocate through the intestinal barrier [57,58,59].

3.2.2. Beneficial Effect of DF Fermentation on the Intestinal Barrier Integrity

It has been previously shown that SCFAs are able to suppress intestinal inflammation in a murine model of colon cancer [39,49] and promote mucus secretion [48,60]. Acetate, an SCFA deriving from Bifidobacteria among others, has been shown to influence GC differentiation in gnotobiotic rats [61]. Arike and colleagues were able to show that germ-free mice lacking SCFAs show shorter MUC2 O-glycans, which correlated with the decrease in the respective glycosyltransferase enzymes responsible for glycan elongation [62]. One example of the beneficial effects of SCFAs has been shown by microbial-derived butyrate, which enhances intestinal barrier integrity in mice [39,63]. Furthermore, SCFAs promoted the secretion of immunoglobulin A (IgA) by B cells [10,48,64]. IgA plays a key role in maintaining gut homeostasis by regulating the host immune system towards tolerance of the commensal gut microbiota rather than responsiveness [10,45,48]. Liu and colleagues also showed that oral administration of SCFAs promoted gut homeostasis in shifting the T cell response towards an anti-inflammatory phenotype by promoting the secretion of interleukin 10 (IL-10) by Treg cells [33]. In addition, several studies showed that environmental factors can affect intestinal homeostasis [10,31,65,66,67]. For example, the use of antibiotics resulted in impaired intestinal barrier integrity [46,68], while a high-fat or westernised diet has been associated with gut microbial dysbiosis. This results in a decreased abundance of SCFA-producing bacteria [37,69] or bacteria promoting mucus secretion [48,61] and an increased intestinal barrier permeability by decreasing TJ expression [70]. Therefore, a leakage of toxic bacterial components such as LPS into the bloodstream could be observed, causing endotoxinaemia [10,31,43,71,72,73]. Simpson and colleagues demonstrated that a westernised diet low in DFs increases the abundance of mucosa-penetrating Proteobacteria [7,10]. Further, it has been shown that a westernised diet alters the gut microbial diversity, resulting in a loss of Bacteriodetes [7,10,74]. Several studies identified beneficial effects of Akkermansia muciniphila, Bifidobacterium spp., Bacteroidetes spp., Lactobacillus spp., and Clostridiales spp. as gut barrier-promoting bacteria, while Oscillibacter spp. and Desulfovibrio deteriorated the integrity of the intestinal barrier [31,71,75,76,77,78,79,80]. In addition, Ding and colleagues found that transplanting gut microbiota from mice fed a high-fat diet to germ-free (GF) mice activated the pro-inflammatory pathway (NFkb1), indicating that diet-induced dysbiosis is sufficient to cause intestinal inflammation [31,81]. Dietary interventions such as supplementation of DFs have been shown to promote gut barrier integrity in mice through an increased SCFA production [52,53].
In addition, DFs promote gut homeostasis and intestinal barrier integrity independent of the microbiota by interacting with intestinal epithelial cells and immune cells. Depending on the type of DF, epithelial TJ protein, GC function, or epithelial cell and glycocalyx maturation can be modulated. While there is high evidence for HMOs and galactooligosaccharides, no such effect has been observed for arabinoxylan and β-glucan [82].

3.2.3. Impact of Dietary Fibres on the Intestinal Barrier Integrity during Enteral Nutrition in Sepsis

Intestinal permeability was significantly reduced in a preclinical study in mice with sepsis by oral feeding containing DFs [18]. Two studies in enterally fed rats with trauma and enterocolitis indicated similar improvement in intestinal barrier function [20,21]. However, the supplementation of cellulose to EN could not improve barrier function in operated rats [19]. In two clinical studies by Spindler-Vesel (2007) and Lopez (2014), intestinal permeability decreased after symbiotic treatment and prebiotic treatment, respectively, compared to control diet [16,17]. This reduction in intestinal permeability was confirmed in a meta-analysis by Liu et al. in critically ill patients without restrictions on the route of nutrition [15] (Table 1).

3.3. Regulation of the Local Cellular Immune Response by Dietary Fibres (3)

A dysregulated host immune response to infection is a key factor of sepsis according to the third international consensus definitions [1]. DFs are metabolised by beneficial gut bacteria, such as Bifidobacteria and certain Bacteroides species, resulting in a release of microbial metabolites. These microbial metabolites such as butyrate or acetate can serve as an energy source for colonic epithelial cells and modulate metabolism and immune function [51,83] (Figure 1).

3.3.1. Mechanism of Local Cellular Immune Response Related to Dietary Fibres

Under normal conditions and if gut intestinal barrier integrity persists, the immune system is able to balance between elimination of pathobionts and tolerance towards beneficial commensal gut bacteria [47,55,59]. Even commensal gut bacteria need continuous monitoring by the intestinal immune system to prevent their outgrowth and mischief [84,85,86,87]. Antibody-presenting cells (APCs), such as dendritic cells (DCs), play an important role in monitoring the gut microbiota. According to which pathogen-associated molecular patterns (PAMPs) they sense, DCs can distinguish commensal microbes from pathobionts and activate the corresponding downstream immune responses [84]. During homeostasis, DC maturation occurs upon SCFA binding among others, resulting in antigen presentation to naïve T cells and differentiation into Treg cells [33]. Several studies emphasise the influence of butyrate on gene expression by the inhibition of histone deacetylases which influences the differentiation of naïve T cells into Treg cells in mice [55,63,65,88,89,90]. Treg cells are important regulatory cells for dampening a pro-inflammatory immune response and therefore avoiding excessive inflammation by secreting anti-inflammatory IL-10 responsible for an immunosuppressive effect [33,91].
Older literature already showed evidence on IgA production, which is highly dependent on the presence of intestinal gut microbes since mucosal IgA levels in GF mice were very low but could be restored after colonisation of these GF animals [92,93,94]. This indicates that IgA may play an important role in maintaining the symbiotic relationship between the host and the gut microbiota [48,95,96,97]. Upon activation by PAMPs or SCFAs via DCs, B cells enter the systemic circulation through the lymph and the blood to then home back to the intestinal mucosal tissue and seed it with IgA-secreting plasma cells [95]. Once the intestinal lumen is reached, IgA prevents bacterial invasion by binding to specific microbes (commensal bacteria to avoid outgrowth or pathobionts to avoid infections) and therefore blocking the attachment to the host [10,94,95,98]. This binding of IgA to microbes marks bacteria for phagocytosis and antigen presentation to DCs [10]. Several studies show indirect evidence of SCFAs promoting the secretion of IgA by B cells [48,64].
DCs can undergo maturation and activation also through other factors, such as diet or PAMPs derived from pathobionts. After DC activation and maturation, antigens are presented to naïve T cells, resulting in their proliferation and differentiation into pro-inflammatory effector T cells, such as Th1, Th17, and invariant natural killer T cells, rather than Treg cells [47].
The communication between the intestinal microbiota and the immune system plays a crucial role in gut homeostasis, and even the smallest changes in their communication may lead to the onset of a disease, microbial dysbiosis, alterations in bacteria-derived metabolites, and impaired intestinal barrier integrity, which then lead to activation of macrophages and DCs, giving rise to a pro-inflammatory immune milieu. Reduced microbial tolerance has been associated with many diseases [47,70,99,100,101,102,103,104]. Park and colleagues showed that if the host was in a situation of fighting against pathobionts, the bacteria-derived SCFAs shifted the immune system towards pro-inflammatory Th1/Th17 effector T cells in order to boost immunity [91,105].

3.3.2. Impact of Dietary Fibres on the Local Immune Response during Enteral Nutrition in Sepsis

Preclinical studies in rats with trauma showed an increase in secretory IgA but no effect on TNF-α and IL-6 in EN with DFs compared to standard EN [21]. There were also conflicting results in two clinical studies where natural killer cell activity was increased [23,24] but there were no significant effects on lymphocytes [22,24] (Table 1).

3.4. Mediation of Systemic Inflammation by Dietary Fibres (4)

During sepsis, DFs might modulate pro- and anti-inflammatory parameters and therefore prevent the development of systemic inflammation (Figure 1).

3.4.1. Development of Local to Systemic Inflammation

It has been shown that at sites of infections, the levels of SCFAs, such as acetate, increased and modulated the subsequent immune responses by increasing IL-10, decreasing pro-inflammatory interferon gamma (IFNγ) and tumor necrosis factor alpha (TNFα) [106], and being distributed systemically [48,107,108]. These findings indicate the importance of metabolites such as SCFAs or bile acids (BAs) on the systemic immune response. Indeed, several studies indicate that the effect of SCFAs is not only limited to the gut [105], which will be discussed later in more detail.
BAs derive from cholesterol in the liver and are secreted into the duodenum and further modified by the gut microbiota in the intestine, resulting in so-called secondary BAs, where they act as potent signalling molecules [35,101]. One main task of BAs is the emulsification of lipids, which explains why BA secretion is enhanced upon a high-fat diet in order to facilitate lipid digestion [109,110]. Another important feature of BAs is their antimicrobial activity [109,111]. Therefore, BAs are able to shape the gut microbiome to their favour, e.g., by promoting BA-metabolising bacteria and preventing the growth of bacteria sensitive to BAs [31,109,112]. BAs have several metabolic effects through their interaction with the farnesoid X receptor (FXR) and TGR5 [53]. Through activation of FXR and TGR5, BAs are able to promote glycogen synthesis and insulin sensitivity in the liver; further, they increase insulin secretion by the pancreas and promote satiety in the brain [53,113,114].

3.4.2. Role of Dietary Fibres in the Mediation of Systemic Inflammation

High-DF diets have been shown to protect against experimental intestinal inflammation and interact with the host immune responses [45,115,116].
As already discussed earlier, microbial-derived metabolites such as SCFAs influence many distant organs and their immune responses, while diet plays an important role in shaping the gut microbiome, including SCFA-producing microbes. Thornburn and colleagues showed that a high-DF diet resulted in high amounts of acetate and suppressed allergic airway disease by enhancing Treg cell immune responses [105,117]. Further, high-DF diets and subsequent production of propionate have been shown to induce haematopoiesis of DCs and reduce a Th2 immune response [105,118]. In the meantime, Braniste and colleagues showed the effect of SCFAs on the blood–brain barrier (BBB), where a colonisation with butyrate-producing Clostridium tyrobutyricum or acetate and propionate-producing Bacteroides thetaiotaomicron decreased BBB permeability [119].
Regarding the impact of DFs on BAs and the following downstream mechanisms, the literature presents evidence on the different types of DFs. Bretin and colleagues identified psyllium, a semi-soluble DF protecting against colitis via altering BA metabolism through FXR activation, which suppresses pro-inflammatory signalling pathways [120]. Others showed that different soluble DFs such as inulin and pectin were able to protect against diet-induced obesity but exacerbate experimental colitis [120,121,122]. Other papers support the fact that the effect of DFs highly depends on the type of DF by showing that inulin DF diets can trigger eosinophilia and Th2 immune cell response not only in the intestine but also in the lungs, which in excess are hallmarks for allergic asthma [123]. Further, Zhou and colleagues showed that some insoluble DFs such as bamboo shoot DF increased SCFA and BA levels influencing lipid metabolism in mice fed a high-fat diet [124]. Furthermore, a low-DF westernised diet was associated with BA profile dysregulations contributing to the development of chronic inflammatory diseases such as diabetes type 2 and colon cancer, and this might be alleviated by DF supplementation since soluble DFs increase the SCFA levels [50,125,126].

3.4.3. Impact of Dietary Fibres on the Systemic Inflammation during Enteral Nutrition in Sepsis

EN with DFs consistently reduced systemic inflammatory response syndrome (SIRS) and C-reactive protein (CRP) levels in critically ill patients compared to standard EN [17,25,26]. The meta-analysis by Liu et al. (2022) confirmed the reduction in CRP levels by DF supplementation in critically ill patients [15]. Albumin, prealbumin, and transferrin levels remained unchanged [22], and prealbumin levels even increased in stroke ICU patients after treatment with pectin-containing EN [24] (Table 1).

4. Clinical Benefits of Dietary Fibre Supplementation in Sepsis

Sepsis is not only a life-threatening condition itself but is closely interlinked with several adverse clinical outcomes, such as diarrhoea, infection, extended length of hospital and ICU stay, and mortality [2]. Based on the potential beneficial effects of DFs on gut homeostasis, we discuss in the following section how DF supplementation may improve sepsis outcomes in clinical care (Table 2).

4.1. Dietary Fibre and Sepsis

Critically ill patients often show complex and fluctuating immune and metabolic states. Malnutrition occurs frequently because of increased catabolism in the early period and augmented anabolism in the late period. Oral nutrition is rarely sufficient in these patients, and administration of nutrients via the enteral or even parenteral routes is needed to cover their nutritional requirements [4]. Adding DFs to EN not only improves the above-mentioned underlying causes of sepsis but could also prevent the development of sepsis. There is no meta-analysis addressing exactly this question; however, the meta-analysis by Chi et al. (2019) showed a decrease in the relative risk for sepsis in pre-term infants when prebiotics were supplemented orally, enterally, or parenterally [127]. Li et al. (2021) found that prebiotics together with probiotics significantly decreased the incidence of sepsis in critically ill adults, whereas the effect of prebiotics alone remained insignificant [128]. A single clinical trial could show a significant decrease in catheter-related sepsis when EN with DFs was used compared to standard EN [129] (Table 2).

4.2. Dietary Fibre on Diarrhoea

Diarrhoea is commonly defined as the passing of at least three unformed stools or more than 250 g of unformed stool per day, often in addition to other bowel symptoms [137]. It is a frequent symptom of many infections that can cause sepsis but also a symptom of sepsis itself. A dysregulated host immune response can trigger inflammation and mucosal necrosis and lead to diarrhoea [138]. Among all age groups, sexes, and locations, diarrhoeal disease is the most common underlying cause of sepsis [2]. Managing diarrhoea while avoiding constipation in ICU patients remains a major challenge where DFs could be an important step towards success. Several clinical studies have shown that EN containing DFs decreased diarrhoea and reduced the number of liquid stools compared to standard EN. Four meta-analyses summarised the current evidence, whereof three concluded that EN with DFs decreased the incidence of diarrhoea [128,130,132] and one showed no significant effect [131] (Table 2).

4.3. Dietary Fibre and Infection

Infections are the precursors of sepsis where organ dysfunction and dysregulated host responses are not yet present [139]. Controlling infections at an early stage has great potential to reduce the incidence of sepsis. In a study on 172 patients with major abdominal surgery, EN containing DFs and probiotics significantly reduced rates of bacterial infections compared to standard EN formulas [133]. Similarly, a meta-analysis showed a significant reduction in infections in pre-term infants after synbiotic treatment, whereas DFs alone did not show any significant effect [140] (Table 2).

4.4. Dietary Fibre and Length of Hospital or ICU Stay

Each hospitalisation for sepsis is estimated to cost USD 35,000, contributing greatly to the rising healthcare costs worldwide [141]. Reducing the length of hospital or ICU stay could decrease these costs immensely. Two studies, one on very low birthweight neonates and one on ICU patients, found significantly reduced length of hospital and/or ICU stay with EN containing DFs compared to standard EN [134,135]. A study on ICU patients with EN and synbiotics could not detect a significant reduction in the length of ICU stay [136] (Table 2).

4.5. Dietary Fibre and Mortality

In 2017, 11 million sepsis-related deaths were recorded worldwide, making sepsis a major cause of global death [2]. The ICU mortality and in-hospital mortality decreased significantly for ICU patients that received EN with DFs together with arginine compared to standard EN [129]. Similarly, the mortality was decreased for very low birthweight neonates and pre-term infants who received DFs [127,134] (Table 2).

5. Implication for Clinical Practice and Future Research

5.1. Clinical Potential

EN compared to parenteral nutrition already prevents and improves sepsis-related outcomes [142,143]. Improvement in EN composition could enhance intestinal barrier integrity, reduce bacterial translocation, and ultimately prevent the occurrence of sepsis. In recent years, clinical evidence that DFs can improve and prevent underlying causes of sepsis has increased drastically. The composition and diversity of the microbiome can be enhanced, resulting in increased levels of SCFAs. Tanes and colleagues showed that a lack of DFs in exclusive EN formulas leads to the slower recovery of the gut microbiome after environmental stress [144]. Although these findings were obtained in healthy individuals, they may be transferable to critically ill patients who are also exposed to high levels of stress and antibiotics. EN with DFs could help to shift the gut microbial composition from dysbiosis back towards eubiosis and improve recovery from sepsis. In addition, most studies have shown that EN with DFs strengthens the intestinal barrier function and decreases permeability. This implies a high potential to inhibit microbial translocation.
Dysregulation of host responses could be reduced, especially in combination with other immunomodulatory nutrients. Interestingly, the meta-analysis by Li and colleagues indicates that sole prebiotic or probiotic supplementation during EN failed to significantly decrease the odds ratio for sepsis. However, when administered together as a synbiotic, the odds ratio was significantly lower compared to patients on standard EN [128]. Many studies have investigated the effect of DFs in EN in addition to other immunonutrients, such as arginine and probiotics, which may overestimate the effect of DFs.
With regard to the consequences of sepsis, DFs have a high potential to reduce diarrhoea in enterally fed critically ill patients. Since diarrhoea is the most common cause of sepsis, this potential should not be overlooked. Clinical evidence suggested that natural diets reduce the incidence of diarrhoea compared with commercial enteral diets and should therefore be considered in addition [145,146]. Data on efficacy in terms of infections, mortality, and shortening of length of hospital or ICU stay are limited, especially regarding the effect of DFs alone during EN. The present results are promising and show a positive trend, but further research is urgently needed. The same applies to the prevention of sepsis.

5.2. Concerns Regarding Dietary Fibre Supplementation

The European Society for Clinical Nutrition and Metabolism (ESPEN) guidelines lack recommendations on the use of DFs for critically ill patients [4]. The American Society for Parenteral and Enteral Nutrition (ASPEN) and the Society of Critical Care Medicine (SCCM) recommend fermentable soluble DF supplements to be considered in stable medical and surgical ICU patients but advise against the routine use of mixed or insoluble DFs [147]. There are particular concerns in patients at high risk of bowel ischaemia or severe dysmotility due to reported bowel obstruction in surgical and post-traumatic patients receiving EN containing insoluble DFs [148,149]. These haemodynamically unstable patients may be susceptible to feeding intolerances. Increased fermentation and bowel distention can occur, which results in an increased risk of adverse events [147]. DFs as a main source of fermentation could enhance negative effects. However, clinical evidence undermining these concerns is scarce.
DFs may be of limited benefit in patients receiving oral antibiotics, and close monitoring of potential side effects, such as abdominal pain or flatulence, is indicated. The faecal microbiota of healthy volunteers exhibited decreases in diversity, richness, and evenness after a 5-day administration of oral ciprofloxacin [150]. Another study with a 7-day clindamycin administration showed that the Bacteroides population, known for its high DF-degrading ability, does not return to its original composition for up to 2 years after treatment. In phases of a reduced abundance of DF-degrading microbes, the DFs may not be able to unfold their full beneficial effects.
Another important point to consider is the interaction between DFs and drugs, which is discussed elsewhere [151,152].

6. Conclusions and Future Directions

There is growing evidence that the benefits of EN containing DFs outweigh the risk in most patients, as they target different underlying mechanisms such as mucosal barrier function, cellular defence, and inflammation.
However, the clinical evidence on the effect of EN containing DFs on sepsis and the associated outcomes is scarce and insufficient. Further research with large and high-quality clinical trials on the effect of DFs alone without other immunonutrients is needed to answer these questions. The type of DF (soluble or insoluble) may also play a role in the beneficial effect of DFs and should be considered. In addition, the role of non-fermentable, insoluble DFs (e.g., cellulose, lignin) should be investigated in more detail, along with the potential additive effect of natural enteral-feeding rich in DFs.
Beyond that, there is a high potential for the supplementation of bacterial metabolites such as butyrate or acetate to bridge the phases of a deprived microbiota. Clinical results on these postbiotics remain scarce.

Author Contributions

Conceptualisation, V.V.H., M.S. and M.L.B.; methodology, V.V.H. and M.L.B.; investigation, V.V.H. and M.S.; writing—original draft preparation, V.V.H. and M.S.; writing—review and editing, M.L.B., K.A.S., Z.S. and S.M.; visualisation, V.V.H. and M.S.; supervision, M.L.B., S.M. and Z.S.; project administration, V.V.H.; funding acquisition, M.L.B., S.M. and Z.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a third-party grant of the Division of Clinical Pharmacy and Epidemiology, University of Basel, grant number FO119900, and the Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, research fund number WFE-002. MLB is funded by SNSF Grant PCEFP3_194618/1 and the Diabetes Center Berne. The APC was funded by the Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

  • Search EN & Fibre & Critically Ill
  • Search Term MEDLINE
  • Ovid MEDLINE(R) ALL <1946 to 8 September 2022>
1.
Enteral Nutrition/ → 21,494
2.
(((Enteral or tube* or force or gastric) adj3 (nutrition or feed*))).ti,ab,kf. → 27,042
3.
Exp Dietary fiber/or dietary carbohydrates/or prebiotics/ → → 47,334
4.
((diet* adj3 fib*) or (wheat adj3 bran*) or roughage* or prebiotic* or (dietary adj3 carbohydrate*) or (resistant adj3 starch) or “alimentary fib*” or “stimulance multi fib*” or alant* or “dahlin” or inulin* or “synanthrin” or xylan* or arabinoxylan or xyloarabinan or hemixylan or (beta adj3 glucan*) or “beta dextroglucan” or macrogard or Pectin* or Methoxy?pectin or guar or Glucotard or slocose or supercol or “cyamopsis gum” or decorpa or fibraguar or galactasol or “gum cyamopsis” or “hepart hp 7000” or lejguar or prefill or galacto?oligosaccharide* or “galactose oligomer” or oligogalactose or GOS or fructo?oligosaccharide* or Idolax or “Raftilose P95” or neosugar or oligofructose or Metamucil or Plantaglucide or Ispaghul* or (Plantago adj Seed*) or Iso?gel or Reguval or agiocur or arcolax or betajel or fybogel or konsyl or metamucil or mucilax or mucilose or mucofalk or “plantaginis semen” or plantaglucid* or “plantago ovata extract” or “plantago ovata seed” or psyllium or regulan or transilane or “vi siblin” or volcolon or Flax* or Linum* or Lin?seed*).ti,ab,kf. → 93,681
5.
Sepsis/or Critical Illness/ → 102,223
6.
(Immuno* or sepsis or septic* or (bloodstream adj infection*) or pyemia* or pyaemia* or (blood adj3 poisoning*) or bacteremi* or fungemi* or parasite* or viremi* or (critical* adj ill*) or (critical* adj car*) or ICU or (intensiv* adj care*)).ti,ab,kf. → 2,636,058
7.
1 or 2 → 35,898
8.
3 or 4 → 123,642
9.
5 or 6 → 2,654,657
10.
7 and 8 and 9 → 205
  • Search Term Google Scholar
  • “Enteral nutrition” “Dietary fiber”|Prebiotics “Critical Illness”|Sepsis|Immunonutrition

References

  1. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef] [PubMed]
  2. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [PubMed]
  3. WHO. Sepsis. Available online: https://www.who.int/news-room/fact-sheets/detail/sepsis (accessed on 14 December 2022).
  4. Singer, P.; Blaser, A.R.; Berger, M.M.; Alhazzani, W.; Calder, P.C.; Casaer, M.P.; Hiesmayr, M.; Mayer, K.; Montejo, J.C.; Pichard, C.; et al. ESPEN guideline on clinical nutrition in the intensive care unit. Clin. Nutr. 2019, 38, 48–79. [Google Scholar] [CrossRef] [PubMed]
  5. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef]
  6. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef]
  7. Simpson, H.L.; Campbell, B.J. Review article: Dietary fibre-microbiota interactions. Aliment. Pharm. 2015, 42, 158–179. [Google Scholar] [CrossRef]
  8. Chen, H.; Mao, X.; He, J.; Yu, B.; Huang, Z.; Yu, J.; Zheng, P.; Chen, D. Dietary fibre affects intestinal mucosal barrier function and regulates intestinal bacteria in weaning piglets. Br. J. Nutr. 2013, 110, 1837–1848. [Google Scholar] [CrossRef]
  9. Bach Knudsen, K.E.; Lærke, H.N.; Hedemann, M.S.; Nielsen, T.S.; Ingerslev, A.K.; Gundelund Nielsen, D.S.; Theil, P.K.; Purup, S.; Hald, S.; Schioldan, A.G.; et al. Impact of Diet-Modulated Butyrate Production on Intestinal Barrier Function and Inflammation. Nutrients 2018, 10, 1499. [Google Scholar] [CrossRef]
  10. Scheithauer, T.P.M.; Rampanelli, E.; Nieuwdorp, M.; Vallance, B.A.; Verchere, C.B.; van Raalte, D.H.; Herrema, H. Gut Microbiota as a Trigger for Metabolic Inflammation in Obesity and Type 2 Diabetes. Front. Immunol. 2020, 11, 571731. [Google Scholar] [CrossRef]
  11. Schley, P.D.; Field, C.J. The immune-enhancing effects of dietary fibres and prebiotics. Br. J. Nutr. 2002, 87 (Suppl. S2), S221–S230. [Google Scholar] [CrossRef]
  12. Freedberg, D.E.; Messina, M.; Lynch, E.; Tess, M.; Miracle, E.; Chong, D.H.; Wahab, R.; Abrams, J.A.; Wang, H.H.; Munck, C. Impact of Fiber-Based Enteral Nutrition on the Gut Microbiome of ICU Patients Receiving Broad-Spectrum Antibiotics: A Randomized Pilot Trial. Crit. Care Explor. 2020, 2, e0135. [Google Scholar] [CrossRef]
  13. Majid, H.A.; Cole, J.; Emery, P.W.; Whelan, K. Additional oligofructose/inulin does not increase faecal bifidobacteria in critically ill patients receiving enteral nutrition: A randomised controlled trial. Clin. Nutr. 2014, 33, 966–972. [Google Scholar] [CrossRef]
  14. Simakachorn, N.; Bibiloni, R.; Yimyaem, P.; Tongpenyai, Y.; Varavithaya, W.; Grathwohl, D.; Reuteler, G.; Maire, J.C.; Blum, S.; Steenhout, P.; et al. Tolerance, safety, and effect on the faecal microbiota of an enteral formula supplemented with pre- and probiotics in critically ill children. J. Pediatr. Gastroenterol. Nutr. 2011, 53, 174–181. [Google Scholar] [CrossRef]
  15. Liu, T.; Wang, C.; Wang, Y.Y.; Wang, L.L.; Ojo, O.; Feng, Q.Q.; Jiang, X.S.; Wang, X.H. Effect of dietary fiber on gut barrier function, gut microbiota, short-chain fatty acids, inflammation, and clinical outcomes in critically ill patients: A systematic review and meta-analysis. J. Parenter. Enter. Nutr. 2022, 46, 997–1010. [Google Scholar] [CrossRef]
  16. Spindler-Vesel, A.; Bengmark, S.; Vovk, I.; Cerovic, O.; Kompan, L. Synbiotics, prebiotics, glutamine, or peptide in early enteral nutrition: A randomized study in trauma patients. J. Parenter. Enter. Nutr. 2007, 31, 119–126. [Google Scholar] [CrossRef]
  17. Lopez de Toro Martin-Consuegra, I.; Sanchez-Casado, M.; Perez-Pedrero Sanchez-Belmonte, M.J.; Lopez-Reina Torrijos, P.; Sanchez-Rodriguez, P.; Raigal-Cano, A.; Heredero-Galvez, E.; Zubigaray, S.B.; Arrese-Cosculluela, M.A. The influence of symbiotics in multi-organ failure: Randomised trial. Med. Clin. 2014, 143, 143–149. [Google Scholar] [CrossRef]
  18. Wang, H.; He, C.; Liu, Y.; Zhao, H.; Long, L.; Gai, X.; Zhao, H. Soluble dietary fiber protects intestinal mucosal barrier by improving intestinal flora in a murine model of sepsis. Biomed. Pharm. 2020, 129, 110343. [Google Scholar] [CrossRef]
  19. Aydogan, A.; Kismet, K.; Kilicoglu, B.; Erel, S.; Ozcan, A.H.; Gollu, A.; Yildiz, E.; Akkus, M.A. Effects of various enteral nutrition solutions on bacterial translocation and intestinal morphology during the postoperative period. Adv. Ther. 2007, 24, 41–49. [Google Scholar] [CrossRef]
  20. Gil Sanchez, M.D.; Rodriguez, J.J.; Marquez Baez, M.A.; Lorenzo Rocha, N.; Ortega Lles, B.; Alarco Hernandez, A.; Gonzalez Hermoso, F. Bacterial translocation: The effect of supplements with dietary fiber in enteral diets in an experimental model of methotrexate-induced enterocolitis. Nutr. Hosp. 1994, 9, 375–384. [Google Scholar]
  21. Hou, H.; Ping, X.; Zhu, Y.; Zhao, Z.; Li, Y.; Li, J. Dietary fiber alleviates intestinal barrier dysfunction in post-trauma rats. Clin. Investig. Med. 2010, 33, E117. [Google Scholar] [CrossRef]
  22. de Luis, D.A.; Aller, R.; Izaola, O.; Cuellar, L.; Terroba, M.C. Postsurgery enteral nutrition in head and neck cancer patients. Eur. J. Clin. Nutr. 2002, 56, 1126–1129. [Google Scholar] [CrossRef] [PubMed]
  23. Lee, J.G.; Kim, Y.S.; Lee, Y.J.; Ahn, H.Y.; Kim, M.; Kim, M.; Cho, M.J.; Cho, Y.; Lee, J.H. Effect of Immune-Enhancing Enteral Nutrition Enriched with or without Beta-Glucan on Immunomodulation in Critically Ill Patients. Nutrients 2016, 8, 336. [Google Scholar] [CrossRef] [PubMed]
  24. Mao, H.Z.; Xiong, F.T.; Hu, M.; Fu, Z. Effects of enteral nutrition semi-curing feeding on nutritional diarrhoea improvement in the patients with severe stroke. Bratisl. Lek. Listy 2022, 123, 214–217. [Google Scholar] [CrossRef] [PubMed]
  25. Abe, T.; Hosoi, T.; Kawai, R.; Uemura, N.; Higaki, E.; An, B.; Kawakami, J.; Saito, T.; Shimizu, Y. Perioperative enteral supplementation with glutamine, fiber, and oligosaccharide reduces early postoperative surgical stress following esophagectomy for esophageal cancer. Esophagus 2019, 16, 63–70. [Google Scholar] [CrossRef]
  26. Olah, A.; Belagyi, T.; Poto, L.; Romics, L., Jr.; Bengmark, S. Synbiotic control of inflammation and infection in severe acute pancreatitis: A prospective, randomized, double blind study. Hepato-Gastroenterol. 2007, 54, 590–594. [Google Scholar]
  27. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef]
  28. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
  29. Zaborin, A.; Smith, D.; Garfield, K.; Quensen, J.; Shakhsheer, B.; Kade, M.; Tirrell, M.; Tiedje, J.; Gilbert, J.A.; Zaborina, O.; et al. Membership and behavior of ultra-low-diversity pathogen communities present in the gut of humans during prolonged critical illness. mBio 2014, 5, e01361-14. [Google Scholar] [CrossRef]
  30. Geva-Zatorsky, N.; Sefik, E.; Kua, L.; Pasman, L.; Tan, T.G.; Ortiz-Lopez, A.; Yanortsang, T.B.; Yang, L.; Jupp, R.; Mathis, D.; et al. Mining the Human Gut Microbiota for Immunomodulatory Organisms. Cell 2017, 168, 928–943.e11. [Google Scholar] [CrossRef]
  31. Malesza, I.J.; Malesza, M.; Walkowiak, J.; Mussin, N.; Walkowiak, D.; Aringazina, R.; Bartkowiak-Wieczorek, J.; Mądry, E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021, 10, 3164. [Google Scholar] [CrossRef]
  32. Patterson, E.; Ryan, P.M.; Cryan, J.F.; Dinan, T.G.; Ross, R.P.; Fitzgerald, G.F.; Stanton, C. Gut microbiota, obesity and diabetes. Postgrad. Med. J. 2016, 92, 286–300. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, P.; Liu, M.; Xi, D.; Bai, Y.; Ma, R.; Mo, Y.; Zeng, G.; Zong, S. Short-chain fatty acids ameliorate spinal cord injury recovery by regulating the balance of regulatory T cells and effector IL-17(+) γδ T cells. J. Zhejiang Univ. Sci. B 2023, 24, 312–325. [Google Scholar] [CrossRef] [PubMed]
  34. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [Google Scholar] [CrossRef] [PubMed]
  35. Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef] [PubMed]
  36. Tilg, H.; Zmora, N.; Adolph, T.E.; Elinav, E. The intestinal microbiota fuelling metabolic inflammation. Nat. Rev. Immunol. 2020, 20, 40–54. [Google Scholar] [CrossRef] [PubMed]
  37. Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef]
  38. Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Cheng, J.; Duncan, A.E.; Kau, A.L.; Griffin, N.W.; Lombard, V.; Henrissat, B.; Bain, J.R.; et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013, 341, 1241214. [Google Scholar] [CrossRef]
  39. Hiippala, K.; Jouhten, H.; Ronkainen, A.; Hartikainen, A.; Kainulainen, V.; Jalanka, J.; Satokari, R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018, 10, 988. [Google Scholar] [CrossRef]
  40. Satokari, R. Contentious host-microbiota relationship in inflammatory bowel disease--can foes become friends again? Scand. J. Gastroenterol. 2015, 50, 34–42. [Google Scholar] [CrossRef]
  41. Cox, L.M.; Yamanishi, S.; Sohn, J.; Alekseyenko, A.V.; Leung, J.M.; Cho, I.; Kim, S.G.; Li, H.; Gao, Z.; Mahana, D.; et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014, 158, 705–721. [Google Scholar] [CrossRef]
  42. Burrello, C.; Garavaglia, F.; Cribiù, F.M.; Ercoli, G.; Bosari, S.; Caprioli, F.; Facciotti, F. Short-term Oral Antibiotics Treatment Promotes Inflammatory Activation of Colonic Invariant Natural Killer T and Conventional CD4(+) T Cells. Front. Med. 2018, 5, 21. [Google Scholar] [CrossRef]
  43. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef]
  44. Mileto, S.J.; Jardé, T.; Childress, K.O.; Jensen, J.L.; Rogers, A.P.; Kerr, G.; Hutton, M.L.; Sheedlo, M.J.; Bloch, S.C.; Shupe, J.A.; et al. Clostridioides difficile infection damages colonic stem cells via TcdB, impairing epithelial repair and recovery from disease. Proc. Natl. Acad. Sci. USA 2020, 117, 8064–8073. [Google Scholar] [CrossRef]
  45. Amoroso, C.; Perillo, F.; Strati, F.; Fantini, M.C.; Caprioli, F.; Facciotti, F. The Role of Gut Microbiota Biomodulators on Mucosal Immunity and Intestinal Inflammation. Cells 2020, 9, 1234. [Google Scholar] [CrossRef]
  46. Lucas, S.E.; Walton, S.L.; Mirabito Colafella, K.M.; Mileto, S.J.; Lyras, D.; Denton, K.M. Antihypertensives and Antibiotics: Impact on Intestinal Dysfunction and Hypertension. Hypertension 2023. [Google Scholar] [CrossRef]
  47. Sittipo, P.; Lobionda, S.; Lee, Y.K.; Maynard, C.L. Intestinal microbiota and the immune system in metabolic diseases. J. Microbiol. 2018, 56, 154–162. [Google Scholar] [CrossRef]
  48. Thorburn, A.N.; Macia, L.; Mackay, C.R. Diet, metabolites, and “western-lifestyle” inflammatory diseases. Immunity 2014, 40, 833–842. [Google Scholar] [CrossRef]
  49. Kim, M.; Friesen, L.; Park, J.; Kim, H.M.; Kim, C.H. Microbial metabolites, short-chain fatty acids, restrain tissue bacterial load, chronic inflammation, and associated cancer in the colon of mice. Eur. J. Immunol. 2018, 48, 1235–1247. [Google Scholar] [CrossRef]
  50. Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef]
  51. Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The role of short-chain fatty acids in health and disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar] [CrossRef]
  52. Macia, L.; Tan, J.; Vieira, A.T.; Leach, K.; Stanley, D.; Luong, S.; Maruya, M.; Ian McKenzie, C.; Hijikata, A.; Wong, C.; et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 2015, 6, 6734. [Google Scholar] [CrossRef]
  53. Beam, A.; Clinger, E.; Hao, L. Effect of Diet and Dietary Components on the Composition of the Gut Microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef]
  54. Rosolem, M.M.; Rabello, L.S.; Lisboa, T.; Caruso, P.; Costa, R.T.; Leal, J.V.; Salluh, J.I.; Soares, M. Critically ill patients with cancer and sepsis: Clinical course and prognostic factors. J. Crit. Care 2012, 27, 301–307. [Google Scholar] [CrossRef] [PubMed]
  55. Allaire, J.M.; Crowley, S.M.; Law, H.T.; Chang, S.Y.; Ko, H.J.; Vallance, B.A. The Intestinal Epithelium: Central Coordinator of Mucosal Immunity. Trends Immunol. 2018, 39, 677–696. [Google Scholar] [CrossRef] [PubMed]
  56. Zihni, C.; Mills, C.; Matter, K.; Balda, M.S. Tight junctions: From simple barriers to multifunctional molecular gates. Nat. Rev. Mol. Cell Biol. 2016, 17, 564–580. [Google Scholar] [CrossRef] [PubMed]
  57. Van der Sluis, M.; De Koning, B.A.; De Bruijn, A.C.; Velcich, A.; Meijerink, J.P.; Van Goudoever, J.B.; Büller, H.A.; Dekker, J.; Van Seuningen, I.; Renes, I.B.; et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 2006, 131, 117–129. [Google Scholar] [CrossRef]
  58. Bergstrom, K.S.; Kissoon-Singh, V.; Gibson, D.L.; Ma, C.; Montero, M.; Sham, H.P.; Ryz, N.; Huang, T.; Velcich, A.; Finlay, B.B.; et al. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog. 2010, 6, e1000902. [Google Scholar] [CrossRef]
  59. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  60. Willemsen, L.E.; Koetsier, M.A.; van Deventer, S.J.; van Tol, E.A. Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E(1) and E(2) production by intestinal myofibroblasts. Gut 2003, 52, 1442–1447. [Google Scholar] [CrossRef]
  61. Wrzosek, L.; Miquel, S.; Noordine, M.L.; Bouet, S.; Joncquel Chevalier-Curt, M.; Robert, V.; Philippe, C.; Bridonneau, C.; Cherbuy, C.; Robbe-Masselot, C.; et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013, 11, 61. [Google Scholar] [CrossRef]
  62. Arike, L.; Holmén-Larsson, J.; Hansson, G.C. Intestinal Muc2 mucin O-glycosylation is affected by microbiota and regulated by differential expression of glycosyltranferases. Glycobiology 2017, 27, 318–328. [Google Scholar] [CrossRef]
  63. Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef]
  64. Ishikawa, T.; Nanjo, F. Dietary cycloinulooligosaccharides enhance intestinal immunoglobulin A production in mice. Biosci. Biotechnol. Biochem. 2009, 73, 677–682. [Google Scholar] [CrossRef]
  65. Byndloss, M.X.; Olsan, E.E.; Rivera-Chávez, F.; Tiffany, C.R.; Cevallos, S.A.; Lokken, K.L.; Torres, T.P.; Byndloss, A.J.; Faber, F.; Gao, Y.; et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [Google Scholar] [CrossRef]
  66. Knoop, K.A.; Gustafsson, J.K.; McDonald, K.G.; Kulkarni, D.H.; Kassel, R.; Newberry, R.D. Antibiotics promote the sampling of luminal antigens and bacteria via colonic goblet cell associated antigen passages. Gut Microbes 2017, 8, 400–411. [Google Scholar] [CrossRef]
  67. Knoop, K.A.; McDonald, K.G.; Kulkarni, D.H.; Newberry, R.D. Antibiotics promote inflammation through the translocation of native commensal colonic bacteria. Gut 2016, 65, 1100–1109. [Google Scholar] [CrossRef]
  68. Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef]
  69. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef]
  70. Bäckhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef]
  71. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef]
  72. Caesar, R.; Reigstad, C.S.; Bäckhed, H.K.; Reinhardt, C.; Ketonen, M.; Lundén, G.; Cani, P.D.; Bäckhed, F. Gut-derived lipopolysaccharide augments adipose macrophage accumulation but is not essential for impaired glucose or insulin tolerance in mice. Gut 2012, 61, 1701–1707. [Google Scholar] [CrossRef] [PubMed]
  73. Crawford, M.; Whisner, C.; Al-Nakkash, L.; Sweazea, K.L. Six-Week High-Fat Diet Alters the Gut Microbiome and Promotes Cecal Inflammation, Endotoxin Production, and Simple Steatosis without Obesity in Male Rats. Lipids 2019, 54, 119–131. [Google Scholar] [CrossRef] [PubMed]
  74. Sonnenburg, J.L.; Bäckhed, F. Diet-microbiota interactions as moderators of human metabolism. Nature 2016, 535, 56–64. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, W.; Lv, L.; Shi, D.; Ye, J.; Fang, D.; Guo, F.; Li, Y.; He, X.; Li, L. Protective Effect of Akkermansia muciniphila against Immune-Mediated Liver Injury in a Mouse Model. Front. Microbiol. 2017, 8, 1804. [Google Scholar] [CrossRef] [PubMed]
  76. Heisel, T.; Montassier, E.; Johnson, A.; Al-Ghalith, G.; Lin, Y.W.; Wei, L.N.; Knights, D.; Gale, C.A. High-Fat Diet Changes Fungal Microbiomes and Interkingdom Relationships in the Murine Gut. mSphere 2017, 2, e00351-17. [Google Scholar] [CrossRef]
  77. Sun, J.; Qiao, Y.; Qi, C.; Jiang, W.; Xiao, H.; Shi, Y.; Le, G.W. High-fat-diet-induced obesity is associated with decreased antiinflammatory Lactobacillus reuteri sensitive to oxidative stress in mouse Peyer’s patches. Nutrition 2016, 32, 265–272. [Google Scholar] [CrossRef]
  78. Devkota, S.; Wang, Y.; Musch, M.W.; Leone, V.; Fehlner-Peach, H.; Nadimpalli, A.; Antonopoulos, D.A.; Jabri, B.; Chang, E.B. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 2012, 487, 104–108. [Google Scholar] [CrossRef]
  79. Lam, Y.Y.; Ha, C.W.; Hoffmann, J.M.; Oscarsson, J.; Dinudom, A.; Mather, T.J.; Cook, D.I.; Hunt, N.H.; Caterson, I.D.; Holmes, A.J.; et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obes. (Silver Spring) 2015, 23, 1429–1439. [Google Scholar] [CrossRef]
  80. Jiao, N.; Baker, S.S.; Nugent, C.A.; Tsompana, M.; Guan, L.; Wang, Y.; Buck, M.J.; Genco, R.J.; Baker, R.D.; Zhu, R. High-fat diet increases Clostridium clusters XIVa in obese rodents. FASEB J. 2017, 31, 965.9. [Google Scholar]
  81. Ding, S.; Chi, M.M.; Scull, B.P.; Rigby, R.; Schwerbrock, N.M.; Magness, S.; Jobin, C.; Lund, P.K. High-fat diet: Bacteria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS ONE 2010, 5, e12191. [Google Scholar] [CrossRef]
  82. Cai, Y.; Folkerts, J.; Folkerts, G.; Maurer, M.; Braber, S. Microbiota-dependent and -independent effects of dietary fibre on human health. Br. J. Pharm. 2020, 177, 1363–1381. [Google Scholar] [CrossRef]
  83. Balmer, M.L.; Ma, E.H.; Bantug, G.R.; Grählert, J.; Pfister, S.; Glatter, T.; Jauch, A.; Dimeloe, S.; Slack, E.; Dehio, P.; et al. Memory CD8(+) T Cells Require Increased Concentrations of Acetate Induced by Stress for Optimal Function. Immunity 2016, 44, 1312–1324. [Google Scholar] [CrossRef]
  84. Iwasaki, A.; Medzhitov, R. Control of adaptive immunity by the innate immune system. Nat. Immunol. 2015, 16, 343–353. [Google Scholar] [CrossRef]
  85. Brestoff, J.R.; Artis, D. Commensal bacteria at the interface of host metabolism and the immune system. Nat. Immunol. 2013, 14, 676–684. [Google Scholar] [CrossRef]
  86. Chow, J.; Lee, S.M.; Shen, Y.; Khosravi, A.; Mazmanian, S.K. Host-bacterial symbiosis in health and disease. Adv. Immunol. 2010, 107, 243–274. [Google Scholar] [CrossRef]
  87. Belkaid, Y.; Naik, S. Compartmentalized and systemic control of tissue immunity by commensals. Nat. Immunol. 2013, 14, 646–653. [Google Scholar] [CrossRef]
  88. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
  89. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
  90. Shi, L.Z.; Wang, R.; Huang, G.; Vogel, P.; Neale, G.; Green, D.R.; Chi, H. HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 2011, 208, 1367–1376. [Google Scholar] [CrossRef]
  91. Park, J.; Kim, M.; Kang, S.G.; Jannasch, A.H.; Cooper, B.; Patterson, J.; Kim, C.H. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol. 2015, 8, 80–93. [Google Scholar] [CrossRef]
  92. Logan, A.C.; Chow, K.P.; George, A.; Weinstein, P.D.; Cebra, J.J. Use of Peyer’s patch and lymph node fragment cultures to compare local immune responses to Morganella morganii. Infect. Immun. 1991, 59, 1024–1031. [Google Scholar] [CrossRef] [PubMed]
  93. Moreau, M.C.; Ducluzeau, R.; Guy-Grand, D.; Muller, M.C. Increase in the population of duodenal immunoglobulin A plasmocytes in axenic mice associated with different living or dead bacterial strains of intestinal origin. Infect. Immun. 1978, 21, 532–539. [Google Scholar] [CrossRef] [PubMed]
  94. Macpherson, A.J.; Köller, Y.; McCoy, K.D. The bilateral responsiveness between intestinal microbes and IgA. Trends Immunol. 2015, 36, 460–470. [Google Scholar] [CrossRef] [PubMed]
  95. Macpherson, A.J.; Slack, E. The functional interactions of commensal bacteria with intestinal secretory IgA. Curr. Opin. Gastroenterol. 2007, 23, 673–678. [Google Scholar] [CrossRef]
  96. Geuking, M.B.; McCoy, K.D.; Macpherson, A.J. The function of secretory IgA in the context of the intestinal continuum of adaptive immune responses in host-microbial mutualism. Semin. Immunol. 2012, 24, 36–42. [Google Scholar] [CrossRef]
  97. Peterson, D.A.; McNulty, N.P.; Guruge, J.L.; Gordon, J.I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2007, 2, 328–339. [Google Scholar] [CrossRef]
  98. Moor, K.; Diard, M.; Sellin, M.E.; Felmy, B.; Wotzka, S.Y.; Toska, A.; Bakkeren, E.; Arnoldini, M.; Bansept, F.; Co, A.D.; et al. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 2017, 544, 498–502. [Google Scholar] [CrossRef]
  99. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef]
  100. Tang, W.H.; Kitai, T.; Hazen, S.L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 1183–1196. [Google Scholar] [CrossRef]
  101. Bäckhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Natl. Acad. Sci. USA 2007, 104, 979–984. [Google Scholar] [CrossRef]
  102. Han, J.L.; Lin, H.L. Intestinal microbiota and type 2 diabetes: From mechanism insights to therapeutic perspective. World J. Gastroenterol. 2014, 20, 17737–17745. [Google Scholar] [CrossRef]
  103. Sun, S.; Lulla, A.; Sioda, M.; Winglee, K.; Wu, M.C.; Jacobs, D.R., Jr.; Shikany, J.M.; Lloyd-Jones, D.M.; Launer, L.J.; Fodor, A.A.; et al. Gut Microbiota Composition and Blood Pressure. Hypertension 2019, 73, 998–1006. [Google Scholar] [CrossRef]
  104. Gómez, H.; Kellum, J.A.; Ronco, C. Metabolic reprogramming and tolerance during sepsis-induced AKI. Nat. Rev. Nephrol. 2017, 13, 143–151. [Google Scholar] [CrossRef]
  105. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
  106. Balmer, M.L.; Ma, E.H.; Thompson, A.J.; Epple, R.; Unterstab, G.; Lötscher, J.; Dehio, P.; Schürch, C.M.; Warncke, J.D.; Perrin, G.; et al. Memory CD8(+) T Cells Balance Pro- and Anti-inflammatory Activity by Reprogramming Cellular Acetate Handling at Sites of Infection. Cell Metab. 2020, 32, 457–467.e5. [Google Scholar] [CrossRef]
  107. Herbst, T.; Sichelstiel, A.; Schär, C.; Yadava, K.; Bürki, K.; Cahenzli, J.; McCoy, K.; Marsland, B.J.; Harris, N.L. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am. J. Respir. Crit. Care Med. 2011, 184, 198–205. [Google Scholar] [CrossRef]
  108. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef]
  109. Islam, K.B.; Fukiya, S.; Hagio, M.; Fujii, N.; Ishizuka, S.; Ooka, T.; Ogura, Y.; Hayashi, T.; Yokota, A. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology 2011, 141, 1773–1781. [Google Scholar] [CrossRef]
  110. Reddy, B.S. Diet and excretion of bile acids. Cancer Res. 1981, 41, 3766–3768. [Google Scholar]
  111. Kurdi, P.; Kawanishi, K.; Mizutani, K.; Yokota, A. Mechanism of growth inhibition by free bile acids in lactobacilli and bifidobacteria. J. Bacteriol. 2006, 188, 1979–1986. [Google Scholar] [CrossRef]
  112. Wahlström, A.; Sayin, S.I.; Marschall, H.U.; Bäckhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed]
  113. Ramírez-Pérez, O.; Cruz-Ramón, V.; Chinchilla-López, P.; Méndez-Sánchez, N. The Role of the Gut Microbiota in Bile Acid Metabolism. Ann. Hepatol. 2017, 16 (Suppl. S1), S21–S26. [Google Scholar] [CrossRef] [PubMed]
  114. Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef] [PubMed]
  115. Silveira, A.L.M.; Ferreira, A.V.M.; de Oliveira, M.C.; Rachid, M.A.; da Cunha Sousa, L.F.; Dos Santos Martins, F.; Gomes-Santos, A.C.; Vieira, A.T.; Teixeira, M.M. Preventive rather than therapeutic treatment with high fiber diet attenuates clinical and inflammatory markers of acute and chronic DSS-induced colitis in mice. Eur. J. Nutr. 2017, 56, 179–191. [Google Scholar] [CrossRef]
  116. Llewellyn, S.R.; Britton, G.J.; Contijoch, E.J.; Vennaro, O.H.; Mortha, A.; Colombel, J.F.; Grinspan, A.; Clemente, J.C.; Merad, M.; Faith, J.J. Interactions Between Diet and the Intestinal Microbiota Alter Intestinal Permeability and Colitis Severity in Mice. Gastroenterology 2018, 154, 1037–1046.e2. [Google Scholar] [CrossRef]
  117. Thorburn, A.N.; McKenzie, C.I.; Shen, S.; Stanley, D.; Macia, L.; Mason, L.J.; Roberts, L.K.; Wong, C.H.; Shim, R.; Robert, R.; et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat. Commun. 2015, 6, 7320. [Google Scholar] [CrossRef]
  118. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
  119. Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Tóth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef]
  120. Bretin, A.; Zou, J.; San Yeoh, B.; Ngo, V.L.; Winer, S.; Winer, D.A.; Reddivari, L.; Pellizzon, M.; Walters, W.A.; Patterson, A.D.; et al. Psyllium Fiber Protects Against Colitis Via Activation of Bile Acid Sensor Farnesoid X Receptor. Cell. Mol. Gastroenterol. Hepatol. 2023, 15, 1421–1442. [Google Scholar] [CrossRef]
  121. Miles, J.P.; Zou, J.; Kumar, M.V.; Pellizzon, M.; Ulman, E.; Ricci, M.; Gewirtz, A.T.; Chassaing, B. Supplementation of Low- and High-fat Diets with Fermentable Fiber Exacerbates Severity of DSS-induced Acute Colitis. Inflamm. Bowel Dis. 2017, 23, 1133–1143. [Google Scholar] [CrossRef]
  122. Singh, V.; Yeoh, B.S.; Walker, R.E.; Xiao, X.; Saha, P.; Golonka, R.M.; Cai, J.; Bretin, A.C.A.; Cheng, X.; Liu, Q.; et al. Microbiota fermentation-NLRP3 axis shapes the impact of dietary fibres on intestinal inflammation. Gut 2019, 68, 1801–1812. [Google Scholar] [CrossRef]
  123. Arifuzzaman, M.; Won, T.H.; Li, T.T.; Yano, H.; Digumarthi, S.; Heras, A.F.; Zhang, W.; Parkhurst, C.N.; Kashyap, S.; Jin, W.B.; et al. Inulin fibre promotes microbiota-derived bile acids and type 2 inflammation. Nature 2022, 611, 578–584. [Google Scholar] [CrossRef]
  124. Zhou, X.; Ma, L.; Dong, L.; Li, D.; Chen, F.; Hu, X. Bamboo shoot dietary fiber alleviates gut microbiota dysbiosis and modulates liver fatty acid metabolism in mice with high-fat diet-induced obesity. Front. Nutr. 2023, 10, 1161698. [Google Scholar] [CrossRef]
  125. Makki, K.; Brolin, H.; Petersen, N.; Henricsson, M.; Christensen, D.P.; Khan, M.T.; Wahlström, A.; Bergh, P.O.; Tremaroli, V.; Schoonjans, K.; et al. 6α-hydroxylated bile acids mediate TGR5 signalling to improve glucose metabolism upon dietary fiber supplementation in mice. Gut 2023, 72, 314–324. [Google Scholar] [CrossRef]
  126. Haeusler, R.A.; Astiarraga, B.; Camastra, S.; Accili, D.; Ferrannini, E. Human insulin resistance is associated with increased plasma levels of 12α-hydroxylated bile acids. Diabetes 2013, 62, 4184–4191. [Google Scholar] [CrossRef]
  127. Chi, C.; Buys, N.; Li, C.; Sun, J.; Yin, C. 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. 2019, 73, 657–670. [Google Scholar] [CrossRef]
  128. Li, C.; Liu, L.; Gao, Z.; Zhang, J.; Chen, H.; Ma, S.; Liu, A.; Mo, M.; Wu, C.; Chen, D.; et al. Synbiotic Therapy Prevents Nosocomial Infection in Critically Ill Adult Patients: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials Based on a Bayesian Framework. Front. Med. 2021, 8, 693188. [Google Scholar] [CrossRef]
  129. Caparros, T.; Lopez, J.; Grau, T. Early enteral nutrition in critically ill patients with a high-protein diet enriched with arginine, fiber, and antioxidants compared with a standard high-protein diet. The effect on nosocomial infections and outcome. J. Parenter. Enter. Nutr. 2001, 25, 299–308, discussion 308–309. [Google Scholar] [CrossRef]
  130. Cara, K.C.; Beauchesne, A.R.; Wallace, T.C.; Chung, M. Safety of Using Enteral Nutrition Formulations Containing Dietary Fiber in Hospitalized Critical Care Patients: A Systematic Review and Meta-Analysis. J. Parenter. Enter. Nutr. 2021, 45, 882–906. [Google Scholar] [CrossRef]
  131. Kamarul Zaman, M.; Chin, K.F.; Rai, V.; Majid, H.A. Fiber and prebiotic supplementation in enteral nutrition: A systematic review and meta-analysis. World J. Gastroenterol. 2015, 21, 5372–5381. [Google Scholar] [CrossRef]
  132. del Olmo, D.; Lopez del Val, T.; Martinez de Icaya, P.; de Juana, P.; Alcazar, V.; Koning, A.; Vazquez, C. Fiber in enteral nutrition: Systematic review of the literature. Nutr. Hosp. 2004, 19, 167–174. [Google Scholar] [PubMed]
  133. Rayes, N.; Seehofer, D.; Muller, A.R.; Hansen, S.; Bengmark, S.; Neuhaus, P. Influence of probiotics and fibre on the incidence of bacterial infections following major abdominal surgery-results of a prospective trial. Z. Gastroenterol. 2002, 40, 869–876. [Google Scholar] [CrossRef] [PubMed]
  134. Dilli, D.; Aydin, B.; Fettah, N.D.; Ozyazici, E.; Beken, S.; Zenciroglu, A.; Okumus, N.; Ozyurt, B.M.; Ipek, M.S.; Akdag, A.; et al. The propre-save study: Effects of probiotics and prebiotics alone or combined on necrotizing enterocolitis in very low birth weight infants. J. Pediatr. 2015, 166, 545–551.e1. [Google Scholar] [CrossRef] [PubMed]
  135. Xi, F.; Xu, X.; Tan, S.; Gao, T.; Shi, J.; Kong, Y.; Yu, W.; Li, J.; Li, N. Efficacy and safety of pectin-supplemented enteral nutrition in intensive care: A randomized controlled trial. Asia Pac. J. Clin. Nutr. 2017, 26, 798–803. [Google Scholar] [CrossRef] [PubMed]
  136. Dehghani, M.H.; Saghafi, F.; Bordbari, Z.; Zare-Kamali, J.; Jafari-Nedooshan, J.; Sahebnasagh, A. Investigating the effect of oral synbiotic on enteral feeding tolerance in critically ill patients: A double-blinded controlled clinical trial of gut microbiota. Nutr. Clin. Pract. 2023, 38, 402–410. [Google Scholar] [CrossRef]
  137. DuPont, H.L. Acute infectious diarrhea in immunocompetent adults. N. Engl. J. Med. 2014, 370, 1532–1540. [Google Scholar] [CrossRef]
  138. Wiesen, P.; Van Gossum, A.; Preiser, J.C. Diarrhoea in the critically ill. Curr. Opin. Crit. Care 2006, 12, 149–154. [Google Scholar] [CrossRef]
  139. Huerta, L.E.; Rice, T.W. Pathologic Difference between Sepsis and Bloodstream Infections. J. Appl. Lab. Med. 2019, 3, 654–663. [Google Scholar] [CrossRef]
  140. Riskin, A.; Hochwald, O.; Bader, D.; Srugo, I.; Naftali, G.; Kugelman, A.; Cohen, E.; Mor, F.; Kaufman, B.; Shaoul, R. The effects of lactulose supplementation to enteral feedings in premature infants: A pilot study. J. Pediatr. 2010, 156, 209–214. [Google Scholar] [CrossRef]
  141. Arefian, H.; Heublein, S.; Scherag, A.; Brunkhorst, F.M.; Younis, M.Z.; Moerer, O.; Fischer, D.; Hartmann, M. Hospital-related cost of sepsis: A systematic review. J. Infect. 2017, 74, 107–117. [Google Scholar] [CrossRef]
  142. Hadfield, R.J.; Sinclair, D.G.; Houldsworth, P.E.; Evans, T.W. Effects of enteral and parenteral nutrition on gut mucosal permeability in the critically ill. Am. J. Respir. Crit. Care Med. 1995, 152, 1545–1548. [Google Scholar] [CrossRef]
  143. Martindale, R.G.; Warren, M. Should enteral nutrition be started in the first week of critical illness? Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 202–206. [Google Scholar] [CrossRef]
  144. Tanes, C.; Bittinger, K.; Gao, Y.; Friedman, E.S.; Nessel, L.; Paladhi, U.R.; Chau, L.; Panfen, E.; Fischbach, M.A.; Braun, J.; et al. Role of dietary fiber in the recovery of the human gut microbiome and its metabolome. Cell Host Microbe 2021, 29, 394–407.e5. [Google Scholar] [CrossRef]
  145. Fabiani, A.; Sanson, G.; Bottigliengo, D.; Dreas, L.; Zanetti, M.; Lorenzoni, G.; Gatti, G.; Sacilotto, M.; Pappalardo, A.; Gregori, D. Impact of a natural versus commercial enteral-feeding on the occurrence of diarrhea in critically ill cardiac surgery patients. A retrospective cohort study. Int. J. Nurs. Stud. 2020, 108, 103605. [Google Scholar] [CrossRef]
  146. Schmidt, S.B.; Kulig, W.; Winter, R.; Vasold, A.S.; Knoll, A.E.; Rollnik, J.D. The effect of a natural food based tube feeding in minimizing diarrhea in critically ill neurological patients. Clin. Nutr. 2019, 38, 332–340. [Google Scholar] [CrossRef]
  147. McClave, S.A.; Taylor, B.E.; Martindale, R.G.; Warren, M.M.; Johnson, D.R.; Braunschweig, C.; McCarthy, M.S.; Davanos, E.; Rice, T.W.; Cresci, G.A.; et al. Guidelines for the Provision and Assessment of Nutrition Support Therapy in the Adult Critically Ill Patient: Society of Critical Care Medicine (SCCM) and American Society for Parenteral and Enteral Nutrition (ASPEN). J. Parenter. Enter. Nutr. 2016, 40, 159–211. [Google Scholar] [CrossRef]
  148. McIvor, A.C.; Meguid, M.M.; Curtas, S.; Warren, J.; Kaplan, D.S. Intestinal obstruction from cecal bezoar; a complication of fiber-containing tube feedings. Nutrition 1990, 6, 115–117. [Google Scholar]
  149. Scaife, C.L.; Saffle, J.R.; Morris, S.E. Intestinal obstruction secondary to enteral feedings in burn trauma patients. J. Trauma 1999, 47, 859–863. [Google Scholar] [CrossRef]
  150. Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef]
  151. de Boer, A.; van Hunsel, F.; Bast, A. Adverse food-drug interactions. Regul. Toxicol. Pharm. 2015, 73, 859–865. [Google Scholar] [CrossRef]
  152. Schmidt, L.E.; Dalhoff, K. Food-drug interactions. Drugs 2002, 62, 1481–1502. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Putative mechanism of dietary fibre in preventing and treating sepsis. Key modes of action are indicated by the numbers 1 to 4. 1. Increase in microbiome diversity and SCFA production, 2. support of intestinal integrity via tight junction expression, 3. improvement of cellular immune responses, 4. regulation of inflammation. DC = dendritic cell, IgA = immunoglobulin A, IL = interleukin, iNKT = invariant natural killer T cell, PAMP = pathogen-associated molecular pattern, SCFA = short-chain fatty acid, Th = T helper cell, TJ = tight junction, TLR = toll-like receptor, TNF-a = tumor necrosis factor alpha, Treg = regulatory T cell. Created with BioRender.com.
Figure 1. Putative mechanism of dietary fibre in preventing and treating sepsis. Key modes of action are indicated by the numbers 1 to 4. 1. Increase in microbiome diversity and SCFA production, 2. support of intestinal integrity via tight junction expression, 3. improvement of cellular immune responses, 4. regulation of inflammation. DC = dendritic cell, IgA = immunoglobulin A, IL = interleukin, iNKT = invariant natural killer T cell, PAMP = pathogen-associated molecular pattern, SCFA = short-chain fatty acid, Th = T helper cell, TJ = tight junction, TLR = toll-like receptor, TNF-a = tumor necrosis factor alpha, Treg = regulatory T cell. Created with BioRender.com.
Nutrients 15 02489 g001
Table 2. Overview of studies and meta-analyses on dietary fibre supplementation in enteral nutrition and clinical benefits.
Table 2. Overview of studies and meta-analyses on dietary fibre supplementation in enteral nutrition and clinical benefits.
OutcomeStudyStudy TypeStudy PopulationPopulation SizeBackground TreatmentResults
for MA: RR/OR/MD (95% CI)
SepsisChi, 2019 [127]MAPre-term infants
(ON, EN, PN)
1106-↓ RR 0.64 (0.51, 0.78)
Li, 2021 [128]MACritically ill adults
(ON, EN, PN)
525Probiotics= OR 0.55 (0.15, 1.90)
↓ OR 0.34 (0.16, 0.70)
Caparros, 2001 [129]RCTCritically ill patients220-↓ Catheter-related sepsis RR 0.07 (0.01 to 0.54)
Cara, 2021 [130]MAHospitalised critical care patients186-↓ MD −2.78 (−4.10, −1.47)
Li, 2021 [128]MACritically ill adults
(ON, EN, PN)
7199Probiotics↓ OR 0.24 (0.05, 0.94)
Kamarul Zaman, 2015 [131]MACritically ill adults936-=OR 0.89 (0.41, 1.92)
Del Olmo, 2004 [132]MACritically ill and post-surgery patientsNA-↓ OR 0.66 (0.46, 0.95)
InfectionsLi, 2021 [128]MACritically ill adults
(ON, EN, PN)
4357Probiotics= RR 0.65 (0.35, 1.15)
↓ RR 0.37 (0.22, 0.61)
Rayes, 2002 [133]RCTMajor abdominal surgery patients172Probiotics↓ Bacterial infections
Length of Hospital/ICU stayDilli, 2015 [134]RCTVLBW neonates200-↓ Length of ICU stay
Xi, 2017 [135]RCTICU patients125-↓ Length of ICU/hospital stay
Dehghani, 2022 [136]RCTICU patients92Probiotics=Length of ICU stay
MortalityChi, 2019 [127]MAPre-term infants
(ON, EN, PN)
924-↓ RR 0.58 (0.36, 0.94)
Dilli, 2015 [134]RCTVLBW neonates200-↓ Mortality
Caparros, 2001 [129]RCTCritically ill patients220Arginine↓ ICU-mortality
↓ In-hospital mortality
↑ significant increase, = no significant effect, ↓ significant decrease, EN = enteral nutrition, MA = meta-analysis, MD = mean difference, ON = oral nutrition, OR = odds ratio, PN = parenteral nutrition, RCT = randomised controlled trial, RR = relative risk. Meta-analyses were preferred over RCTs.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Huwiler, V.V.; Scalise, M.; Schönenberger, K.A.; Mühlebach, S.; Stanga, Z.; Balmer, M.L. The Role of Dietary Fibre in Enteral Nutrition in Sepsis Prevention and Therapy: A Narrative Review. Nutrients 2023, 15, 2489. https://doi.org/10.3390/nu15112489

AMA Style

Huwiler VV, Scalise M, Schönenberger KA, Mühlebach S, Stanga Z, Balmer ML. The Role of Dietary Fibre in Enteral Nutrition in Sepsis Prevention and Therapy: A Narrative Review. Nutrients. 2023; 15(11):2489. https://doi.org/10.3390/nu15112489

Chicago/Turabian Style

Huwiler, Valentina V., Melanie Scalise, Katja A. Schönenberger, Stefan Mühlebach, Zeno Stanga, and Maria L. Balmer. 2023. "The Role of Dietary Fibre in Enteral Nutrition in Sepsis Prevention and Therapy: A Narrative Review" Nutrients 15, no. 11: 2489. https://doi.org/10.3390/nu15112489

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

Article Metrics

Back to TopTop