SciELO - Scientific Electronic Library Online

 
vol.37 número1Influencia de la ingesta de alimentos o grupos de alimentos en la aparición y/o protección de los diversos tipos de cáncer: revisión sistemáticaIngestas de energía y nutrientes recomendadas en la Unión Europea: actualización para vitamina K, riboflavina, sodio y cloruro índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Nutrición Hospitalaria

versión On-line ISSN 1699-5198versión impresa ISSN 0212-1611

Nutr. Hosp. vol.37 no.1 Madrid ene./feb. 2020  Epub 08-Jun-2020

https://dx.doi.org/10.20960/nh.02775 

Revisiones

Immunonutritional contribution of gut microbiota to fatty liver disease

Contribución inmunonutricional de la microbiota intestinal a la hepatopatía grasa

Ricardo Fernández-Musoles1  , Aurora García-Tejedor1  , José Moisés Laparra2 

1Food Science and Health Group, Valencian International University. Valencia, Spain.

2Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA Alimentación). Grupo de Inmunonutrición Molecular. Madrid, Spain.

Abstract

Non-alcoholic fatty liver disease (NAFLD) is indisputably the most widespread liver disease worldwide, leading to a significant increase in patient morbidity, mortality, and health care utilization. The gut microbiota and its genome (microbiome) have emerged as a novel modulator of the immunometabolic processes that NAFLD implies, but microbiota-targeted interventions have resulted both astounding and at the same time unsuccessful. The most relevant alteration appears to be the overgrowth of Gram-negative bacteria, characterized by an increased ratio of Firmicutes to Bacteroidetes, although current evidence indicates species- and strain-specific effects influencing energy harvest, the host's innate and adaptive immune systems, and epigenetic regulation as determinants of the immunomodulatory milieu in NAFLD. The genera Lactobacillus and Bifidobacterium deserve special attention since many of their probiotic strains are marketed for human consumption, even more so when considering that, in conjunction with prebiotics, they are potential modulators of gut microbiota composition and/or metabolic activity. Here, a better understanding of the major intestinal microbial factors with a detrimental or preventive role in NAFLD, and of the dynamic interplay between gut microbiome and host factors, appears crucial in defining the exposome for the prevention and treatment of NAFLD and associated diseases such as metabolic syndrome, type-2 diabetes, and obesity.

Key words: Biomarkers; Gastrointestinal; Global health; Microbiome; Phenotype(s)

Resumen

La esteatosis hepática no alcohólica (NAFLD, por sus siglas en inglés) es indiscutiblemente la patología hepática más extendida a escala mundial y conlleva un aumento significativo de la utilización de la atención médica de los pacientes, así como de la morbilidad y la mortalidad. La microbiota intestinal y su genoma (microbioma) se han revelado como uno de los factores moduladores de los procesos inmunometabólicos subyacentes que desencadenan la NAFLD. Las intervenciones dirigidas a modificar la composición y/o la actividad de la microbiota han resultado sorprendentes y, al mismo tiempo, infructuosas. La disbiosis más relevante en la patología es un aumento de la proporción entre Firmicutes y Bacteroidetes. La evidencia actual indica que los efectos específicos de la especie y la cepa influyen en el resultado funcional de la microbiota sobre el metabolismo de los nutrientes, la rama innata y la adaptativa del sistema inmune, y la regulación epigenética del genoma humano en relación al NAFLD. Los géneros Lactobacillus y Bifidobacterium merecen especial atención ya que muchas cepas probióticas de estos géneros se comercializan para consumo humano, e incluso más si se considera que, junto con los prebióticos, son moduladores potenciales de la composición de la microbiota intestinal y/o su actividad metabólica. En este contexto, una mejor comprensión de los principales factores microbianos con papel perjudicial o preventivo en la NAFLD, y de la interacción dinámica entre el microbioma intestinal y los factores del huésped, parece crucial para definir el exposoma de la prevención y el tratamiento de la NAFLD y sus enfermedades asociadas, como el síndrome metabólico, la diabetes de tipo 2 y la obesidad.

Palabras clave: Biomarcadores; Gastrointestinal; Salud global; Microbioma; Fenotipo(s)

INTRODUCTION

Lifestyle and dietary habits are major contributors to the growing incidence of non-communicable diseases. As a consequence of a dramatic increase in risk factors such as obesity, type-2 diabetes and altered food supply and preferences, non-alcoholic fatty liver disease (NAFLD) is one of the most important causes of liver disease worldwide, affecting an estimated ≥ 24 % of most populations (1). Genetic predisposition must be also placed in the context of environmental factors. Nowadays, accumulating evidence describes a ‘lean' manifestation of NAFLD, which was initially described in Asian populations and affects at least 5 % of the population in western societies (2). This ‘lean NAFLD phenotype' represents, among others, a ‘metabolically obese, normal weight phenotype' (1,2).

A significant body of data suggests that dietary composition and its interaction with the gut microbiota - and eventually the immune system - may predispose individuals to NAFLD, rather than total calorie intake, with a continuous positive balance promoting obesity, fatty liver, and finally NASH and/or metabolic syndrome. Over the past ten years, different mouse models - germ-free, antibiotic-treated and with or without colonization by specific microbial strains (3 4-5) - have been used to prove metabolic and immune effects derived from microbes for hosts. Here, imbalances in the gut microbiota have been linked to major risk factors for NAFLD (6,7). However, direct evidence for an association between functional alterations in the gut microbiota composition, as well as the microbiota-related molecular signature, and NAFLD and NASH remains largely inferential (3,5). In this scenario, surrounding environmental conditions, including diet (within 24 h), have proven to significantly impact microbial genome (microbiome) influencing host metabolic stress and dietary energy expenditure (8). Otherwise, long-term dietary habits were more clearly associated with imbalances in microbial levels categorized into enterotypes. The metabolic activity of gut microbiota has led to consider it as a ‘supraorgan' likely comparable to the liver. Notably, gut microbiome exhibits high variability between individuals (i.e., genders) and may constitute a source of variation for the effects observed, but evidence at the population level is incompletely characterized.

The liver is a central organ for the turnover and transformation of the metabolites supplied from the intestine. In addition, the intestine constitutes an important source of immune signals that stem from the intestinal mucosa and immune system. This so-called ‘gut-liver' axis is sensitive to the gut microbiota, which modulates energy supply and homeostasis, ectopic fat deposition, intestinal and fatty tissue inflammation, and immune maturation (9 10-11). The regulation of the energy balance operates both in the central nervous system and at peripheral sites, including adipose tissue and the liver, playing a critical role in the development of NAFL/NASH and liver fibrosis (12,13). In this sense, contradictory findings have been reported suggesting an important role for the diversity of gut microbiota as well as its composition, worsening or improving, both qualitatively and quantitatively, liver lipid accumulation (14,15). Besides, only a few studies have investigated the role (bidirectional) of the gut microbiota in host lipid metabolism (16) to establish steady-state commensalism and tissue metabolic homeostasis (17).

Preclinical data also support the relevance of pre- and post-natal nutrition to the developmental programming of NAFLD (18). The importance of perinatal outcomes in microbiota modulation at critical periods for development have been highlighted for their potential role in the prevention (19) and treatment of human disease. This is increasingly thought to occur through epigenetic changes as integral underlying mechanisms. Here, it can be established that an indirect gut microbiota-mediated epigenetic regulation may modify host immunometabolic responses and, ultimately, gene expression (20,21). These are novel perspectives connecting the intrauterine milieu to risk factors for NAFLD, i.e., diabetes, obesity, and hyperlipidemia, as well as for metabolic syndrome as a whole. However, how immunonutritional food components influence these aspects in NAFLD remains poorly understood.

In relation to the above, several recent reviews (22,23) have focused on reporting experimental and clinical evidence of intestinal dysbiosis in NAFLD. From these studies, conclusions were drawn on the therapeutic potential of manipulating the gut microbiota to decrease the incidence and prevalence of fatty liver disease. However, relatively few protocols have been registered in relation to gut microbiota manipulation in NAFLD patients (24). There is still an open debate about how and what microbes or their genomes may modulate NAFLD severity and (consequently) energy expenditure or nutrient fate to prevent metabolic and immune complications. Currently obtained results are promising, but as yet they do not permit definitive clinical conclusions in relation to their clear efficacy in microbiome-targeted therapies (24,25).

This review provides a comprehensive overview about the role of the gut microbiota as an important contributor to immunometabolic factors influencing liver disease. To this end a comprehensive update of the dynamic interplay between diet, nutrients, and microbes, as well as the microbial phylogenetic changes that occur in obese versus lean individuals is reported. Particularly, this manuscript is focused on the immune function and includes the current knowledge on the functional outcome obtained from modifications in microbial ecosystems.

EXPOSOME AND LIVER FUNCTION

The mammalian gut harbours highly complex microbial communities, which develop multifaceted interactions in the health and nutrition of the host and are influenced by the exposome (i.e., environmental exposures to various exogenous - diet - and endogenous factors), and have influential effects on both metabolism and immunity. Microbial products derive from either saccharolytic or proteolytic fermentation and may affect the gut-liver axis via multiple mechanisms, hence contributing to the pathogenesis of NAFLD.

Different microbial-mediated processes have received major attention, including: a) efficiency of energy harvest, i.e., utilization of nutrients (fermentation of complex dietary carbohydrates) favouring short-chain fatty acid (SCFA) production (5); b) modulation of low-grade liver (and adipose tissue) inflammation, e.g., via activation of free fatty acid receptor 2 (FFA2, a G-protein-coupled receptor also known as GPR43) (26); c) impact on the innate immune response, such as endotoxin production from Gram-negative bacteria (27), and the adaptive immune response, by binding to leukocyte receptors (butyrate binding to GPR41 and GPR43) suppressing lymphocyte proliferation, to inhibit cytokine production by Th1-lymphocytes and to induce T-lymphocyte apoptosis (28).

SCFAs can regulate lipogenesis through their interaction with the endocannabinoid system or stimulation of orexigenic hormone release (e.g., leptin) in adipose tissue (29). Also, they can reduce or increase insulin resistance (30) via a GPR40-mediated and calcium-dependent mechanism regulating the expression of PPARδ. Otherwise, the impact or modulatory role of bacterial metabolites originating from a proteolytic metabolism on host physiology has received much less attention.

Microbes gradually shift to their proteolytic metabolism as carbohydrates are depleted in the intestinal lumen. Bacterial protein degradation products are considered deleterious for the host's physiology due to the production of phenolic compounds (i.e., indole) derived from aromatic amino acid degradation. Numerous Gram-negative, as well as Gram-positive, bacterial species able to produce indole have been identified (including Escherichia coli, Clostridiumspp., and Bacteroidesspp), including Lactobacilliand Bifidobacteriathat are considered beneficial for the host's health. Accumulating evidence suggests that microbial tryptophan catabolites resulting from proteolysis have an impact on mammalian gut immune homeostasis through binding to the aryl hydrocarbon receptor (AHR), stimulate hormone (i.e., GLP-1) secretion, and modulate gastrointestinal motility (31). Furthermore, adequate tryptophan metabolism can prevent redox imbalances due to decreased production of NAD equivalents, which are used to compensate the fermentation of carbohydrates.

Nutritional deficiency of choline has also been associated with changes in the microbiome of the human gut and the development of fatty liver, as previously found in rodent models of NASH (32). Accordingly, changes have been reported in various physiological processes such as lipid metabolism, enterohepatic circulation of bile acids, and cholesterol metabolism (33). These observations were accompanied by increased numbers of the Gram-negative bacteria Gammaproteobacteriaand Erysipelotrichi, whose presence has been associated with increases in fat content in the liver (34). The Erysipelotrichaceaefamily is relatively distant, from a phylogenetic point of view, from the Clostridialesorder, for which oxidative enzymes contributing to the production of oxysterols have been identified. This bacterial metabolic potential opens new avenues to the microbial contribution to synergistic lipotoxic effects leading to mitochondrial function impairment (35).

Collectively, existing data demonstrate that the gut microbiome can affect the liver, the adipose tissue, and metabolic diseases both in a beneficial and in detrimental way. However, owing to the complexity and multifaceted nature of the interactions between gut microbiota and its microbiome with the host's nutritional and health status, no human in vivodata on a direct link between microbial-derived metabolites, liver function, and NAFLD progression are available.

ENERGY HARVEST AND FATE OF NUTRIENTS

The commensal microbiota regulates nutrient acquisition and utilization in symbiosis with the host, providing additional enzymes (36,37). Here, it is implied a regulated expression of various genes that are involved in the utilization of dietary carbohydrates and lipids. Metagenomic studies identified gut microbial enzymes specialized in the utilization of otherwise non-digestible nutritional carbohydrates and host-derived glycoconjugates (e.g., mucins, proteoglycans). These genes participate in the deconjugation and dehydroxylation of bile acids, in cholesterol reduction (the latter being mainly converted to coprostanol by intestinal fermentative bacteria, either directly by the enzyme cholesterol reductase or via an alternative indirect pathway), in the biosynthesis of vitamins and isoprenoids, and in the metabolism of amino acids and xenobiotics (36,38).

Thus, novel human microbiome-targeted interventions have been proposed, while an effective approach has yet to be successfully developed (24). Human trials support potential positive roles for prebiotics (i.e., non-digestible food ingredients, mostly oligosaccharides that stimulate growth and activity in several specific, beneficial intestinal bacteria) enhancing NAFLD/NASH progression likely through their modulatory effects on glucose homeostasis and lipid metabolism (5). Also, the few existing studies revealed changes in the microbiome after very short-term dietary interventions (10 days) with high-fat/low fibre versus low-fat/high fibre (39). However, direct evidence of the influence of defined prebiotic structures on specific microbial genes and their physiological functions is lacking. Nowadays, prebiotic contribution to establishing a ‘score' of disease and validating its use in clinical practice remains elusive.

The potential contribution of microbial fermentation has also been a matter of controversy since it improves energy harvest from the diet, which may contribute as much as 10 % to daily calorie intake (40). Studies in rodents clearly showed the causative role of the gut microbiota in adiposity and insulin resistance, linking changes in host phenotypes to variations in gut microbial composition (14, 41, 42, 43). Thus, germ-free animals did not develop diet-induced obesity and related comorbidities, like visceral adipose tissue inflammation with macrophage infiltration and insulin resistance (42). Otherwise, these mice gained excess fat mass when conventionalized with microbiota from lean non-germ-free animals kept on a normal diet (14). The persistent lean phenotype of the germ-free animals displayed increased skeletal muscle and liver levels of phosphorylated AMP-activated protein kinase (AMPK), accompanied by its downstream targets involved in fatty-acid oxidation (acetylCoA carboxylase, carnitine-palmitoyl transferase) (14). Fat mass gain was greater when microbiota from ob/obmice (obese due to defective leptin signalling) was transferred to germ-free animals (41). The ob/obmicrobiome showed a higher proportion of environmental gene tags encoding many enzymes involved in breaking down otherwise indigestible dietary polysaccharides, including pathways for starch/sucrose, galactose and butanoate metabolism, indicating an improved energy harvest (41). In these scenarios, a comparison of microbial families associated to diabetes-related clinical traits revealed a positive correlation with Lactobacillaceae, while Bacteroidaceaewas negatively associated with weight gain (43). Dietary supplementation (12 weeks) with prebiotic galacto-oligosaccharides (GOS) was proven effective to increase fecal numbers of Bifidobacteriumspp. without improving insulin sensitivity in overweight or obese prediabetic men and women (44). Metagenomic studies in patients with T2D revealed that changes in the microbiome rather than in microbial taxa have a greater influence on the risk factors for T2D (45).

MICROBIOTA IN METABOLIC AND ENDOCRINE PROCESSES

Promising reports on the effectiveness of microbiome-based targeted interventions demonstrated changes in hepatic gene expression when animals were fed prebiotics (46,47). Gene expression implied that different genes are involved in diverse biochemical processes: lipogenesis, cholesterol production and mobilisation, or induction of PPARα-mediated fatty-acid oxidation. Notably, the composition of the gut microbiota, including the activity and growth of lactobacilli and bifidobacteria, which promote the production of SCFA (e.g., acetate, butyrate and propionate), appeared as a critical factor driving the magnitude and final biological outcomes. Apart from trophic effects on intestinal epithelia (48,49), reported findings remark the high dependence of biological effects in the host's physiology on the SCFA profile produced. While the production of luminal acetate appears to be a precursor for hepatic lipid synthesis (16), propionate oppositely impacts lipid metabolism inhibiting the effects of acetate. Generally, butyrate is regarded as a healthy metabolite as it is utilized by enterocytes, positively influencing cell growth and differentiation, and promoting anti-inflammatory effects. Collectively, it was found that increasing proportions of SCFA producers significantly alleviate hepatic steatosis, decreasing the content of triglycerides and free fatty acids in rats fed a high-fat diet (50). The latter could open new interpretations to the role of lactobacilli, major contributors to the efficiency of energy harvest and nutrient utilization, and their potential contribution to the onset and progression of NAFLD. SCFAs also affect endocrine functions due to their interaction with G-protein-coupled FA receptors (i.e., GRP43 and GRP41) (51), the stimulation of which increases the production of peptide YY and leptin (orexigenic and anorexigenic peptides, respectively) (52,53).

Much of the beneficial effects of prebiotics on plasma lipid and hepatic triglyceride concentrations are attributed to regulation of the expression of the different transcription factors involved in lipid metabolism (i.e., acetyl-CoA carboxylase, fatty-acid synthase, malic enzyme, and ATP citrate lyase). These effects occur via regulation of glucose metabolism (54) and insulin resistance (55). This could point to enhanced enteroendocrine function in L-cells (mainly located in the ileum and colon) (56) as mediators of the trophic effects associated with reduced mucosal inflammation (48,57) and induction of the antidiabetic (PPAR-mediated regulation of Akt pathway influencing GLUT4 expression in liver) and anorexigenic incretin glucagon-like peptide 1 (GLP-1).

Prebiotics have also been proven to influence physiological endocrine systems up-regulating the production of GLP-2 and expression of the endocannabinoid system (eCB) (58). Thus alleviating gut permeability, plasma lipopolysaccharides (endotoxaemia) from Gram-negative bacteria (LPS) and cytokines decreased the hepatic expression of inflammatory cytokines (e.g., TNF-α, MCP-I) and oxidative stress markers in obese (leptin-deficient) mice. It has been recently shown that a dysregulated expression of eCB 1 and 2 in the colon and liver is associated to metabolic imbalances derived from sirtuin 3 (SIRT3) deficiency (59). These observations were followed by increased endotoxaemia in correlation with gut microbiota dysbiosis. Yet the underlying mechanism linking SIRT3 with gut microbiota remains obscure, and SIRT3 deficiency may be attenuated by sodium butyrate. Prebiotic-treated obese (ob/ob) mice exhibited a normalized eCB system tone (expression of eCB-1r, plasma eCB levels or adipose tissue eCB content) in both the gut and adipose tissue, as well as endotoxaemia (29). Figure 2summarizes the interrelationship between prebiotics, gut permeability and eCB-induced adipogenesis and NAFL.

Figure 2. Interrelationship between prebiotics, gut permeability, and influence of the endocannabinoid (eCB) system in adipogenesis and NAFL development. 

Relatively rapid shifts in gut microbiota composition and their related functional outcomes in response to high-sugar and high-fat diets have been reported (8,60,61). These responses modify both gene expression and the resultant metabolite generation and flow in the host. At the same time, alterations in the composition of the gut microbiota influence strain-specific susceptibility to diet-induced metabolic disease (43). This bidirectional interaction determines the microbial capacity for processing dietary sugars and for generating hydrophobic bile acids and, thereby susceptibility to metabolic disease. The impact of metabolic disease on bacterial functional outcomes is reflected, whether worsening or improving the disease, in the role of GPR43 as sensor for excessive dietary energy (15). Apparently, this receptor loses its inhibitory influence on insulin signaling, reducing fat accumulation in adipocytes. An example is vancomycin-treated mice, which display microbial changes relatively similar to obese patients, with a significantly reduced diversity of the gut microbiota and a markedly increased abundance of Lactobacillusspp (62). However, these bacterial changes were accompanied by significant increases in body weight and colonic GPR43 expression. Microbial alterations in obese people (60,63,64) favor higher glycoside hydrolase activities and a higher activity of host transport proteins importing the breakdown products of indigestible carbohydrates.

MICROBIOME AND LIVER FUNCTION

Grain-based food, sugars and dairy products are the nutritional staples of western diet, which produce high glycemic and fat loads that have been recognized as aggravating factors of fatty liver disease.

Western diets, with the concomitant specific changes in gut microbiota to pro-inflammatory microbes (Gammaproteobacteriagenera including Klebsiellaspp., Enterobacterspp., and Escherichiaspp.), also reduce the availability and utilization of several amino acids originating from both alimentary and endogenous proteins. These metabolic conditions favour the development of fatty liver inflammation in patients on controlled diets with lowered levels of choline (32,65). Dietary choline deficiency is an example of one (of several) nutritional factors in modern diets that may favor the development of NAFL and NASH. Most studies documented that choline is an essential nutrient with a key role on fatty liver progression by decreasing methionine levels, favoring abnormal phospholipid synthesis and lipoprotein secretion, mitochondrial dysfunction, and endoplasmic reticulum stress (32,33). In addition, choline deficiency contributes to dysregulate methionine levels and tissue polyamines, a significant part of which have their origin in the intestinal microbiota, impairing glucose, lipid, and energy homeostasis.

Studies correlating the content of polyunsaturated fatty acids (PUFA) with the development and extent of NAFL have produced divergent results (66 67-68). Dietary deficiency of ω-3 PUFA caused imbalances on microbial composition in mice, with decreased cecal numbers of Lactobacillusspp. and Bacteroidetes-Prevotellaspp. (67). However, ω-3-depleted mice displayed a slightly increased proportion of Bifidobacteriumspp., which was significantly associated with increased insulin resistance and steatosis. Here, dietary supplementation with fructo-oligosaccharides (FOS), for only 24 days, increased the proportion of lactobacilli, contributing to restore an efficient fat storage without lessening liver steatosis (67). Thus, these studies demonstrate different effects of prebiotic fibres in fatty liver disease depending on dietary microbiome adaptations. Also, the results may be interpreted to suggest a direct role for microbial fermentation in steatosis, insulin resistance, and down-regulation of hepatic inflammatory regulators (i.e., peroxisome proliferator-activated receptor (PPAR)-α, and receptor gamma coactivator α). Collectively, it can be delineated a significant bulk of evidence supporting the bidirectional impact of either nutrient availability or gut microbial imbalances on each other to trigger profound effects on mammalian lipid metabolism. Table Isummarizes the effects observed in serum and liver lipids, as well as in the biomarkers associated to NAFL, in different animal models fed a high-fat diet.

Table I. Examples of the effects of prebiotic fibres on serum and liver lipids and biomarkers associated to NAFL in animal models fed a high-fat diet 

Indirectly, gut microbiota can modify the lipid profile in the enterohepatic circulation affecting overall hepatic lipid metabolism. Solely microbial transformation of cholesterol and bile acids constitute the origin of coprostanol and secondary bile acids - deoxycholic acid (3α,12α-dihydroxy-5β-cholan-24-oic acid) and lithocholic acid (3α-hydroxy-5β-cholan-24-oic acid). Under physiological conditions these bile acids are absorbed but human hepatic metabolic capacities cannot remove them from the bile acid pool causing hepatic toxicity (69). 7α-dehydroxylation is the most quantitatively important bacterial bile salt biotransformation in the human colon, and 16S rDNA phylogenetic analyses have led to classify most competent intestinal bacteria into the genus Clostridium(70). The order Clostridiales, including ClostridiumXIVa and IV subgroups, within the superfamily Lachnospiraceaeis peripherally related to the family Erysipelotrichaceaewithin phylum Firmicutes(71) mostly associated to obese individuals (72). Unfortunately, there are scarce studies addressing to what extent these differences in hydroxysteroid dehydrogenases could be responsible, at least in part, for the significant detrimental effects in NAFLD as 7β-dehydroxylation activity has also been described in Bacteroides distasonis(73).

Despite the direct effect of bile acids helping in the intestinal absorption of lipids and fat-soluble vitamins, they also play a role in regulating lipid-glucose homeostasis, inflammation, and thereby innate immune cells polarization via specific nuclear receptors (FXR) and G-protein-coupled receptors (TGR5). FXR is highly expressed in the liver, intestine, kidney and adrenal gland, and may play a role in reducing hepatic triglyceride levels via different gene pathways, including FGF21 (74), ChREBP (75), PPARα (76), and AKR1B7 (77). TGR5 is highly expressed in the gallbladder, intestine (primarily in the ileum and colon), and liver, where it has been shown to antagonize the activity of NFkB in macrophages and hepatic Kupffer cells (78). Moreover, in intestinal L-cells TGR5 induces GLP1 secretion, and in the liver TGR5 attenuated triglyceride accumulation through a yet-to-be-determined mechanism (79).

MICROBIAL IMBALANCES ASSOCIATED WITH LIVER DISEASE

A direct link between NAFL/NASH status and imbalances in the composition of the gut microbiota is now being established (3,27). The most relevant alteration is an increased ratio of Firmicutesto Bacteroidetes, associated to the overgrowth of Gram-negative bacteria and decreased proportions of Bifidobacteriumspp. in the cecal contents of obese humans (63,80). However, contrasting results have also been obtained from clinical studies where the proportion of Bacteroideswas found significantly increased in subjects with liver fibrosis (81). In a similar way to the procedures followed to uncover the role of gut microbiota in obese and T2D patients, ‘infection' models served to establish the microbial contribution to the induction of adaptive and innate immune memory (82). This bacterial contribution seems to occur at the epigenetic, metabolic, or functional level. However, despite its potentially important role, to the best of our knowledge no population-based evidence is available regarding the ability of the mucosal immune system to co-operate with systemic immunity in response to certain commensals in the setting of NAFL.

Scientific evidence available in the literature tried to establish a relationship between bacterial translocation through the intestinal barrier and NAFL aggravated symptoms and disease progression (83,84). The overgrowth of small intestinal bacteria (SIBO) is directly associated with an increase in intestinal permeability affecting nutrient absorption and utilization. Notwithstanding this, the impact and magnitude of intestinal functional impairment seem to be associated to the NAFLD phenotype.

For example, a study population (n = 35 patients) with biopsy-proven NAFLD determined the presence of a "leaky" intestine with evidence of increased SIBO (83). Furthermore, a cross-sectional study performed in outpatients with metabolic risk factors for NAFLD development (type-2 diabetes, obesity, dyslipidemia) did not find any significant association between SIBO and NAFLD or hepatic steatosis (85). However, it was found that central obesity was significantly associated with SIBO.

Metagenome-wide association studies revealed that gut microbiota in T2D patients was characterized by decreased numbers of some universal butyrate-producing bacteria (Eubacterium rectale, Faecalibacterium prausnitzii, Roseburia intestinalisand R. inulinivorans). Besides this, an increase in various opportunistic pathogens (Bacteroides caccae, Clostrium hathewayi, Clostridium ramosum, Clostridium symbiosum, Eggerthella lenta, and Escherichia coli) was quantified, as well as an enrichment of other microbial functions conferring sulphate reduction and oxidative stress resistance. Obesity was associated with reduced bacterial diversity, with lower numbers of Bacteroidetesand increased numbers of Actinobacteria(30,63,86). Some other components of the obesity-associated gut microbiome are distributed among Proteobacetria, Fusobacteriaand Verrucomicrobia(72). Otherwise, contrasting data are reported in relation to the proportion of Methanobrevibacter smithiiin obese subjects (63,72). Unexpectedly, M. smithiiand Escherichia coliwere found to negatively correlate with body mass index in overweight and obese people (87). Recent extensive studies have revealed that molecular hydrogen (H2) has a great impact on obesity development with a decreased expression of H2S-synthesizing enzymes and reduced H2S levels in adipose tissues in mice. Also, significant increases in some other bacterial groups such as Staphylococcus aureus(88,89) and Faecalibacterium prausnitzii(90) have been associated with NAFL comorbidities. However, a decreased abundance of both the Faecalibacteriumand Ruminococcusgroup was recently demonstrated in ‘lean' NASH patients (91).

The role of Lactobacillusspp. in obesity has been a subject of controversy, with part of the risk for causing public health problems in humans being attributed to the promotion of consumption of products containing these bacteria. Analyses on the proportions of Lactobacillusspp. evidenced contrasting changes in this microbial population. While significant increases (63,92), particularly in the species L. reuteriand L. ingluviei(63,93), have been found in overweight people, significant reductions in the fecal concentrations of the Lactobacillus gasserisubgroup were observed in obese infants at baseline (80). Nowadays few interesting data are available regarding the role of dietary supplementation with probiotics in the management of obesity, as most probiotics for human consumption belong to the Lactobacillusgenus. In this sense, a recent study in obese children who received daily supplementation with L. casei(Shirota strain) microbial effectiveness contributing to weight loss while improving lipid metabolism was demonstrated. Notably, these effects could be associated to a significant increase in the fecal numbers of Bifidobacteriumspp. and acetic acid concentration (80). These results suggest that microbial metabolic associations are important to determine the functional outcomes of gut microbiota composition in the development of metabolic diseases. Table IIsummarizes the most significant bacterial groups associated with overweight in obese animals and humans. The specific role of gut microbiota components and their bioactive factors remains unclear in NAFLD. In addition, the direct role of gut microbiome in the development of obesity and its associated phenotypes has been demonstrated to be transmissible by fecal transplantation into germ-free mice (43,94). Here, bacterial colonization in obesity and associated disorders appears to be dependent on host metabolic responses to diet.

Table II. Summary of the most significant bacterial groups associated with overweight and obese animals and humans 

In addition to the gut microbiota metabolic participation in the modulation of lipid metabolism, it has also been associated with an indirect regulatory influence in the central nervous system (CNS), the so-called ‘gut-brain' axis. Thereby, microbial-mediated CNS control could influence food intake and expenditure, the regulation of hepatic lipogenesis, and gut integrity, as well as hepatic low-grade inflammation. Preclinical studies have shown that microbial colonization of germ-free mice can reduce the levels of circulating fasting-induced adipose factor (Fiaf), and the levels of phosphorylated AMP-activated protein kinase (both in the skeletal muscle and liver), thus contributing to fat storage (7,95) (Fig. 1). Taken together, these functional outcomes of gut microbiota beyond energy harvest from diet have led to the concept that gut microbiota serves as an important regulator of energy homeostasis and ectopic fat deposition. Nevertheless, the functional characterization of transplanted microbial communities suggest that variation in metabolic outcomes is under the control of both host genetics and gut microbiota (43).

Figure 1. Beneficial effects of short-chain fatty acids (SCFA) derived from bacterial fermentation of dietary fructo-oligosaccharides and galacto-oligosaccharides on non-alcoholic fatty liver (NAFL) development. 

Data from scarce nutritional interventions in humans also strongly suggest the microbiota-mediated positive effects in patients with nonalcoholic steatohepatitis (NASH) (96). A study in 66 patients who received a symbiotic mixture of Bifidobacterium longumwith FOS for 24 weeks concluded that, compared to lifestyle modification alone, dietary supplementation with the symbiotic formulation significantly reduced NASH activity index, serum concentrations of LDL cholesterol, inflammatory markers, and insulin resistance. Otherwise, knockout mice for phosphatidylethanolamine N-methyltransferase (PEMT) develop severe NASH (97). However, these mice are protected from obesity and whole-body insulin resistance. Here, the study of the influence of feeding prebiotics to animals with metabolic risk factors for NAFLD development has been inferential. Feeding the prebiotic kojibiose (2-O-α-D-glucopyranosyl-α-D-glucopyranose) to hyperglycaemic animals treated with arachidic acid significantly reduced the plasma levels of lyso-phosphatidylcholine (98). These changes were accompanied by body weight loss and increased numbers of Bifidobacteriumspp. (by 12 %), Bacteroidesspp. (by 2 %), and Enterobacteriaceae(by 4 %) relative to control animals.

POTENTIAL DUAL ROLE OF PROBIOTICS

To date there is a controversial debate concerning the species- or strain-dependent role, whether positive or negative, of Lactobacillusspp. in body weight gain (92,99). Different investigations point out specific alterations in gut microbiota composition causing markedly opposite regulatory effects on lipogenesis associated with increased numbers of Lactobacillusspp., and thereby Firmicutesin fecal contents. This is of particular importance since most probiotics for human consumption belong to this genus. While some species (L. acidophilusand L. reuteri) have been associated with obesity-associated gut microbiota (63,64), others (L. plantarumand L. gasseri) are associated with weight loss in obese humans and animals (63). Feeding L. ingluievito mice preceded an increase in weight and increased FAS and TNF-α transcript levels in the liver (93). L. acidophilusor L. caseiwere likely protective against diabetes since animals fed with these probiotics did not show any evidence of metabolic syndrome such as insulin resistance (100). However, in some cases strain-specific beneficial effects have been reported, for example, in patients dietary supplemented with L. plantarum299v or a yoghurt supplemented with L. acidophilus145, B. longum913 and oligofructose (101,102). Most prominent effects significantly reduced serum concentrations of LDL-cholesterol (by 9.6 %) and fibrinogen (101) or increased HDL-cholesterol (by 25.8 %) (102). Feeding L. rhamnosusGG and L. sakeiNR28, supplemented within a normal chow, to mice resulted in a significant reduction of epididymal fat mass, as well as obesity-related biomarkers like AA, FAS, and steatoryl-CoA desaturase-1 in the liver (103). Taken together, all studies seem to highlight strain-dependent effects on lipid metabolism and liver/adipose tissue inflammation for Bifidobacteriumand Bacteroidesspp. that counteract NAFL/NASH.

Another important aspect is that most species of these genera possess β-glucosidase activity contributing to nutrient utilization. Otherwise, metagenomic studies have also demonstrated the expression of β-glucuronidase activity in some species of Firmicutes(104). β-glucuronidase activity might contribute to the production of toxins and/or mutagens resulting from increased intestinal reuptake and hepatic retention of usually deconjugated, potentially toxic bile acids. However, the role of bacterial β-glucosidases in the development of NAFL/NASH needs further study, since opposite bacterial patterns have been found as a function of the aetiology of fatty liver disease. For example, enteric dysbiosis with relative abundance of Bacteroidetesand Verrucomicrobiacompared to Firmicutes, a pattern that should correlate with amelioration of NAFL/NASH or obesity, has been associated with alcohol-induced steatosis and steatohepatitis (105).

INNATE IMMUNITY

NAFLD has been strongly associated with gut injury and increased intestinal permeability, and NASH frequently recurs in patients who underwent weight reduction surgery to treat obesity. These conditions favour translocation of bacterial antigens across the intestinal mucosal wall into the portal circulation in 25-70 % of patients with liver disease, enhancing hepatic inflammation and disease progression to NASH (9,11). Studies in animals and humans indicate that gut-derived bacterial endotoxin (i.e., lipopolysaccharide, LPS) and other cell wall components play important roles in the development of NAFL and its progression to NASH by stimulating, for instance, toll like receptor (TLR)-4. This receptor is prominently expressed on innate immune cells, increasing the release of proinflammatory cytokines and chemokines favouring insulin resistance (106).

Immune crosstalk between gut microbiota and its host takes place by interaction of the gastrointestinal epithelium and different cell types in the gut-associated lymphatic tissue that possess TLRs (107 108-109). These receptors recognize specific molecular patterns (proteins, nucleic acids, lipopeptides, etc.) found in both commensals and pathogens. Especially, recent reports highlighted the potential key role of defined Bidifobacteriumspp. (109) and Lactobacillusspp. (108) strains as promising modulators of inflammatory activity by regulating TLR-2/4 and NF-κB expression. Amelioration of LPS-induced NF-κB activation by potentially beneficial bacteria could, at least in part, explain their positive effects on the inflammatory signals originating in the intestine, preventing further amplification for those within the gut liver axis. The importance of innate immunity has been demonstrated in fructose-treated mice, as a model of steatosis characterized by high lipogenesis, where increased accumulation of 16S rDNA sequences in the liver was associated with significant induction of TLR 1-4 and 6-8 (34). However, mice genetically deficient in TLR5 also exhibit hyperphagia and develop hyperlipidemia, hypertension, insulin resistance, and increased adiposity (110). In contrast, TLR2-deficient mice under germ-free conditions are protected from diet-induced insulin resistance resulting in a phenotype reminiscent of metabolic syndrome (111). These animals were characterized by differences in gut microbiota, with a 3-fold increase in Firmicutesand a slight increase in Bacteroidetesas compared with controls, although both were present in similar proportions (47.9 %) (111). These authors suggested that the presence of gut microbiota could reverse the phenotype of an animal developing as a consequence of glucose intolerance and later obesity. Fructose disposition (solid or liquid formulations) has been shown to differentially affect gut microbiota composition and Bacteroidescolonization in mice (112). The decreased numbers of, among others, Bacteroidesspp. and Lactobacillusspp. were associated with up-regulated protein expression in the lipopolysaccharide-dependent proinflammatory TLR4/NLRP3 inflammasome pathway. Recovery of the intestinal Bacteroidetespopulation could contribute to better balance immunostimulatory thresholds because of the pivotal role played by the outer membrane vesicles (OMVs) of Gram-negative bacteria and their interaction with TLRs. Bacteroides uniformisCECT 7771 was reported to help in developing adequate immune responses, improving macrophage activity and liver steatosis (reduced fat vacuoles in liver tissue) in mice (113).

Studies conducted in rodent NASH models have demonstrated a direct implication of gut microbiota in enhancing NASH severity (114). When feeding a methionine-deficient diet for 4 weeks, animals develop defective NRLP3 and NRPL6 inflammasome (damage-associated tissue) sensing. This nutritional deficiency causes an aberrant colitogenic intestinal microbiota characterized by increased proportions of Bacteroidetes(Prevotellaceae) and bacterial phylum TM7, and reductions in the genus Lactobacillus in the Firmicutes phylum (115). This microbiota composition, although contrasting to that reported in obese and T2D patients, was identified as responsible for colonic inflammation (CCL5 secretion), favouring the translocation of TLR-4/9 stimulants towards the portal circulation (114). Similar inflammatory conditions were reported in fibrogenic animals administered CCl4 associated to bacterial translocation with increased proportions of aerobic bacteria and decreased numbers of Gram-positive anaerobics in the Clostridiumgroup (116).

Metabolic endotoxemia can result from multiple mechanisms, including increased production of endotoxins upon changes in gut microbiota composition, and triggers TLR4 receptor complex (i.e., TLR4/MD2/CD14) signaling in murine liver cells (mice fed a choline-deficient diet for 3 weeks) to fuel liver and adipose tissue inflammation (117). Thus, a functional relationship between metabolism and the innate immune system is increasingly recognized, suggesting that innate immune alterations can favour the development of metabolic syndrome (109,118). In this regard food ingestion, even in lean subjects, triggers a mild inflammatory response in the liver and adipose tissue, which usually does not lead to negative consequences. However, with the advent of immunometabolic imbalances such as obesity coupled with an unfavourable lifestyle and certain medications or genetic predisposition, inflammatory responses can get out of control, thus contributing to NASH development and severity (119). Accordingly, high calorie feeding induced a postprandial increase in surrogate serum markers of liver fibrosis in patients with NAFL and T2D (120). Here, inflammation is driven by classically activated (M1 polarized) macrophages, both in the liver and adipose tissue (121), where TLR4 receptor complex activity significantly contributes to macrophage polarization and functional differentiation (117,122). Thus, TLR4-/-mice fed a choline-deficient diet (for 22 weeks) displayed reduced hepatic macrophage infiltration with decreased macrophage chemoattractant protein (MCP)-1 and chemokine-chemokine receptor (CCR)-2 expression (123). Also, the hepatic proportion of F4/80+macrophages significantly increased in GPR43-/-mice fed a high-fat diet (15). Taken together, these studies highlight a direct immunometabolic control by microbial metabolites on the immune innate cell population of macrophages. In this regard, microbial butyrate has been proven to modulate macrophage metabolism, reducing mTOR kinase activity and increasing their antimicrobial phenotype (124).

HOST GENOME-MICROBIOME INTERACTION

Accumulated evidence regarding the immunometabolic implications of intestinal dysbiosis proved the (epi)genomic relationship between the exposome and host genetics to influence gut microbiome. The bidirectional interaction of gut microbiota and its microbiome with the host's physiological processes also induces changes in bacterial metabolism. This metabolic adaptation can lead to pattern changes in microbial metabolites, which may be important indirect regulators of the host epigenome, worsening or improving the development of liver disease. An example of this microbiome adaptation involves the reported changes in the gene composition of gut microbiota, which are similar to those seen in the gut microbiota of patients suffering from inflammatory bowel disease as compared to lean controls (86,125). Obesity and type-2 diabetes microbiomes present the hallmarks of adaptation to low microbial diversity environments (43,125). These microbiomes are enriched with genes encoding enzymes frequently involved in membrane transportation (125) and exhibit differences in glucose-stimulated insulin secretion (43). Furthermore, these microbiomes are depleted of cofactor, vitamin and nucleotide metabolism and transcription (125). Genomic studies have shown a strong influence of microbial metabolites (SCFAs) in the regulation of epigenetic programming in different critical tissues (liver, white adipose tissue, and proximal colon), determining the onset and severity of NAFLD (125). Collectively, these studies demonstrated that intestinal dysbiosis yields a core microbiome closely related at a functional level to systemic immunometabolic diseases and different physiologic states.

The pathogenesis and severity of NAFLD has been associated with several nonsynonymous single-nucleotide polymorphisms in the PNPLA3gene (rs738409, rs143392071) (127), increasing the risk for both steatosis and fibrosis (128); the MTTP (rs756998920) gene (129), and the adiponectin (rs1501299) gene (61). Also, a splicing mutation in TM6SF2 (rs58542926) affecting postprandial lipoprotein metabolism and glucose homeostasis in NAFLD has been identified (130). Research advances in this sense determined that the PNPLA3, TM6SF2, and MBOAT7 variants are associated with increased liver injury. The TM6SF2 variant seems to modulate predominantly hepatic fat accumulation, and the MBOAT7 polymorphism is commonly linked to fibrosis.

Data from animal models indicate that maternal overnutrition, even in the absence of pregnancy-related obesity, can promote metabolic dysregulation and predispose to T2D in the offspring (131). Metabolic programming of disease is being increasingly thought to occur via epigenetic changes as integral underlying mechanisms (132). Accordingly, epigenetic studies in diabetic and obese subjects revealed significant methylation differences in the promoter region of innate immune mediators such as TLR-2 and -4 (133), and FFAR3 (134) in comparison to lean controls, suggesting that changes in gut microbiota are involved in the epigenetic regulation of inflammatory reactions.

CONCLUSIONS

NAFLD has been revealed to be a severe liver dysfunction with an increasing prevalence in both obese and lean patients with metabolic syndrome where overnutrition and sedentary lifestyles are considered major contributors. There is a cumulative body of evidence for a correlation between alterations in microbial diversity and the functional output of gut microbiota. These changes appear as important modulators for the development of NAFLD or its progression toward NASH. Phylogenetic changes occur in the microbiota of obese versus lean individuals, and are characterized by an increased Firmicutesto Bacteroidetesratio. Here, the relationship between Lactobacillusspp. and Bifidobacteriumspp. appears to be critical for energy harvest and endocrine regulation. Thus, it deserves special attention since many probiotic strains within these genera are marketed for human consumption and, together with prebiotics, represent dietary strategies that shape either gut microbiota composition and/or its metabolic activity. The latter effects appear to be mediated not only at species but also at strain level. This points to the potential use of gut microbiota composition as predictive/therapeutic tool for NAFLD progression and severity. Notwithstanding this, microbiome-targeted interventions have shown promising but unsatisfactory clinical benefits. Microbiome-targeted therapies have shown significant immune or metabolic benefits for a minority of patients. The sharp influence of exogenous (i.e., diet and lifestyle, geography, prenatal nutrition) and endogenous (i.e., genes, ethnicity) factors on the transgenerational inheritance of heterogeneity in human gut microbiome composition still appears to represent a key step determining the clinical benefits for NAFLD patients. Overall, there is still a clinical need for contributions a) to understand the potential role of specific consortia of microbes; b) to foster interdisciplinary science establishing variations in gut microbiota composition and its genome within a ‘score of disease'; and c) to guide future therapeutic and management strategies for most patients.

REFERENCES

1. Younossi Z, Anstee QM, Marietti M, Hardy T, Henry L, Eslam M, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 2018;15(1):11-20. [ Links ]

2. Conus F, Rabasa-Lhoret R, Peronnet F. Characteristics of metabolically obese normal-weight (MONW) subjects. Appl Physiol Nutr Metab 2007;32:4-12. [ Links ]

3. Solga SF, Diehl AM. Non-alcoholic fatty liver disease: lumen-liver interactions and possible role for probiotics. J Hepatol 2003;38(5):681-7. [ Links ]

4. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev 2010;90(3):859-904. [ Links ]

5. Parnell JA, Raman M, Rioux KP, Reimer RA. The potential role of prebiotic fibre for treatment and management of non-alcoholic fatty liver disease and associated obesity and insulin resistance. Liver International 2012;32(5):707-11. [ Links ]

6. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007;56: 1761-72. [ Links ]

7. Cani PD, Osto M, Geurts L, Everard A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes 2012;3(4):279-88. [ Links ]

8. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334(6052):105-8. [ Links ]

9. Szabo G, Bala S, Petrasek J, Gattu A. Gut-liver axis and sensing microbes. Dig Dis 2010;28(6):737-44. [ Links ]

10. Musso G, Gambino R, Cassader M. Gut microbiota as a regulator of energy homeostasis and ectopic fat deposition: mechanisms and implications for metabolic disorders. Curr Opin Lipidol 2010;21:76-83. [ Links ]

11. Compare D, Coccoli P, Rocco A, Nardone OM, De Maria S, CartenìM, et al. Gut-liver axis: The impact of gut microbiota on non-alcoholic fatty liver disease. Nutr Metab Cardiovasc Dis 2012;22(6):471-6. [ Links ]

12. Anty R, Lemoine M. Liver fibrogenesis and metabolic factors. Clin Res Hepatol Gastroenterol 2011;35(S1):S10-20. [ Links ]

13. Schattenberg JM, Schuppan D. Non-alcoholic steatohepatitis: the therapeutic hcalenge of a global epidemic. Curr Opin Lipidol 2011;22(6):479-88. [ Links ]

14. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA 2004;101:15718-23. [ Links ]

15. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun 2013;4:1829. [ Links ]

16. Kindt A, Liebisch G, Clavel T, Haller D, Hörmannsperger G, Yoon H, et al. The gut microbiota promotes hepatic fatty acid desaturation and elongation in mice. Nat Commun 2018;9(1):3760. [ Links ]

17. Mao K, Baptista AP, Tamoutounour S, Zhuang L, Bouladoux N, Martins AJ, et al. Innate and adaptive lymphocytes sequentially shape the gut microbiota and lipid metabolism. Nature 2018;554(7691):255-9. [ Links ]

18. Lynch C, Chan CS, Drake AJ. Early life programming and the risk of non-alcoholic fatty liver disease. J Dev Orig Health Dis 2017;8(3):263-72. [ Links ]

19. VandeVusse L, Hanson L, Safdar N. Perinatal outcomes of prenatal probiotic and prebiotic administration: an integrative review. J Perinat Neonatal Nurs 2013;27(4):288-301. [ Links ]

20. Tian Y, Mok MT, Yang P, Cheng AS. Epigenetic Activation of Wnt/?-Catenin Signaling in NAFLD-Associated Hepatocarcinogenesis. Cancers (Basel) 2016;8(8):E76. [ Links ]

21. Cortese R, Lu L, Yu Y, Ruden D, Claud EC. Epigenome-Microbiome crosstalk: A potential new paradigm influencing neonatal susceptibility to disease. Epigenetics 2016;11(3):205-15. [ Links ]

22. Campo L, Eiseler S, Apfel T, Pyrsopoulos N. Fatty Liver Disease and Gut Microbiota: A Comprehensive Update. J Clin Transl Hepatol 2019;7(1): 56-60. [ Links ]

23. Augustyn M, Grys I, Kukla M. Small intestinal bacterial overgrowth and nonalcoholic fatty liver disease. Clin Exp Hepatol 2019;5(1):1-10. [ Links ]

24. Cho MS, Kim SY, Suk KT, Kim BY. Modulation of gut microbiome in nonalcoholic fatty liver disease: pro-, pre-, syn-, and antibiotics. J Microbiol 2018;56(12):855-67. [ Links ]

25. Sharpton SR, Maraj B, Harding-Theobald E, Vittinghoff E, Terrault NA. Gut microbiome-targeted therapies in nonalcoholic fatty liver disease: a systematic review, meta-analysis, and meta-regression. Am J Clin Nutr 2019; pii:nqz042. DOI:10.1093/ajcn/nqz042 [ Links ]

26. Al-Lahham S, Roelofsen H, Rezaee F, Weening D, Hoek A, Vonk R, et al. Propionic acid affects immune status and metabolism in adipose tissue from overweight subjects. Eur J Clin Invest 2012;42(4):357-64. [ Links ]

27. Abu-Shanab A, Quigley EM. The role of the gut microbiota in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2010;7(12):691-701. [ Links ]

28. Cavaglieri CR, Nishiyama A, Fernandes LC, Curi R, Miles EA, Calder PC. Differential effects of short-chain fatty acids on proliferation and production of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sci 2003;73(13):1683-90. [ Links ]

29. Muccioli GG, Naslain D, Backhed F, Reigstad CS, Lambert DM, Delzenne NM, et al. The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol 2010;6:392. [ Links ]

30. Serino M, Chabo C, Burcelin R. Intestinal microbiomics to define health and disease in human mice. Curr Pharm Biotechnol 2012;13(5):746-58. [ Links ]

31. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun 2018;9(1):3294. [ Links ]

32. Spencer MD, Hamp TJ, Reid RW, Fischer LM, Zeisel SH, Fodor AA. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 2011;140(3):976-86. [ Links ]

33. Corbin KD, Zeisel SH. Choline metabolism provides novel insights into non-alcoholic fatty liver disease and its progression. Curr Opin Gastroenterol 2012;28:159-65. [ Links ]

34. Wagnerberger S, Spruss A, Kanuri G, Volynets V, Stahl C, Bischoff SC, et al. Toll-like receptors 1-9 are elevated in livers with fructose-induced hepatic steatosis. Br J Nutr 2012;107:1727-38. [ Links ]

35. Bellanti F, Villani R, Tamborra R, Blonda M, Iannelli G, di Bello G, et al. Synergistic interaction of fatty acids and oxysterols impairs mitochondrial function and limits liver adaptation during nafld progression. Redox Biol 2018;15: 86-96. [ Links ]

36. Hooper LV, Midtvedt T, Gordon JI. How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr 2002;22:283-307. [ Links ]

37. Cani PD, Delzenne NM. The gut microbiome as therapeutic target. Pharmacology & Therapeutics 2011;130:202-12. [ Links ]

38. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Metagenomic analysis of the human distal gut microbiome. Science 2006;312:1355-9. [ Links ]

39. Flint HJ, Bayer EA, Rincon MT, Lamed R, White BA. Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis. Nat Rev Microbiol 2008;6:121-31. [ Links ]

40. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006;444:1027-31. [ Links ]

41. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008;57(6):1470-81. [ Links ]

42. Kreznar JH, Keller MP, Traeger LL, Rabaglia ME, Schueler KL, Stapleton DS, et al. Host Genotype and Gut Microbiome Modulate Insulin Secretion and Diet-Induced Metabolic Phenotypes. Cell Rep 2017;18(7):1739-50. [ Links ]

43. Canfora EE, van der Beek CM, Hermes GDA, Goossens GH, Jocken JWE, Holst JJ, et al. Supplementation of Diet with Galacto-oligosaccharides Increases Bifidobacteria, but Not Insulin Sensitivity, in Obese Prediabetic Individuals. Gastroenterology 2017;153(1):87-97. [ Links ]

44. Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ, Fagerberg B, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013;498(7452):99-103. [ Links ]

45. Pachikian BD, Essaghir A, Demoulin JB, Neyrinck AM, Catry E, De Backer FC, et al. Hepatic n-3 polyunsaturated fatty acid depletion promotes steatosis and insulin resistance in mice:genomic analysis of cellular targets. PLoS One 2011;6(8):e23365. [ Links ]

46. Delzenne NM, Neyrinck AM, Cani PD. Gut microbiota and metabolic disorders: How prebiotic can work Br J Nutr 2013;109(S2):S81-5. [ Links ]

47. Sauer J, Richter KK, Pool-Zobel BL. Products formed during fermentation of the prebiotic inulin with human gut flora enhance expression of biotransformation genes in human primary colon cells. Br J Nutr 2007;97(5):928-37. [ Links ]

48. Delzenne NM, Kok NN. Biochemical basis of oligofructose-induced hypolipidemia in animal models. J Nutr 1999;129:1467S-70S. [ Links ]

49. Yin X, Peng J, Zhao L, Yu Y, Zhang X, Liu P, et al. Structural changes of gut microbiota in a rat non-alcoholic fatty liver disease model treated with a Chinese herbal formula. Syst Appl Microbiol 2013;36:188-96. [ Links ]

50. Xiong Y, Miyamoto N, Shibata K, Valasek MA, Motoike T, Kedzierski RM, et al. Short-chain fatty acids stimulate leptin production in adipocytes through the G-protein-coupled receptor GRP41. Proc Nat Acad Sci USA 2004;101:1045-50. [ Links ]

51. Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr 2009;90:1236-43. [ Links ]

52. Zaibi MS, Stocker CJ, O'Dowd J, Davies A, Bellahcene M, Cawthorne MA, et al. Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett 2010;584(11):2381-6. [ Links ]

53. Agheli N, Kabir M, Berni-Canani S, Petitjean E, Boussairi A, Luo J, et al. Plasma lipids and fatty acid synthase activity are regulated by short chain fructo-oligosaccharides in sucrose-fed insulin-resistant rats. J Nutr 1998;128:1283-8. [ Links ]

54. Chambers ES, Byrne CS, Morrison DJ, Murphy KG, Preston T, Tedford C, et al. Dietary supplementation with inulin-propionate ester or inulin improves insulin sensitivity in adults with overweight and obesity with distinct effects on the gut microbiota, plasma metabolome and systemic inflammatory responses: a randomised cross-over trial. Gut 2019;68(8):1430-8. DOI:10.1136/gutjnl-2019-318424. Epub 2019 Apr 10 [ Links ]

55. Cani PD, Daubioul CA, Reusens B, Remacle C, Catillon G, Delzenne NM. Involvement of glucagon-like peptide 1 (7-36) amide on glycaemia-lowering effect of oligofructose in streptocin-treated rats. J Endocrinol 2005;185:457-65. [ Links ]

56. Guarner F. Inulin and oligofructose: impact on intestinal diseases and disorders. Br J Nutr 2005;93(S1):S61-5. [ Links ]

57. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009;58(8):1091-103. [ Links ]

58. Chen M, Hui S, Lang H, Zhou M, Zhang Y, Kang C, et al. SIRT3 Deficiency Promotes High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease in Correlation with Impaired Intestinal Permeability through Gut Microbial Dysbiosis. Mol Nutr Food Res 2019;63(4):e1800612. [ Links ]

59. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med 2009;1:6ra14. [ Links ]

60. Liu J, Xing J, Wang B, Wei C, Yang R, Zhu Y, et al. Correlation Between Adiponectin Gene rs1501299 Polymorphism and Nonalcoholic Fatty Liver Disease Susceptibility: A Systematic Review and Meta-Analysis. Med Sci Monit 2019;25:1078-86. [ Links ]

61. Xu X, Fukui H, Ran Y, Tomita T, Oshima T, Watari J, et al. Alteration of GLP-1/GPR43 expression and gastrointestinal motility in dysbiotic mice treated with vancomycin. Sci Rep 2019;9(1):4381. [ Links ]

62. Million M, Angelakis E, Paul M, Armougom F, Leibovici L, Raoult D. Comparative meta-analysis of the effect of Lactobacillus species on weight gain in humans and animals. Microb Pathog 2012;53(2):100-8. [ Links ]

63. Million M, Maraninchi M, Henry M, Armougom F, Richet H, Carrieri P, et al. Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii. Int J Obes (Lond) 2012;36(6):817-25. [ Links ]

64. Neis EP, Dejong CH, Rensen SS. The role of microbial amino acid metabolism in host metabolism. Nutrients 2015;7(4):2930-46. [ Links ]

65. Zelber-Sagi S, Nitzan-Kaluski D, Goldsmith R, Webb M, Blendis L, Halpern Z, et al. Long term nutritional intake and the risk for non-alcoholic fatty liver disease (NAFLD): a population based study. J Hepatol 2007;47(5):711-7. [ Links ]

66. Pachikian BD, Neyrinck AM, Portois L, De Backer FC, Sohet FM, Hacquebard M, et al. Involvement of gut microbial fermentation in the metabolic alterations occurring in n-3 polyunsaturated fatty acids-depleted mice. Nutr Metab (Lond) 2011;8(1):44. [ Links ]

67. Petit JM, Guiu B, Duvillard L, Jooste V, Brindisi MC, Athias A, et al. Increased erythrocytes n-3 and n-6 polyunsaturated fatty acids is significantly associated with a lower prevalence of steatosis in patients with type 2 diabetes. Clin Nutr 2011;31(4):520-5. [ Links ]

68. Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lip Res 2006;47:241-59. [ Links ]

69. Kitahara M, Takamine F, Imamura T, Benno Y. Assignment of Eubacterium sp. VPI 12708 and related strains with high bile acid 7a-dehydroxylating activity to Clostridium scindens and proposal of Clostridium hylemonae sp. nov., isolated from human feces. Int J Syst Evol Microbiol 2000;50:971-8. [ Links ]

70. Marounek M, Duskova D. Metabolism of pectin in rumen bacteria Butyrivibrio fibrisolvens and Prevotella ruminicola. Lett Appl Microbiol 1999;29:429-33. [ Links ]

71. Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci USA 2009;106:2365-70. [ Links ]

72. Takamine F, Imamura T. 7b-dehydroxylation of 3,7-dihydroxy bile acids by a Eubacterium species strain C-25 and stimulation of 7b-dehydroxylation by Bacteroides distasonis strain K-5. Microbiol Immun 1985;29:1247-252 [ Links ]

73. Cyphert HA, Ge X, Kohan AB, Salati LM, Zhang Y, Hillgartner FB. Activation of the farnesoid X receptor induces hepatic expression and secretion of fibroblast growth factor 21. J Biol Chem 2012;287:25123-38. [ Links ]

74. Ip E, Farrell GC, Robertson G, Hall P, Kirsch R, Leclercq I. Central role of PPARalpha-dependent hepatic lipid turnover in dietary steatohepatitis in mice. Hepatology 2003;38:123-32. [ Links ]

75. Prawitt J, Abdelkarim M, Stroeve JH, Popescu I, Duez H, Velagapudi VR, et al. Farnesoid receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes 2011;60:1861-71. [ Links ]

76. Ge X, Yin L, Ma H, Li T, Chiang JY, Zhang Y. Aldo-keto reductase 1B7 is a target gene of FXR and regulates lipid and glucose homeostasis. J Lipid Res 2011;52:1561-8. [ Links ]

77. Wang YD, Chen WD, Yu D, Forman BM, Huang W. The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor kappa light-chain enhancer of activated B cells (NF-kappaB) in mice. Hepatology 2011;54:1421-32. [ Links ]

78. Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, et al. TGR5- mediated bile acid sensing controls glucose homeostasis. Cell Metab 2009;10:167-77. [ Links ]

79. Nagata S, Chiba Y, Wang C, Yamashiro Y. The effects of the Lactobacillus casei strain on obesity in children:a pilot study. Benef Microbes 2017;8(4):535-43. [ Links ]

80. Bastian WP, Hasan I, Lesmana CRA, Rinaldi I, Gani RA. Gut Microbiota Profiles in Nonalcoholic Fatty Liver Disease and Its Possible Impact on Disease Progression Evaluated with Transient Elastography: Lesson Learnt from 60 Cases. Case Rep Gastroenterol 2019;13(1):125-33. [ Links ]

81. McCoy KD, Burkhard R, Geuking MB. The microbiome and immune memory formation. Immunol Cell Biol 2019;97(7):625-35. DOI:10.1111/imcb.12273 [ Links ]

82. Miele L, Valenza V, La Torre G, Montalto M, Cammarota G, Ricci R, et al. Increased intestinal permeability and tight junction alterations in non-alcoholic fatty liver disease. Hepatology 2009;49:1877-87. [ Links ]

83. Brandt A, Hernández-Arriaga A, Kehm R, Sánchez V, Jin CJ, Nier A, et al. Metformin attenuates the onset of non-alcoholic fatty liver disease and affects intestinal microbiota and barrier in small intestine. Sci Rep 2019;9(1):6668. [ Links ]

84. Fitriakusumah Y, Lesmana CRA, Bastian WP, Jasirwan COM, Hasan I, Simadibrata M, et al. The role of Small Intestinal Bacterial Overgrowth (SIBO) in Non-alcoholic Fatty Liver Disease (NAFLD) patients evaluated using Controlled Attenuation Parameter (CAP) Transient Elastography (TE): a tertiary referral center experience. BMC Gastroenterol 2019;19(1):43. [ Links ]

85. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean twins. Nature 2009;457:480-4. [ Links ]

86. Million M, Angelakis E, Maraninchi M, Henry M, Giorgi R, Valero R, et al. Correlation between body mass index and gut concentrations of Lactobacillus reuteri, Bifidobacterium animalis, Methanobrevibacter smithii and Escherichia coli. Int J Obes (Lond) 2013;37(11):1460-6. [ Links ]

87. Kalliomaki M, Collado MC, Salminen S, Isolauri E. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr 2008;87:534-8. [ Links ]

88. Santacruz A, Collado MC, García-Valdés L, Segura MT, Martín-Lagos JA, Anjos T, et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br J Nutr 2010;104(1):83-92. [ Links ]

89. Balamurugan R, George G, Kabeerdoss J, Hepsiba J, Chandragunasekaran AM, Ramakrishna BS. Quantitative differences in intestinal Faecalibacterium prausnitzii in obese Indian children. Br J Nutr 2010;103:335-8. [ Links ]

90. Duarte SMB, Stefano JT, Miele L, Ponziani FR, Souza-Basqueira M, Okada LSRR, et al. Gut microbiome composition in lean patients with NASH is associated with liver damage independent of caloric intake:a prospective pilot study. Nutr Metab Cardiovasc Dis 2018; 28: 369-84. [ Links ]

91. Armougom F, Henry M, Vialettes B, Raccah D, Raoult D. Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. PLoS One 2009; 4: e7125. [ Links ]

92. Angelakis E, Bastelica D, Ben Amara A, El Filali A, Dutour A, Mege JL, et al. An evaluation of the effects of Lactobacillus ingluievi on body weight, the intestinal microbiome and metabolism in mice. Micro Pathog 2012;52(1):61-8. [ Links ]

93. Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013; 341(6150): 1241214. [ Links ]

94. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA 2007;104:979-84. [ Links ]

95. Malaguarnera M, Vacante M, Antic T, Giordano M, Chisari G, Acquaviva R, et al. Bifidobacterium longum with fructo-oligosaccharides in patients with non-alcoholic steatohepatitis. Dig Dis Sci 2012;57:545-53. [ Links ]

96. van der Veen JN, Lingrell S, McCloskey N, LeBlond ND, Galleguillos D, Zhao YY, et al. A role for phosphatidylcholine and phosphatidylethanolamine in hepatic insulin signaling. FASEB J 2019;33(4):5045-57. [ Links ]

97. Laparra JM, Díez-Municio M, Moreno FJ, Herrero M. Kojibiose ameliorates arachidic acid-induced metabolic alterations in hyperglycaemic rats. Br J Nutr 2015;114(9):1395-402. [ Links ]

98. Luoto R, Kalliomäki M, Laitinen K, Delzenne NM, Cani PD, Salminen S, et al. Initial dietary and microbiological environments deviate in normal-weight compared to overweight children at 10 years of age. J Pediatr Gastroenterol Nutr 2011;52:90-5. [ Links ]

99. Yadav H, Jain S, Sinha P. Oral administration of dahi containing probiotic Lactobacillus acidophilus and Lactobacillus casei delayed the progression of streptozotocininduced diabetes in rats. J Dairy Res 2008;75:189-95. [ Links ]

100. Bukowska H, Pieczul-Mróz J, Jastrzebska M, Chełstowski K, Naruszewicz M. Decrease in fibrinogen and LDL-cholesterol levels upon supplementation of diet with Lactobacillus plantarum in subjects with moderately elevated cholesterol. Atherosclerosis 1998;137(2):437-8. [ Links ]

101. Kiessling G, Schneider J, Jahreis G. Long-term consumption of fermented dairy products over 6 months increases HDL cholesterol. Eur J Clin Nutr 2002;56(9):843-9. [ Links ]

102. Ji YS, Kim HN, Park HJ, Lee JE, Yeo SY, Yang JS, et al. Modulation of the murine microbiome with a concomitant anti-obesity effect by Lactobacillus rhamnosus GG and Lactobacillus sakei NR28. Benef Microbes 2012;3(1):13-22. [ Links ]

103. Dabek M, McCrae SI, Stevens VJ, Duncan SH, Louis P. Distribution of beta-glucosidase and beta-glucuronidase activity and of beta-glucuronidase gene gus in human colonic bacteria. FEMS Microbiol Ecol 2008;66:487-95. [ Links ]

104. Yan AW, Fouts DE, Brandl J, Stärkel P, Torralba M, Schott E, et al. Enteric dysbiosis associated with a mouse model of alcoholic liver disease. Hepatology 2011;53(1):96-105. [ Links ]

105. Li Z, Yang S, Lin H, Huang J, Watkins PA, Moser AB, et al. Probiotics and antibodies to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology 2003;37(2):343-50. [ Links ]

106. Shibolet O, Podolsky DK. TLRs in the Gut. IV. Negative regulation of Toll-like receptors and intestinal homeostasis: addition by subtraction. Am J Physiol Gastrointest Liver Physiol 2007;292(6):G1469-73. [ Links ]

107. Lee SI, Kim HS, Koo JM, Kim IH. Lactobacillus acidophilus modulates inflammatory activity by regulating the TLR4 and NF-?B expression in porcine peripheral blood mononuclear cells after lipopolysaccharide challenge. Br J Nutr 2016;115(4):567-75. [ Links ]

108. Castro-Bravo N, Margolles A, Wells JM, Ruas-Madiedo P. Exopolysaccharides synthesized by Bifidobacterium animalis subsp. lactis interact with TLR4 in intestinal epithelial cells. Anaerobe 2019;56:98-101. [ Links ]

109. Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 2010;328(5975):228-31. [ Links ]

110. Caricilli AM, Picardi PK, de Abreu LL, Ueno M, Prada PO, Ropelle ER, et al. Gut microbiota is a key modulator of insulin resistance in TLR 2 knockout mice. PLoS Biol 2011;9(12):e1001212. [ Links ]

111. Mastrocola R, Ferrocino I, Liberto E, Chiazza F, Cento AS, Collotta D, et al. Fructose liquid and solid formulations differently affect gut integrity, microbiota composition and related liver toxicity:a comparative in vivo study. J Nutr Biochem 2018;55:185-99. [ Links ]

112. Gauffin Cano P, Santacruz A, Moya Á, Sanz Y. Bacteroides uniformis CECT 7771 ameliorates metabolic and immunological dysfunction in mice with high-fat-diet induced obesity. PLoS One 2012;7(7):e41079. [ Links ]

113. Henao-Mejia J, Elinav E, Jin C, Hao L, Mehal WZ, Strowig T, et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012;482(7384):179-85. [ Links ]

114. Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011;145:745-57. [ Links ]

115. Gómez-Hurtado I, Santacruz A, Peiró G, Zapater P, Gutiérrez A, Pérez-Mateo M, et al. Gut microbiota dysbiosis is associated with inflammation and bacterial translocation in mice with CCl4-induced fibrosis. PLoS One 2011;6(7):e23037. [ Links ]

116. Rivera CA, Adegboyega P, van Rooijen N, Tagalicud A, Allman M, Wallace M. Toll-like receptor-4 signalling and Kupfer cells play pivotal roles in the pathogenesis of non-alcoholic esteatohepatitis. J Hepatology 2007;47:571-9. [ Links ]

117. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol 2010;72:219-46. [ Links ]

118. Hotamisligil GS. Inflammation and metabolic disorders. Nature 2006;444(7121):860-7. [ Links ]

119. Khoo EY, Stevenson MC, Leverton E, Cross R, Eriksson JW, Poucher SM, et al. Elevation of Alanine Transaminase and Markers of Liver Fibrosis After a Mixed Meal Challenge in Individuals with Type 2 Diabetes. Dig Dis Sci 2012;57(11):3017-25. [ Links ]

120. Lee YS, Park MS, Choung JS, Kim SS, Oh HH, Choi CS, et al. Glucagon-like peptide-1 inhibits adipose tissue macrophage infiltration and inflammation in an obese mouse model of diabetes. Diabetologia 2012;55(9):2456-68. [ Links ]

121. Hardy OT, Kim A, Ciccarelli C, Hayman LL, Wiecha J. Increased Toll-like receptor (TLR) mRNA expression in monocytes is a feature of metabolic syndrome in adolescents. Pediatr Obes 2013;8(1):e19-23. [ Links ]

122. Miura K, Yang L, van Rooijen N, Ohnishi H, Seki E. Hepatic recruitment of macrophages promotes nonalcoholic steatohepatitis through CCR2. Am J Physiol Gastrointest Liver Physiol 2012;302(11):G1310-21. [ Links ]

123. Schulthess J, Pandey S, Capitani M, Rue-Albrecht KC, Arnold I, Franchini F, et al. The Short Chain Fatty Acid Butyrate Imprints an Antimicrobial Program in Macrophages. Immunity 2019;50(2):432-45. [ Links ]

124. Greenblum S, Turnbaugh PJ, Borenstein E. Metagenomic systems biology of the human gut microbiome reveals topological shifts associated with obesity and inflammatory bowel disease. Proc Natl Acad Sci USA 2012;109(2):594-9. [ Links ]

125. Krautkramer KA, Kreznar JH, Romano KA, Vivas EI, Barrett-Wilt GA, Rabaglia ME, et al. Diet-Microbiota Interactions Mediate Global Epigenetic Programming in Multiple Host Tissues. Mol Cell 2016;64:982-92. [ Links ]

126. Mazo DF, Malta FM, Stefano JT, Salles APM, Gomes-Gouvea MS, Nastri ACS, et al. Validation of PNPLA3 polymorphisms as risk factor for NAFLD and liver fibrosis in an admixed population. Ann Hepatol 2019;18(3):466-471. DOI:10.1016/j.aohep.2018.10.004. Epub 2019 Apr 18 [ Links ]

127. Krawczyk M, Rau M, Schattenberg JM, Bantel H, Pathil A, Demir M, et al. Combined effects of the PNPLA3 rs738409, TM6SF2 rs58542;926, and MBOAT7 rs641738 variants on NAFLD severity: a multicenter biopsy-based study. J Lipid Res 2017;58(1):247-55. [ Links ]

128. Boonvisut S, Yoshida K, Nakayama K, Watanabe K, Miyashita H, Iwamoto S. Identification of deleterious rare variants in MTTP, PNPLA3, and TM6SF2 in Japanese males and association studies with NAFLD. Lipids Health Dis 2017;16(1):183. [ Links ]

129. Musso G, Cipolla U, Cassader M, Pinach S, Saba F, De Michieli F, et al. TM6SF2 rs58542;926 variant affects postprandial lipoprotein metabolism and glucose homeostasis in NAFLD. J Lipid Res 2017;58(6):1221-9. [ Links ]

130. Latouche C, Heywood SE, Henry SL, Ziemann M, Lazarus R, El-Osta A, et al. Maternal overnutrition programs changes in the expression of skeletal muscle genes that are associated with insulin resistance and defects of oxidative phosphorylation in adult male rat offspring J Nutr 2014;144(3):237-44. [ Links ]

131. Bäckhed F. Programming of host metabolism by the gut microbiota. Ann Nutr Metab 2011;58(S2):44-52. [ Links ]

132. Remely M, Aumueller E, Jahn D, Hippe B, Brath H, Haslberger AG. Microbiota and epigenetic regulation of inflammatory mediators in type 2 diabetes and obesity. Benef Microbes 2014;5(1):33-43. [ Links ]

133. Remely M, Aumueller E, Merold C, Dworzak S, Hippe B, Zanner J, et al. Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene 2014;537(1):85-92. [ Links ]

134. Kok NN, Morgan LM, Williams CM, Roberfroid MB, Thissen JP, Delzenne NM. Insulin, glucagon-like peptide 1, glucose-dependent insulinotropic polypeptide and insulin-like growth factor I as putative mediators of the hypolipidemic effect of oligofructose in rats. J Nutr 1998;128(7):1099-103. [ Links ]

135. Wada T, Sugatani J, Terada E, Ohguchi M, Miwa M. Physicochemical characterization and biological effects of inulin enzymatically synthesized from sucrose. J Agric Food Chem 2005;53:1246-53. [ Links ]

136. Sugatani J, Wada T, Osabe M, Yamakawa K, Yoshinari K, Miwa M. Dietary inulin alleviates hepatic steatosis and xenobiotics-induced liver injury in rats fed a high-fat and high-sucrose diet: association with the hepatic suppression of cytochrome P450 and hepatocyte nuclear factor 4alpha expression. Drug Metab Dispos 2006;34:1677-87. [ Links ]

137. Sugatani J, Osabe M, Wada T, Yamakawa K, Yamazaki Y, Takahashi T, et al. Comparison of enzymatically synthesized inulin, resistant maltodextrin and clofibrate effects on biomarkers of metabolic disease in rats fed a high-fat and high-sucrose (cafeteria) diet. Eur J Nutr 2008;47(4):192-200. [ Links ]

138. Chen HL, Wang CH, Kuo YW, Tsai CH. Antioxidative and hepatoprotective effects of fructo-oligosaccharide in D-galactose-treated Balb/cJ mice. Br J Nutr 2011;105:805-9. [ Links ]

139. Fan JG, Xu ZJ, Wang GL. Effect of lactulose on establishment of a rat non-alcoholic steatohepatitis model. World J Gastroenterol 2005;11(32):5053-6. [ Links ]

140. Parnell JA, Reimer RA. Effect of prebiotic fibre supplementation on hepatic gene expression and serum lipids: a dose-response study in JCR: LA-cp rats. Br J Nutr 2010;103:1577-84. [ Links ]

141. Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci USA 2005;102:11070-5. [ Links ]

142. Schwiertz A, Taras D, Schafer K, Beijer NA. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2009;18:190-5. [ Links ]

143. Collado MC, Isolauri E, Laitinen K, Salminen S. Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am J Clin Nutr 2008;88:894-9. [ Links ]

144. Santacruz A, Marcos A, Wärnberg J, MartíA, Martín-Matillas M, Campoy C, et al. Interplay between weight loss and gut microbiota composition in overweight adolescents. Obesity 2009;17(10):1906-15. [ Links ]

145. Nadal I, Santacruz A, Marcos A, Warnberg JM, Garagorri JM, Moreno LA, et al. Shifts in clostridia, bacteroides and immunoglobulin-coating fecal bacteria associated with weight loss in obese adolescents. Int J Obesity 2009;33:758-67. [ Links ]

146. Million M, Lagier JC, Yahav D, Paul M. Gut bacterial microbiota and obesity. Clinical Microbiology and Infection 2013;19(4);305-13. [ Links ]

147. Pedersen R, Andersen AD, Mølbak L, Stagsted J, Boye M. Changes in the gut microbiota of cloned and non-cloned control pigs during development of obesity: gut microbiota during development of obesity in cloned pigs. BMC Microbiology 2013;13:30. [ Links ]

148. Ley RE. Obesity and the human microbiome. Curr Opin Gastroenterol 2010;26:5-11. [ Links ]

149. Guo Y, Huang ZP, Liu CQ, Qi L, Sheng Y, Zou DJ. Modulation of the gut microbiome: a systematic review of the effect of bariatric surgery. Eur J Endocrinol 2018;178:43-56. [ Links ]

150. Karlsson CL, Onnerfält J, Xu J, Molin S, AhrnéK. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity (Silver Spring) 2012;20:2257-61. [ Links ]

151. Chakraborti CK. New-found link between microbiota and obesity. World J Gastrointest Pathophysiol 2015;6:110-9. [ Links ]

Author´s contribution: JML thanks MICINN for his ‘Ramon y Cajal’ contract (RYC-2015-18083). This work was supported by grants RYC-2015-18083 from the Spanish Ministry of Science and Innovation (MICINN, Spain).

Fernández-Musoles R, García-Tejedor A, Laparra JM. Immunonutritional contribution of gut microbiota to fatty liver disease. Nutr Hosp 2020;37(1):193-20.

Received: July 05, 2019; Accepted: October 13, 2019

Correspondence: José Moisés Laparra Llopis. Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA Alimentación). Grupo de Inmunonutrición Molecular. Ctra. de Cantoblanco, 8. 28049 Madrid, Spain e-mail: moises.laparra@imdea.org

Conflict of interest:

The authors declare no conflict of interest.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License