Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Eur J Med Res ; 29(1): 485, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39367507

RESUMEN

Metabolic dysfunction-associated steatohepatitis (MASH) and progression to hepatocellular carcinoma (HCC) exhibits distinct molecular and immune characteristics. These traits are influenced by multiple factors, including the gut microbiome, which interacts with the liver through the "gut-liver axis". This bidirectional relationship between the gut and its microbiota and the liver plays a key role in driving various liver diseases, with microbial metabolites and immune responses being central to these processes. Our review consolidates the latest research on how gut microbiota contributes to MASH development and its progression to HCC, emphasizing new diagnostic and therapeutic possibilities. We performed a comprehensive literature review across PubMed/MedLine, Scopus, and Web of Science from January 2000 to August 2024, focusing on both preclinical and clinical studies that investigate the gut microbiota's roles in MASH and HCC. This includes research on pathogenesis, as well as diagnostic and therapeutic advancements related to the gut microbiota. This evidence emphasizes the critical role of the gut microbiome in the pathogenesis of MASH and HCC, highlighting the need for further clinical studies and trials. This is to refine diagnostic techniques and develop targeted therapies that exploit the microbiome's capabilities, aiming to enhance patient care in liver diseases.


Asunto(s)
Carcinoma Hepatocelular , Microbioma Gastrointestinal , Neoplasias Hepáticas , Humanos , Microbioma Gastrointestinal/fisiología , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/microbiología , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/microbiología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/metabolismo , Hígado Graso/diagnóstico , Hígado Graso/terapia , Hígado Graso/microbiología , Hígado Graso/metabolismo , Hígado Graso/etiología
2.
Gut Microbes ; 16(1): 2399260, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239875

RESUMEN

The gut microbiota drives progression to liver fibrosis, the main determinant of mortality in metabolic dysfunction-associated steatohepatitis (MASH). In this study, we aimed to identify bacterial species associated with protection against liver fibrosis in a high-risk population, and test their potential to protect against liver fibrosis in vivo. Based on stool shotgun metagenomic sequencing of 340 subjects from a population cohort disproportionally affected by MASH, we identified bacterial species from the Bacteroidales and Clostridiales orders associated with reduced risk of liver fibrosis. A bacterial consortium was subsequently tested in a mouse model of MASH, which demonstrated protective effects against liver fibrosis. Six of the eight inoculated bacteria were detected in mouse stool and liver. Intrahepatic presence of bacteria was further confirmed by bacterial culture of mouse liver tissue. Changes in liver histological parameters, gut functional profiles, and amino acid profiles were additionally assessed. Comparison between fibrosis-associated human metagenome and bacteria-induced metagenome changes in mice identified microbial functions likely to mediate the protective effect against liver fibrosis. Amino acid profiling confirmed an increase in cysteine synthase activity, associated with reduced fibrosis. Other microbiota-induced changes in amino acids associated with reduced fibrosis included increased gut asparaginase activity and decreased hepatic tryptophan-to-kynurenine conversion. This human-to-mouse study identified bacterial species and their effects on amino acid metabolism as innovative strategies to protect against liver fibrosis in MASH.


Asunto(s)
Aminoácidos , Bacterias , Microbioma Gastrointestinal , Cirrosis Hepática , Hígado , Animales , Humanos , Cirrosis Hepática/microbiología , Cirrosis Hepática/metabolismo , Ratones , Aminoácidos/metabolismo , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Masculino , Hígado/metabolismo , Hígado/patología , Hígado/microbiología , Femenino , Heces/microbiología , Ratones Endogámicos C57BL , Persona de Mediana Edad , Hígado Graso/metabolismo , Hígado Graso/microbiología , Modelos Animales de Enfermedad , Metagenoma , Adulto
3.
FASEB J ; 38(13): e23766, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38967214

RESUMEN

Dysbiosis of gut microbiota may account for pathobiology in simple fatty liver (SFL), metabolic dysfunction-associated steatohepatitis (MASH), fibrotic progression, and transformation to MASH-associated hepatocellular carcinoma (MASH-HCC). The aim of the present study is to investigate gut dysbiosis in this progression. Fecal microbial rRNA-16S sequencing, absolute quantification, histopathologic, and biochemical tests were performed in mice fed high fat/calorie diet plus high fructose and glucose in drinking water (HFCD-HF/G) or control diet (CD) for 2, 16 weeks, or 14 months. Histopathologic examination verified an early stage of SFL, MASH, fibrotic, or MASH-HCC progression with disturbance of lipid metabolism, liver injury, and impaired gut mucosal barrier as indicated by loss of occludin in ileum mucosa. Gut dysbiosis occurred as early as 2 weeks with reduced α diversity, expansion of Kineothrix, Lactococcus, Akkermansia; and shrinkage in Bifidobacterium, Lactobacillus, etc., at a genus level. Dysbiosis was found as early as MAHS initiation, and was much more profound through the MASH-fibrotic and oncogenic progression. Moreover, the expansion of specific species, such as Lactobacillus johnsonii and Kineothrix alysoides, was confirmed by an optimized method for absolute quantification. Dynamic alterations of gut microbiota were characterized in three stages of early SFL, MASH, and its HCC transformation. The findings suggest that the extent of dysbiosis was accompanied with MASH progression and its transformation to HCC, and the shrinking or emerging of specific microbial species may account at least in part for pathologic, metabolic, and immunologic alterations in fibrogenic progression and malignant transition in the liver.


Asunto(s)
Carcinoma Hepatocelular , Disbiosis , Microbioma Gastrointestinal , Neoplasias Hepáticas , Ratones Endogámicos C57BL , Animales , Ratones , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/microbiología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/microbiología , Neoplasias Hepáticas/etiología , Disbiosis/microbiología , Masculino , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/microbiología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología
4.
Sci Rep ; 14(1): 16122, 2024 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997279

RESUMEN

Alcoholic-associated liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD) show a high prevalence rate worldwide. As gut microbiota represents current state of ALD and MASLD via gut-liver axis, typical characteristics of gut microbiota can be used as a potential diagnostic marker in ALD and MASLD. Machine learning (ML) algorithms improve diagnostic performance in various diseases. Using gut microbiota-based ML algorithms, we evaluated the diagnostic index for ALD and MASLD. Fecal 16S rRNA sequencing data of 263 ALD (control, elevated liver enzyme [ELE], cirrhosis, and hepatocellular carcinoma [HCC]) and 201 MASLD (control and ELE) subjects were collected. For external validation, 126 ALD and 84 MASLD subjects were recruited. Four supervised ML algorithms (support vector machine, random forest, multilevel perceptron, and convolutional neural network) were used for classification with 20, 40, 60, and 80 features, in which three nonsupervised ML algorithms (independent component analysis, principal component analysis, linear discriminant analysis, and random projection) were used for feature reduction. A total of 52 combinations of ML algorithms for each pair of subgroups were performed with 60 hyperparameter variations and Stratified ShuffleSplit tenfold cross validation. The ML models of the convolutional neural network combined with principal component analysis achieved areas under the receiver operating characteristic curve (AUCs) > 0.90. In ALD, the diagnostic AUC values of the ML strategy (vs. control) were 0.94, 0.97, and 0.96 for ELE, cirrhosis, and liver cancer, respectively. The AUC value (vs. control) for MASLD (ELE) was 0.93. In the external validation, the AUC values of ALD and MASLD (vs control) were > 0.90 and 0.88, respectively. The gut microbiota-based ML strategy can be used for the diagnosis of ALD and MASLD.ClinicalTrials.gov NCT04339725.


Asunto(s)
Microbioma Gastrointestinal , Aprendizaje Automático , Humanos , Masculino , Femenino , Persona de Mediana Edad , Adulto , Algoritmos , Hepatopatías Alcohólicas/microbiología , Hepatopatías Alcohólicas/diagnóstico , Hepatopatías Alcohólicas/metabolismo , ARN Ribosómico 16S/genética , Anciano , Curva ROC , Heces/microbiología , Hígado Graso/microbiología , Hígado Graso/diagnóstico , Hígado Graso/metabolismo
5.
Hepatol Int ; 18(Suppl 2): 861-872, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38995341

RESUMEN

In 2020, a revised definition of fatty liver disease associated with metabolic dysfunction (MAFLD) was proposed to replace non-alcoholic fatty liver (NAFLD). Liver steatosis and at least one of the three metabolic risk factors, including type 2 diabetes, obesity, or signs of metabolic dysregulation, are used to diagnose MAFLD. MAFLD, similarly to NAFLD, is characterized by a spectrum of disease ranging from simple steatosis to advanced metabolic steatohepatitis with or without fibrosis, and may progress to cirrhosis and liver cancer, including increased risk of other critical extrahepatic diseases. Even though the pathophysiology of MAFLD and potential therapeutic targets have been explored in great detail, there is yet no Food and Drug Administration approved treatment. Recently, gut microbiome-derived products (e.g., endotoxins and metabolites) involved in intestinal barrier disruption, systemic inflammation, and modification of intrahepatic immunity have been associated with MAFLD development and progression. Therefore, different strategies could be adopted to modify the gut microbiome to improve outcomes in early and progressive MAFLD. Here, we provide an overview of mechanisms that may link the gut microbiome and immune response during the onset of liver steatosis and progression to steatohepatitis and fibrosis in patients with MAFLD. Finally, gut microbiota-based approaches are discussed as potential personalized treatments against MAFLD.


Asunto(s)
Microbioma Gastrointestinal , Microbioma Gastrointestinal/inmunología , Humanos , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Hígado Graso/inmunología , Hígado Graso/microbiología , Progresión de la Enfermedad , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/microbiología
6.
Gigascience ; 132024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38837944

RESUMEN

Hepatic steatosis is the initial manifestation of abnormal liver functions and often leads to liver diseases such as nonalcoholic fatty liver disease in humans and fatty liver syndrome in animals. In this study, we conducted a comprehensive analysis of a large chicken population consisting of 705 adult hens by combining host genome resequencing; liver transcriptome, proteome, and metabolome analysis; and microbial 16S ribosomal RNA gene sequencing of each gut segment. The results showed the heritability (h2 = 0.25) and duodenal microbiability (m2 = 0.26) of hepatic steatosis were relatively high, indicating a large effect of host genetics and duodenal microbiota on chicken hepatic steatosis. Individuals with hepatic steatosis had low microbiota diversity and a decreased genetic potential to process triglyceride output from hepatocytes, fatty acid ß-oxidation activity, and resistance to fatty acid peroxidation. Furthermore, we revealed a molecular network linking host genomic variants (GGA6: 5.59-5.69 Mb), hepatic gene/protein expression (PEMT, phosphatidyl-ethanolamine N-methyltransferase), metabolite abundances (folate, S-adenosylmethionine, homocysteine, phosphatidyl-ethanolamine, and phosphatidylcholine), and duodenal microbes (genus Lactobacillus) to hepatic steatosis, which could provide new insights into the regulatory mechanism of fatty liver development.


Asunto(s)
Pollos , Hígado Graso , Microbioma Gastrointestinal , Animales , Pollos/microbiología , Microbioma Gastrointestinal/genética , Hígado Graso/genética , Hígado Graso/microbiología , Hígado Graso/veterinaria , Hígado Graso/metabolismo , Hígado/metabolismo , Hígado/microbiología , Transcriptoma , Genoma , Metaboloma , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/genética
7.
Inflamm Res ; 73(7): 1081-1098, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38619583

RESUMEN

BACKGROUND: Tsumura-Suzuki non-obese (TSNO) mice exhibit a severe form of metabolic dysfunction-associated steatohepatitis (MASH) with advanced liver fibrosis upon feeding a high-fat/cholesterol/cholate-based (iHFC) diet. Another ddY strain, Tsumura-Suzuki diabetes obese (TSOD) mice, are impaired in the progression of iHFC diet-induced MASH. AIM: To elucidate the underlying mechanisms contributing to the differences in MASH progression between TSNO and TSOD mice. METHODS: We analyzed differences in the immune system, gut microbiota, and bile acid metabolism in TSNO and TSOD mice fed with a normal diet (ND) or an iHFC diet. RESULTS: TSOD mice had more anti-inflammatory macrophages in the liver than TSNO mice under ND feeding, and were impaired in the iHFC diet-induced accumulation of fibrosis-associated macrophages and formation of histological hepatic crown-like structures in the liver. The gut microbiota of TSOD mice also exhibited a distinct community composition with lower diversity and higher abundance of Akkermansia muciniphila compared with that in TSNO mice. Finally, TSOD mice had lower levels of bile acids linked to intestinal barrier disruption under iHFC feeding. CONCLUSIONS: The dynamics of liver macrophage subsets, and the compositions of the gut microbiota and bile acids at steady state and post-onset of MASH, had major impacts on MASH development.


Asunto(s)
Ácidos y Sales Biliares , Dieta Alta en Grasa , Microbioma Gastrointestinal , Hígado , Macrófagos , Animales , Ácidos y Sales Biliares/metabolismo , Hígado/patología , Hígado/metabolismo , Dieta Alta en Grasa/efectos adversos , Macrófagos/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/microbiología , Akkermansia , Progresión de la Enfermedad , Colesterol en la Dieta/efectos adversos
8.
Int J Mol Sci ; 25(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38673972

RESUMEN

Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex disorder whose prevalence is rapidly growing in South America. The disturbances in the microbiota-gut-liver axis impact the liver damaging processes toward fibrosis. Gut microbiota status is shaped by dietary and lifestyle factors, depending on geographic location. We aimed to identify microbial signatures in a group of Chilean MASLD patients. Forty subjects were recruited, including healthy controls (HCs), overweight/obese subjects (Ow/Ob), patients with MASLD without fibrosis (MASLD/F-), and MASLD with fibrosis (MASLD/F+). Both MASLD and fibrosis were detected through elastography and/or biopsy, and fecal microbiota were analyzed through deep sequencing. Despite no differences in α- and ß-diversity among all groups, a higher abundance of Bilophila and a lower presence of Defluviitaleaceae, Lachnospiraceae ND3007, and Coprobacter was found in MASLD/F- and MASLD/F+, compared to HC. Ruminococcaceae UCG-013 and Sellimonas were more abundant in MASLD/F+ than in Ow/Ob; both significantly differed between MASLD/F- and MASLD/F+, compared to HC. Significant positive correlations were observed between liver stiffness and Bifidobacterium, Prevotella, Sarcina, and Acidaminococcus abundance. Our results show that MASLD is associated with changes in bacterial taxa that are known to be involved in bile acid metabolism and SCFA production, with some of them being more specifically linked to fibrosis.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Masculino , Femenino , Persona de Mediana Edad , Adulto , Cirrosis Hepática/microbiología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Heces/microbiología , Hígado/metabolismo , Hígado/patología , Hígado Graso/microbiología , Hígado Graso/metabolismo , Hígado Graso/patología , Progresión de la Enfermedad , Obesidad/microbiología , Obesidad/complicaciones , Obesidad/metabolismo , Chile , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Bacterias/metabolismo , Anciano
9.
Chin Med Sci J ; 38(4): 286-296, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38097345

RESUMEN

Metabolic associated fatty liver disease (MAFLD) has become a prevalent chronic liver disease worldwide because of lifestyle and dietary changes. Gut microbiota and its metabolites have been shown to play a critical role in the pathogenesis of MAFLD. Understanding of the function of gut microbiota and its metabolites in MAFLD may help to elucidate pathological mechanisms, identify diagnostic markers, and develop drugs or probiotics for the treatment of MAFLD. Here we review the pathogenesis of MAFLD by gut microbiota and its metabolites and discuss the feasibility of treating MAFLD from the perspective of gut microbes.


Asunto(s)
Hígado Graso , Microbioma Gastrointestinal , Hígado Graso/microbiología , Humanos
10.
mBio ; 12(4): e0115521, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34225483

RESUMEN

Nonalcoholic fatty liver disease (NAFLD), characterized by excessive fat deposition in the liver unrelated to alcohol consumption, is highly prevalent worldwide. However, effective therapeutic agents approved for NAFLD treatment are lacking. An ileal bile acid transporter inhibitor (IBATi), which represents a new mode of treatment of chronic idiopathic constipation, leads to increased delivery of bile acids to the colon. We investigated the effect of IBATi against NAFLD through modification of the gut microbiota in mice. IBATi treatment significantly suppressed body weight gain, liver dysfunction, and serum low-density lipoprotein levels and significantly decreased NAFLD activity scores in high-fat diet (HFD) mice. Treatment with IBATi ameliorated the decreased hepatic cholesterol 7-a-monooxygenase (Cyp7a1) and increased ileal fibroblast growth factor 15 (Fgf15) mRNA expression in HFD mice. Further, IBATi treatment changed the α-diversity in the gut microbiota reduced by HFD, which was analyzed in feces using 16S rRNA sequencing. To establish the mechanism underlying improvement in NAFLD induced by IBATi, we recolonized antibiotic solution-treated mice by fecal microbiome transplantation (FMT) using stool from HFD or HFD plus IBATi mice. This is the first report that fecally transplanted gut microbiota from HFD plus IBATi mice prevented hepatic steatosis caused by HFD. In conclusion, IBATi improved hepatic steatosis by ameliorating gut microbiota dysbiosis in NAFLD model mice, suggesting a potential therapeutic agent for NAFLD treatment. IMPORTANCE NAFLD is an increasingly recognized condition that may progress to liver cirrhosis and hepatocellular carcinoma, and community surveys have assessed that the prevalence is 14 to 32% worldwide. The first line of treatment for NAFLD is lifestyle modification to achieve weight reduction, particularly through diet and exercise. However, weight reduction is difficult to achieve and maintain, and pharmacological agents approved for the treatment of NAFLD are lacking. This study investigated the influence of the gut microbiota and the effect of an IBATi on NAFLD using a murine model. Treatment with IBATi significantly improved NAFLD in HFD mice. Further, fecal microbiome transplantation using stool from HFD plus IBATi mice prevented hepatic steatosis caused by HFD. Our study makes a significant contribution to the literature because the study findings suggest a potential treatment strategy for NAFLD patients by ameliorating gut microbiota dysbiosis.


Asunto(s)
Disbiosis/prevención & control , Hígado Graso/prevención & control , Microbioma Gastrointestinal/fisiología , Enfermedad del Hígado Graso no Alcohólico/microbiología , Animales , Ácidos y Sales Biliares/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Disbiosis/microbiología , Hígado Graso/microbiología , Microbioma Gastrointestinal/genética , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética
11.
Medicine (Baltimore) ; 99(50): e22867, 2020 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-33327227

RESUMEN

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) has emerged as a major health problem worldwide; according to statistics, 10% to 25% of patients with NAFLD can progress to nonalcoholic steatohepatitis (NASH). A link between the composition and metabolites of intestinal microbiota and the development of NAFLD is becoming clearer. It is believed that microbiota factors are driving forces of hepatic steatosis and inflammation. The formulated food that contains prebiotics and dietary fiber may improve NAFLD by altering the intestinal flora and its metabolites. METHODS: The study plan to recruit adult patients (18-75 years, n = 120) with NAFLD, range of alanine aminotransferase is 1.5 to 5 times upper limit of normal (ULN) or liver biopsy is confirmed as NASH. Participants will be randomly allocated into 2 groups: formulated food (n = 80) and a placebo group (n = 40) for 24 weeks. Both groups will receive lifestyle and nutritional advice. The primary endpoint is a decrease in MRS-PDFF by more than 30% from baseline at 24 weeks. The secondary endpoints include the change of anthropometric, liver function, glycolipid metabolism, and systemic inflammation at 4, 12, and 24 weeks. In addition, we consider the changes in intestinal microbiota as an exploration to assess the abundance and diversity at 24 weeks. Weeks 24 to 36 are the follow-up period of drug withdrawal. DISCUSSION: This clinical trial will provide evidence of efficacy and safety of formulated food as a potential new therapeutic agent for NAFLD patients. TRIAL REGISTRATION: The trial is registered in the China Clinical Trial Center (ChiCTR1800016178).


Asunto(s)
Alimentos Formulados/efectos adversos , Microbioma Gastrointestinal/fisiología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Adulto , Anciano , Alanina Transaminasa/análisis , Bacterias/metabolismo , Estudios de Casos y Controles , China/epidemiología , Hígado Graso/dietoterapia , Hígado Graso/microbiología , Femenino , Estudios de Seguimiento , Alimentos Formulados/microbiología , Alimentos Formulados/provisión & distribución , Humanos , Inflamación/dietoterapia , Inflamación/microbiología , Metabolismo de los Lípidos/fisiología , Hígado/patología , Masculino , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/dietoterapia , Enfermedad del Hígado Graso no Alcohólico/patología , Seguridad , Resultado del Tratamiento
12.
Food Funct ; 10(9): 5566-5573, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31429848

RESUMEN

The interactions among the liver, intestine and immune system play an important role in alcoholic liver injury. In this study, C57BL/6N mice with alcoholic injury were treated with unfermented and Lactobacillus fermentum KP-3-fermented ginseng. The indicators of hepatic steatosis, inflammation and injury were evaluated. The number of beneficial and harmful bacteria in the mice ileum and colon was counted by a traditional method; moreover, the diversity analysis of the cecum flora was performed. The alcohol exposure increased the levels of ALT, AST, TNF-α and IL-6 inflammatory factors and liver steatosis. In addition, the alcohol-fed miceexhibited a lower number of Lactobacilli and Bifidobacteria in the ileum and colon; the cecum flora diversity in the mice showed that alcohol obviously enhanced the abundance of the unclassified S24-7 of the Bacteroidetes phylum and the Proteobacteria genus of the Sutterella phylum and reduced the abundance of short-chain fatty acid-producing bacteria such as Akkermansia in the Verrucomicrobia phylum and those belonging to the Allobaculum genus, the Ruminococcus genus, and the Adlercreutzia genus in the Actinobacteria phylum. All these changes were improved by fermented ginseng. Conclusively, fermented ginseng could alleviate the alcoholic liver injury and disorder of the intestine by adjusting the intestinal flora.


Asunto(s)
Hígado Graso/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Limosilactobacillus fermentum/metabolismo , Panax/química , Extractos Vegetales/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Colon/microbiología , Etanol/efectos adversos , Ácidos Grasos Volátiles/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/microbiología , Fermentación , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Panax/microbiología , Extractos Vegetales/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
Gut ; 68(3): 453-464, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30064988

RESUMEN

OBJECTIVE: The consumption of fruits is strongly associated with better health and higher bacterial diversity in the gut microbiota (GM). Camu camu (Myrciaria dubia) is an Amazonian fruit with a unique phytochemical profile, strong antioxidant potential and purported anti-inflammatory potential. DESIGN: By using metabolic tests coupled with 16S rRNA gene-based taxonomic profiling and faecal microbial transplantation (FMT), we have assessed the effect of a crude extract of camu camu (CC) on obesity and associated immunometabolic disorders in high fat/high sucrose (HFHS)-fed mice. RESULTS: Treatment of HFHS-fed mice with CC prevented weight gain, lowered fat accumulation and blunted metabolic inflammation and endotoxaemia. CC-treated mice displayed improved glucose tolerance and insulin sensitivity and were also fully protected against hepatic steatosis. These effects were linked to increased energy expenditure and upregulation of uncoupling protein 1 mRNA expression in the brown adipose tissue (BAT) of CC-treated mice, which strongly correlated with the mRNA expression of the membrane bile acid (BA) receptor TGR5. Moreover, CC-treated mice showed altered plasma BA pool size and composition and drastic changes in the GM (eg, bloom of Akkermansia muciniphila and a strong reduction of Lactobacillus). Germ-free (GF) mice reconstituted with the GM of CC-treated mice gained less weight and displayed higher energy expenditure than GF-mice colonised with the FM of HFHS controls. CONCLUSION: Our results show that CC prevents visceral and liver fat deposition through BAT activation and increased energy expenditure, a mechanism that is dependent on the GM and linked to major changes in the BA pool size and composition.


Asunto(s)
Metabolismo Energético/fisiología , Frutas/química , Microbioma Gastrointestinal/efectos de los fármacos , Obesidad/prevención & control , Animales , Ácido Ascórbico/uso terapéutico , Glucemia/metabolismo , Endotoxemia/prevención & control , Hígado Graso/microbiología , Hígado Graso/fisiopatología , Hígado Graso/prevención & control , Trasplante de Microbiota Fecal , Homeostasis/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/microbiología , Obesidad/fisiopatología , Paniculitis/prevención & control , Extractos Vegetales/química , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico
14.
Nutrients ; 10(10)2018 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297615

RESUMEN

A variety of dietary natural products have shown hepatoprotective effects. Increasing evidence has also demonstrated that gut microorganisms play an important role in the hepatoprotection contributed by natural products. Gut dysbiosis could increase permeability of the gut barrier, resulting in translocated bacteria and leaked gut-derived products, which can reach the liver through the portal vein and might lead to increased oxidative stress and inflammation, thereby threatening liver health. Targeting gut microbiota modulation represents a promising strategy for hepatoprotection. Many natural products could protect the liver from various injuries or mitigate hepatic disorders by reverting gut dysbiosis, improving intestinal permeability, altering the primary bile acid, and inhibiting hepatic fatty acid accumulation. The mechanisms underlying their beneficial effects also include reducing oxidative stress, suppressing inflammation, attenuating fibrosis, and decreasing apoptosis. This review discusses the hepatoprotective effects of dietary natural products via modulating the gut microbiota, mainly focusing on the mechanisms of action.


Asunto(s)
Productos Biológicos/farmacología , Dieta , Hígado Graso/microbiología , Microbioma Gastrointestinal , Neoplasias Hepáticas/microbiología , Hígado , Probióticos , Apoptosis/efectos de los fármacos , Productos Biológicos/uso terapéutico , Disbiosis/complicaciones , Disbiosis/tratamiento farmacológico , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/prevención & control , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/microbiología , Hígado Graso Alcohólico/patología , Hígado Graso Alcohólico/prevención & control , Fibrosis/tratamiento farmacológico , Humanos , Inflamación/tratamiento farmacológico , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/prevención & control , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/microbiología , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico
15.
J Cell Physiol ; 234(1): 122-133, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30191979

RESUMEN

Previous studies have shown that gut-derived bacterial endotoxins contribute in the progression of simple steatosis to steatohepatitis, although the mechanism(s) remains inaccurate to date. As hepatic stellate cells (HSC) play a pivotal role in the accumulation of excessive extracellular matrix (ECM), leading to collagen deposition, fibrosis, and perpetuation of inflammatory response, an in vitro model was developed to investigate the crosstalk between HSC and hepatocytes (human hepatoma cell) pretreated with palmitate. Bacterial lipopolysaccharide (LPS) stimulated HSC with phosphorylation of the p38 mitogen-activated protein kinase/NF-κB pathway, while several important pro-inflammatory cytokines were upregulated in the presence of hepatocyte-HSC. Concurrently, fibrosis-related genes were regulated by palmitate and the inflammatory effect of endotoxin where cells were more exposed or sensitive to reactive oxygen species (ROS). This interaction was accompanied by increased expression of the mitochondrial master regulator, proliferator-activated receptor gamma coactivator alpha, and a cytoprotective effect of the agent N-acetylcysteine suppressing ROS production, transforming growth factor-ß1, and tissue inhibitor of metalloproteinase-1. In summary, our results demonstrate that pro-inflammatory mediators LPS-induced promote ECM rearrangement in hepatic cells transcriptionally committed to the regulation of genes encoding enzymes for fatty acid metabolism in light of differences that might require an alternative therapeutic approach targeting ROS regulation.


Asunto(s)
Comunicación Celular/genética , Hígado Graso/genética , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Comunicación Celular/efectos de los fármacos , Citocinas/genética , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Hígado Graso/microbiología , Hígado Graso/patología , Fibrosis/genética , Fibrosis/patología , Células Estrelladas Hepáticas/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Lipopolisacáridos/toxicidad , Palmitatos/farmacología , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/genética , Factor de Crecimiento Transformador beta1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
16.
Infection ; 46(6): 793-799, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30066228

RESUMEN

PURPOSE: Endotoxin is a component of the outer membrane of gram-negative bacteria that live in the intestine. Endotoxinemia is reported in non-alcoholic fatty liver disease and in cirrhotic patients, causing various biological and clinical effects in the host. It is not known whether endotoxinemia occurs in chronic hepatitis C patients (CHC), therefore we evaluated the occurrence of endotoxinemia and its effect on inflammation, liver damage, insulin resistance (IR) and atherosclerosis. METHODS: Consecutive CHC patients assessed by liver biopsy were enrolled. Endotoxinemia was evaluated by LAL test. IR was estimated by HOMA-IR. Serum TNF-α, IL-8, adiponectin and MCP-1 were measured with ELISA tests. Oxidative stress was estimated by circulating IgG against malondialdehyde adducts with human serum albumin (MDA-HAS). Carotid atherosclerosis was assessed by ultrasonography. RESULTS: Endotoxinemia was found in 60% of the 126 patients enrolled. A serum level-dependent association between endotoxinemia, steatosis (p < 0.001) and HOMA-IR (p < 0.006) was observed. Patients with endotoxinemia showed significant increase in TNF-α and IL8 levels. TNF-α correlated with steatosis (p <  0.001) and HOMA-IR (p < 0.03), whereas IL8 correlated with steatosis (p =  <0.001), TNF-α (p < 0.04) and atherosclerosis (p < 0.01). The highest levels of endotoxinemia were associated with oxidative stress and a higher prevalence of carotid atherosclerosis. Multivariate logistic regression analysis showed that the independent factors associated with endotoxinemia were hepatic steatosis, HOMA-IR, IL8 and MDA-HAS. CONCLUSIONS: Endotoxinemia occurs with high frequency in CHC patients and contributes to the development of hepatic steatosis, IR and atherosclerosis through increased pro-inflammatory cytokines and oxidative stress. Anti-endotoxin treatment could be of clinical relevance.


Asunto(s)
Aterosclerosis/microbiología , Endotoxemia/epidemiología , Fibrosis/microbiología , Hepatitis C Crónica/complicaciones , Inflamación/microbiología , Resistencia a la Insulina , Estrés Oxidativo , Adolescente , Adulto , Anciano , Quimiocinas/metabolismo , Citocinas/metabolismo , Endotoxemia/complicaciones , Endotoxemia/microbiología , Hígado Graso/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Adulto Joven
17.
Nutr Diabetes ; 8(1): 15, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29549311

RESUMEN

BACKGROUND: Non-digestible carbohydrates present in cereals such as fructans and arabinoxylans represent promising prebiotic nutrients to prevent the development of obesity and related metabolic disorders. OBJECTIVE AND DESIGN: The aim of this study was to determine the corrective effects of wheat bran-derived arabinoxylan oligosaccharides in obese mice fed a western diet (WD). WD was given for 4 weeks before wheat bran extract (WBE) supplementation (5%) for an additional 4 weeks, whereas a control group received the standard diet. RESULTS: Bifidogenic effect of WBE was evidenced by an induction of both Bifidobacterium animalis and Bifidobacterium pseudolongum in the caecal content. WBE supplementation normalised WD-induced fat-mass expansion, steatosis, hypercholesterolemia, hyperleptinemia, hyperglycemia and hyperinsulinemia reaching the values of control mice. The reduced glucose-dependent insulinotropic polypeptide (GIP) release observed in WD + WBE mice may be a protective mechanism in terms of reducing adipose tissue storage, hepatic steatosis and glucose homoeostasis. CONCLUSION: We found that WBE completely abolished WD-induced metabolic disorders. Those results might be useful to take into account nutritional advices to treat obesity and related metabolic disorders such as type 2 diabetes, hypercholesterolaemia and fatty liver diseases when obesity was already established.


Asunto(s)
Bifidobacterium/efectos de los fármacos , Dieta Occidental/efectos adversos , Enfermedades Metabólicas/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Oligosacáridos/uso terapéutico , Triticum/química , Xilanos/uso terapéutico , Tejido Adiposo/metabolismo , Animales , Bifidobacterium/crecimiento & desarrollo , Glucemia/metabolismo , Ciego/microbiología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/microbiología , Fibras de la Dieta , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Hígado Graso/microbiología , Polipéptido Inhibidor Gástrico/sangre , Hipercolesterolemia/sangre , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/microbiología , Hiperglucemia/sangre , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/microbiología , Hiperinsulinismo/sangre , Hiperinsulinismo/tratamiento farmacológico , Hiperinsulinismo/microbiología , Leptina/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/microbiología , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/complicaciones , Obesidad/metabolismo , Obesidad/microbiología , Oligosacáridos/farmacología , Prebióticos , Xilanos/farmacología
18.
Am J Physiol Endocrinol Metab ; 314(4): E334-E352, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28874357

RESUMEN

Increasing evidence suggests that polyphenols have a significant potential in the prevention and treatment of risk factors associated with metabolic syndrome. The objective of this study was to assess the metabolic outcomes of two polyphenol-containing extracts from cinnamon bark (CBE) and grape pomace (GPE) on C57BL/6J mice fed a high-fat diet (HFD) for 8 wk. Both CBE and GPE were able to decrease fat mass gain and adipose tissue inflammation in mice fed a HFD without reducing food intake. This was associated with reduced liver steatosis and lower plasma nonesterified fatty acid levels. We also observed a beneficial effect on glucose homeostasis, as evidenced by an improved glucose tolerance and a lower insulin resistance index. These ameliorations of the overall metabolic profile were associated with a significant impact on the microbial composition, which was more profound for the GPE than for the CBE. At the genus level, Peptococcus were decreased in the CBE group. In the GPE-treated group, several key genera that have been previously found to be linked with HFD, metabolic effects, and gut barrier integrity were affected: we observed a decrease of Desulfovibrio, Lactococcus, whereas Allobaculum and Roseburia were increased. In addition, the expression of several antimicrobial peptides and tight junction proteins was increased in response to both CBE and GPE supplementation, indicating an improvement of the gut barrier function. Collectively, these data suggest that CBE and GPE can ameliorate the overall metabolic profile of mice on a high-fat diet, partly by acting on the gut microbiota.


Asunto(s)
Cinnamomum zeylanicum/química , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Enfermedades Metabólicas/prevención & control , Extractos Vegetales/farmacología , Vitis/química , Animales , Biomarcadores/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Experimental/prevención & control , Dieta Alta en Grasa/efectos adversos , Hígado Graso/metabolismo , Hígado Graso/microbiología , Hígado Graso/prevención & control , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/microbiología , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/microbiología , Obesidad/prevención & control , Permeabilidad , Extractos Vegetales/uso terapéutico
19.
Biochem Biophys Res Commun ; 493(2): 1075-1081, 2017 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-28923247

RESUMEN

Different edible oils such as lard and soybean oil have been reported to interact with the gut microbiota, affecting host lipid metabolism. However, whether bacteria derived from the environment influence host lipid metabolism remains unclear. This study aimed to clarify the roles of environmental bacteria in host lipid storage and distribution with various edible oils. Gnotobiotic C57BL/6JNarl mice were inoculated with Lysinibacillus xylanilyticus and Paenibacillus azoreducens and then fed either a normal diet (LabDiet 5010, control group) or a diet containing 60% lard (L-group) or soybean oil (S-group) for 18 months. Interestingly, the S-group accumulated massive amounts of white adipose tissue compared to the L- and control groups, while the L-group displayed more hepatic steatosis and fatty droplets than the other groups. The expression of fatty acid synthase (FAS), hydroxymethylglutaryl-coenzyme A reductase (HMGCR), sterol regulatory element-binding protein 2 (SREBP2), and peroxisome proliferator-activated receptor gamma (PPARγ) in the livers of the L-group were markedly elevated compared to the S-group. FAS and PPARγ protein levels were also markedly elevated. However, there were no differences in the expression of the pro-inflammatory cytokines interleukin-6 and tumor necrosis factor-α between the groups. Our results suggest that environmental bacteria may affect host hepatic inflammation and lipid distribution in the presence of high-fat diets, with different effects depending on the fat type consumed.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hígado Graso/metabolismo , Hígado Graso/microbiología , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/microbiología , Animales , Bacillaceae/fisiología , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/metabolismo , Hígado Graso/patología , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Paenibacillus/fisiología , Aceite de Soja/efectos adversos , Aceite de Soja/metabolismo
20.
Am J Pathol ; 187(8): 1800-1813, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28711154

RESUMEN

Patients who have liver cirrhosis and liver cancer also have reduced farnesoid X receptor (FXR). The current study analyzes the effect of diet through microbiota that affect hepatic inflammation in FXR knockout (KO) mice. Wild-type and FXR KO mice were on a control (CD) or Western diet (WD) for 10 months. In addition, both CD- and WD-fed FXR KO male mice, which had hepatic lymphocyte and neutrophil infiltration, were treated by vancomycin, polymyxin B, and Abx (ampicillin, neomycin, metronidazole, and vancomycin). Mice were subjected to morphological analysis as well as gut microbiota and bile acid profiling. Male WD-fed FXR KO mice had the most severe steatohepatitis. FXR KO also had reduced Firmicutes and increased Proteobacteria, which could be reversed by Abx. In addition, Abx eliminated hepatic neutrophils and lymphocytes in CD-fed, but not WD-fed, FXR KO mice. Proteobacteria and Bacteroidetes persisted in WD-fed FXR KO mice even after Abx treatment. Only polymyxin B could reduce hepatic lymphocytes in WD-fed FXR KO mice. The reduced hepatic inflammation by antibiotics was accompanied by decreased free and conjugated secondary bile acids as well as changes in gut microbiota. Our data revealed that Lactococcus, Lactobacillus, and Coprococcus protect the liver from inflammation.


Asunto(s)
Dieta Occidental/efectos adversos , Disbiosis/patología , Hígado Graso/patología , Inflamación/patología , Hígado/patología , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Antibacterianos/farmacología , Disbiosis/etiología , Disbiosis/metabolismo , Disbiosis/microbiología , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/microbiología , Inflamación/metabolismo , Inflamación/microbiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/microbiología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Linfocitos/patología , Masculino , Ratones , Ratones Noqueados , Microbiota , Infiltración Neutrófila , Receptores Citoplasmáticos y Nucleares/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA