Open Access
Open access
Small Structures, pages 2200312

Prebiotic‐Based Nanoamorphous Atorvastatin Attenuates Nonalcoholic Fatty Liver Disease by Retrieving Gut and Liver Health

Publication typeJournal Article
Publication date2023-01-18
Journal: Small Structures
SJR
CiteScore17.3
Impact factor13.9
ISSN26884062
General Medicine
Abstract

The pathogenesis of nonalcoholic fatty liver disease (NAFLD) is multifactorial and composite, with the disorder of lipid metabolism-induced lipotoxicity being one of the main risk factors. Atorvastatin (AT), the most widely prescribed lipid-lowering drug, has pleiotropic actions benefiting NAFLD treatment. However, low absorption rate in the gut and potential disruption of AT on gut flora hindered its further applications. Notably, gut dysbiosis is involved in and is thus a promising management strategy for NAFLD. In this study, we constructed a prebiotic-based AT nanoamorphous (PANA) to improve the efficacy of AT against NAFLD by retrieving liver and gut health. After oral administration, PANA showed superior drug accumulation in the liver tissue compared with pure AT. Moreover, PANA intervention effectively restored gut healthiness, indicated by reconstructed gut flora, and improved intestinal immunity, barrier integrity, and inflammation. Consequently, compared with AT, PANA treatment caused profound inhibition of weight gain and fat deposition, decreased plasma lipid levels, and alleviated hepatic steatosis and liver inflammation. The transcriptome analysis in the gut and liver tissues identified improved immunity and inflammation as potential mechanisms. This study suggests a promising strategy to treat NAFLD, assisted with nanotechnology in synergy with functional biomaterials.

Lin S., Wu F., Cao Z., Liu J.
2022-03-23 citations by CoLab: 9 PDF Abstract  
The intestinal barrier plays a vital role in maintaining the homeostasis of the gut and the organism. Various nanomedicines with advantages in interacting with the intestinal barrier have been developed for disease treatment. Previous studies indicate multiple interactions between these nanomaterials and the intestinal barrier, boosting their applications in promoting the efficiency of oral drug delivery. Recently, numerous elegant nanomedicines have been developed for preventing and treating both intestinal and extraintestinal diseases. Integration of multiple elements with diverse functions into a single type of nanomedicine is beneficial for comprehensive regulation of its interaction with the intestinal barrier and the corresponding treatment efficacy. In this review, the interactions between nanomaterials and the intestinal barrier are discussed following an introduction of the structure and function of the intestinal barrier, with an aim to better understanding the rational design of effective nanomedicines, particularly for oral delivery. Lastly, the applications of oral nanomedicines in treating intestinal barrier dysfunction and relevant diseases, such as inflammatory bowel disease, diabetes, arthritis, and cancer, are highlighted. It is anticipated that this review will stimulate more innovative thinking focused on the intestinal barrier as a critical target for drug delivery and disease treatment.
Su Y., Hu X., Tang H., Lu K., Li H., Liu S., Xing B., Ji R.
Nature Nanotechnology scimago Q1 wos Q1
2021-11-11 citations by CoLab: 130 Abstract  
Silicone-rubber baby teats used to bottle-feed infants are frequently disinfected by moist heating. However, infant exposure to small microplastics (<10 μm) potentially released from the heated teats by hydrothermal decomposition has not been studied, owing to the limitations of conventional spectroscopy in visualizing microplastic formation and in characterizing the particles at the submicrometre scale. Here both the surfaces of silicone teats subjected to steam disinfection and the wash waters of the steamed teats were analysed using optical-photothermal infrared microspectroscopy. This new technique revealed submicrometre-resolved steam etching on and chemical modification of the teat surface. Numerous flake- or oil-film-shaped micro(nano)plastics (MNPs) (in the size range of 0.6–332 μm) presented in the wash waters, including cyclic and branched polysiloxanes or imides, which were generated by the steam-induced degradation of the base polydimethylsiloxane elastomer and the polyamide resin additive. The results indicated that by the age of one year, a baby could ingest >0.66 million elastomer-derived micro-sized plastics (MPs) (roughly 81% in 1.5–10 μm). Global MP emission from teat disinfection may be as high as 5.2 × 1013 particles per year. Our findings highlight an entry route for surface-active silicone-rubber-derived MNPs into both the human body and the environment. The health and environmental risks of the particles are as yet unknown. Steam disinfection of silicone-rubber baby teats can lead to steam etching and chemical modification of the teat surface. This can release micro- and nanoplastics and result in ingestion. The results suggested that by the age of one year, a baby could ingest more than 600,000 microplastics.
Zhu R., Lang T., Yan W., Zhu X., Huang X., Yin Q., Li Y.
Advanced Science scimago Q1 wos Q1 Open Access
2021-03-09 citations by CoLab: 36 PDF Abstract  
Gut microbiota have close interactions with the host. It can affect cancer progression and the outcomes of cancer therapy, including chemotherapy, immunotherapy, and radiotherapy. Therefore, approaches toward the modulation of gut microbiota will enhance cancer prevention and treatment. Modern drug delivery systems (DDS) are emerging as rational and promising tools for microbiota intervention. These delivery systems have compensated for the obstacles associated with traditional treatments. In this review, the essential roles of gut microbiota in carcinogenesis, cancer progression, and various cancer therapies are first introduced. Next, advances in DDS that are aimed at enhancing the efficacy of cancer therapy by modulating or engineering gut microbiota are highlighted. Finally, the challenges and opportunities associated with the application of DDS targeting gut microbiota for cancer prevention and treatment are briefly discussed.
Sharpton S.R., Schnabl B., Knight R., Loomba R.
Cell Metabolism scimago Q1 wos Q1
2021-01-01 citations by CoLab: 125 Abstract  
Nonalcoholic fatty liver disease (NALFD) is now a leading cause of chronic liver disease worldwide, in part, as a consequence of rapidly rising levels of obesity and metabolic syndrome and is a major risk factor for cirrhosis, hepatocellular carcinoma, and liver-related mortality. From NAFLD stems a myriad of clinical challenges related to both diagnosis and management. A growing body of evidence suggests an intricate linkage between the gut microbiome and the pathogenesis of NAFLD. We highlight how our current knowledge of the gut-liver axis in NAFLD may be leveraged to develop gut microbiome-based personalized approaches for disease management, including its use as a non-invasive biomarker for diagnosis and staging, as a target for therapeutic modulation, and as a marker of drug response. We will also discuss current limitations of these microbiome-based approaches. Ultimately, a better understanding of microbiota-host interactions in NAFLD will inform the development of novel preventative strategies and precise therapeutic targets.
Wang R., Tang R., Li B., Ma X., Schnabl B., Tilg H.
2020-12-14 citations by CoLab: 266 Abstract  
The gut microbiota is a complex and plastic consortium of microorganisms that are intricately connected with human physiology. The liver is a central immunological organ that is particularly enriched in innate immune cells and constantly exposed to circulating nutrients and endotoxins derived from the gut microbiota. The delicate interaction between the gut and liver prevents accidental immune activation against otherwise harmless antigens. Work on the interplay between the gut microbiota and liver has assisted in understanding the pathophysiology of various liver diseases. Of immense importance is the step from high-throughput sequencing (correlation) to mechanistic studies (causality) and therapeutic intervention. Here, we review the gut microbiota, liver immunology, and the interaction between the gut and liver. In addition, the impairment in the gut–liver axis found in various liver diseases is reviewed here, with an emphasis on alcohol-associated liver disease (ALD), nonalcoholic fatty liver disease (NAFLD), and autoimmune liver disease (AILD). On the basis of growing evidence from these preclinical studies, we propose that the gut–liver axis paves the way for targeted therapeutic modalities for liver diseases.
Zhu X., Zhao L., Liu Z., Zhou Q., Zhu Y., Zhao Y., Yang X.
Nano Research scimago Q1 wos Q1
2020-12-03 citations by CoLab: 33 Abstract  
Titanium dioxide nanoparticles (TiO2-NPs) are commonly used as food additives, including some high-fat foods that are risk factors for obesity. However, little is known about the effects of chronic TiO2-NPs digestion in the population on high fat diet (HFD). Herein, we reported that TiO2-NPs exacerbated HFD-induced obesity by disruption of mucus layer and alterations of gut microbiota. Oral intake of TiO2-NPs significantly increased body weight, liver weight, and amount of adipose tissues, especially in HFD-fed mice. Mechanistic studies revealed TiO2-NPs induced colonic mucus layer disruption and obesity-related microbiota dysbiosis. The damage on mucus was demonstrated through down-regulation of Muc2 gene and the absorption of mucin protein by TiO2-NPs. Consequently, mucus layer damage combined microbiota dysbiosis escalated the low-grade systemic inflammation, which exacerbated HFD-induced obesity. In contrast, gut microbiota depletion eliminated these effects, indicating gut microbiota were necessary for TiO2-NPs-induced inflammation and obesity. All the results stated the alarming role of TiO2-NPs in the HFD-driven obesity and emphasized the reevaluating the health impacts of nanoparticles commonly used in daily life, particularly, in susceptible population.
Stachowska E., Portincasa P., Jamioł-Milc D., Maciejewska-Markiewicz D., Skonieczna-Żydecka K.
Nutrients scimago Q1 wos Q1 Open Access
2020-11-11 citations by CoLab: 42 PDF Abstract  
We aim to systematically review the efficacy of prebiotics in reducing anthropometric and biochemical parameters in individuals with non-alcoholic fatty liver disease (NAFLD). A systematic search using PubMed/MEDLINE, Embase, clinicaltrials.gov, Cinahl, and Web of Science of articles published up to 20 March 2020 was performed for randomized controlled trials enrolling >20 adult patients. Random-effect meta-analysis for metabolic outcomes in NAFLD patients was performed for anthropometric data in addition to liver enzyme, carbohydrate, and lipid parameters. We found six trials (comprising a total of 242 patients) with NAFLD, with subjects aged 38–52 years. The mean time of fiber administration varied between 10 and 12 weeks. The main fiber types were psyllium (seeds or powder), Ocimum basilicum (seeds), and high-performance inulin and oligofructose powder at doses of either 10 or 16 g per day. The control group received either maltodextrin (powder or capsules) or crushed wheat (powder). Patients on the diet with added fiber had improvements in body mass index (BMI) (standardized mean difference (SMD) = −0.494, 95% confidence interval (CI): −0.864 to −0.125, p = 0.009); alanine aminotransferase (ALT) (SMD = −0.667, 95% CI: −1.046 to −0.288, p = 0.001); aspartate aminotransferase (AST) (SMD = −0.466, 95% CI: −0.840 to −0.091, p = 0.015); fasting insulin (SMD = −0.705, 95% CI: −1.115 to −0.295, p = 0.001); and homeostasis model assessment for insulin resistance (HOMA-IR) (SMD = −0.619, 95% CI: −1.026 to −0.211, p = 0.003). Hence, the results show that fiber supplements result in favorable changes as reflected in the measurement of anthropometric, metabolic, and liver-related biomarkers, i.e., body mass index (BMI), homeostasis model assessment for insulin resistance (HOMA-IR), insulin, alanine aminotransferase (ALT), and aspartate aminotransferase (AST). These effects suggest the potential benefits of fiber consumption for NAFLD populations. More prospective, controlled studies should be conducted to reveal specific details regarding the fiber type, dosage, and duration for optimal intervention.
Oh T.G., Kim S.M., Caussy C., Fu T., Guo J., Bassirian S., Singh S., Madamba E.V., Bettencourt R., Richards L., Yu R.T., Atkins A.R., Huan T., Brenner D.A., Sirlin C.B., et. al.
Cell Metabolism scimago Q1 wos Q1
2020-11-01 citations by CoLab: 194 Abstract  
Dysregulation of the gut microbiome has been implicated in the progression of non-alcoholic fatty liver disease (NAFLD) to advanced fibrosis and cirrhosis. To determine the diagnostic capacity of this association, we compared stool microbiomes across 163 well-characterized participants encompassing non-NAFLD controls, NAFLD-cirrhosis patients, and their first-degree relatives. Interrogation of shotgun metagenomic and untargeted metabolomic profiles by using the random forest machine learning algorithm and differential abundance analysis identified discrete metagenomic and metabolomic signatures that were similarly effective in detecting cirrhosis (diagnostic accuracy 0.91, area under curve [AUC]). Combining the metagenomic signature with age and serum albumin levels accurately distinguished cirrhosis in etiologically and genetically distinct cohorts from geographically separated regions. Additional inclusion of serum aspartate aminotransferase levels, which are increased in cirrhosis patients, enabled discrimination of cirrhosis from earlier stages of fibrosis. These findings demonstrate that a core set of gut microbiome species might offer universal utility as a non-invasive diagnostic test for cirrhosis.
Lee J.I., Lee H.W., Lee K.S., Lee H.S., Park J.
2020-10-07 citations by CoLab: 82 Abstract  
The use of statins in nonalcoholic fatty liver disease (NAFLD) may reduce cardiovascular morbidity, although their effect on NAFLD itself is not well known. We aimed to investigate the role of statins on the development of de novo NAFLD and progression of significant liver fibrosis.This study included 11,593,409 subjects from the National Health Information Database of the Republic of Korea entered in 2010 and followed up until 2016. NAFLD was diagnosed by calculating fatty liver index (FLI), and significant liver fibrosis was evaluated using the BARD score. Controls were randomly selected at a ratio of 1:5 from individuals who were at risk of becoming the case subjects at the time of selection.Among 5,339,901 subjects that had a FLI < 30 and included in the non-NAFLD cohort, 164,856 subjects eventually had NAFLD developed. The use of statin was associated with a reduced risk of NAFLD development (adjusted odds ratio [AOR] 0.66; 95% confidence interval [CI] 0.65-0.67) and was independent of associated diabetes mellitus (DM) (with DM: AOR 0.44; 95% CI 0.41-0.46, without DM: AOR 0.71; 95% CI 0.69-0.72). From 712,262 subjects with a FLI > 60 and selected in the NAFLD cohort, 111,257 subjects showed a BARD score ≥ 2 and were defined as liver fibrosis cases. The use of statins reduced the risk of significant liver fibrosis (AOR 0.43; 95% CI 0.42-0.44), independent of DM (with DM: AOR 0.31; 95% CI 0.31-0.32, without DM: AOR 0.52; 95% CI 0.51-0.52).In this large population-based study, statin use decreased the risk of NAFLD occurrence and the risk of liver fibrosis once NAFLD developed.
Zhang X., Coker O.O., Chu E.S., Fu K., Lau H.C., Wang Y., Chan A.W., Wei H., Yang X., Sung J.J., Yu J.
Gut scimago Q1 wos Q1
2020-07-21 citations by CoLab: 554 Abstract  
ObjectiveNon-alcoholic fatty liver disease (NAFLD)-associated hepatocellular carcinoma (HCC) is an increasing healthcare burden worldwide. We examined the role of dietary cholesterol in driving NAFLD–HCC through modulating gut microbiota and its metabolites.DesignHigh-fat/high-cholesterol (HFHC), high-fat/low-cholesterol or normal chow diet was fed to C57BL/6 male littermates for 14 months. Cholesterol-lowering drug atorvastatin was administered to HFHC-fed mice. Germ-free mice were transplanted with stools from mice fed different diets to determine the direct role of cholesterol modulated-microbiota in NAFLD–HCC. Gut microbiota was analysed by 16S rRNA sequencing and serum metabolites by liquid chromatography–mass spectrometry (LC–MS) metabolomic analysis. Faecal microbial compositions were examined in 59 hypercholesterolemia patients and 39 healthy controls.ResultsHigh dietary cholesterol led to the sequential progression of steatosis, steatohepatitis, fibrosis and eventually HCC in mice, concomitant with insulin resistance. Cholesterol-induced NAFLD–HCC formation was associated with gut microbiota dysbiosis. The microbiota composition clustered distinctly along stages of steatosis, steatohepatitis and HCC. Mucispirillum, Desulfovibrio, Anaerotruncus and Desulfovibrionaceae increased sequentially; while Bifidobacterium and Bacteroides were depleted in HFHC-fed mice, which was corroborated in human hypercholesteremia patients. Dietary cholesterol induced gut bacterial metabolites alteration including increased taurocholic acid and decreased 3-indolepropionic acid. Germ-free mice gavaged with stools from mice fed HFHC manifested hepatic lipid accumulation, inflammation and cell proliferation. Moreover, atorvastatin restored cholesterol-induced gut microbiota dysbiosis and completely prevented NAFLD–HCC development.ConclusionsDietary cholesterol drives NAFLD–HCC formation by inducing alteration of gut microbiota and metabolites in mice. Cholesterol inhibitory therapy and gut microbiota manipulation may be effective strategies for NAFLD–HCC prevention.
Pengrattanachot N., Cherngwelling R., Jaikumkao K., Pongchaidecha A., Thongnak L., Swe M.T., Chatsudthipong V., Lungkaphin A.
2020-06-01 citations by CoLab: 35 Abstract  
An excessive consumption of high-fat diet can lead to the alterations of glucose and lipid metabolism, impaired insulin signaling and increased ectopic lipid accumulation resulting in renal lipotoxicity and subsequent renal dysfunction. Atorvastatin is a lipid-lowering drug in clinical treatment. Several studies have reported that atorvastatin has several significant pleiotropic effects including anti-inflammatory, antioxidant, and anti-apoptotic effects. However, the effects of atorvastatin on metabolic disturbance and renal lipotoxicity in obesity are not fully understood. In this study, obesity in rat was developed by high-fat diet (HFD) feeding for 16 weeks. After that, the HFD-fed rats were received either a vehicle (HF), atorvastatin (HFA) or vildagliptin (HFVIL), by oral gavage for 4 weeks. We found that HF rats showed insulin resistance, visceral fat expansion and renal lipid accumulation. Impaired renal function and renal organic anion transporter 3 (Oat3) function and expression were also observed in HF rats. The marked increases in MDA level, renal injury and NF-κB, TGF-β, NOX-4, PKC-α expression were demonstrated in HF rats. Atorvastatin or vildagliptin treatment attenuated insulin resistance and renal lipid accumulation-induced lipotoxicity in HFA and HFVIL rats. Moreover, the proteins involved in renal inflammation, fibrosis, oxidative stress and apoptosis were attenuated leading to improved renal Oat3 function and renal function in the treated groups. Interestingly, atorvastatin showed higher efficacy than vildagliptin in improving insulin resistance, renal lipid accumulation and in exerting renoprotective effects in obesity-induced renal injury and impaired renal Oat3 function. • Atorvastatin is able to ameliorate insulin resistance in high-fat induced obese rats. • Atorvastatin can attenuate renal lipid accumulation in high-fat induced obese rats. • Atorvastatin can reduce oxidative stress and proinflammatory cytokines in the kidney of high-fat induced obese rats. • Impaired renal Oat3 function and renal function are restored by atorvastatin treatment.
Zimmermann F., Roessler J., Schmidt D., Jasina A., Schumann P., Gast M., Poller W., Leistner D., Giral H., Kränkel N., Kratzer A., Schuchardt S., Heimesaat M.M., Landmesser U., Haghikia A.
Journal of Clinical Medicine scimago Q1 wos Q1 Open Access
2020-05-25 citations by CoLab: 21 PDF Abstract  
Background and Aims: The mechanisms of interindividual variation of lipid regulation by statins, such as the low-density lipoprotein cholesterol (LDL) lowering effects, are not fully understood yet. Here, we used a gut microbiota depleted mouse model to investigate the relation between the gut microbiota and the regulatory property of atorvastatin on blood lipids. Methods: Mice (C57BL/6) with intact gut microbiota or antibiotic induced abiotic mice (ABS) were put on standard chow diet (SCD) or high fat diet (HFD) for six weeks. Atorvastatin (10 mg/kg body weight/day) or a control vehicle were applied per gavage for the last four weeks of dietary treatment. Blood lipids including total cholesterol, very low-density lipoprotein, low-density lipoprotein, high-density lipoprotein and sphingolipids were measured to probe microbiota-dependent effects of atorvastatin. The expression of genes involved in hepatic and intestinal cholesterol metabolism was analyzed with qRT-PCR. The alteration of the microbiota profile was examined using 16S rRNA qPCR in mice with intact gut microbiota. Results: HFD feeding significantly increased total blood cholesterol and LDL levels, as compared to SCD in both mice with intact and depleted gut microbiota. The cholesterol lowering effect of atorvastatin was significantly attenuated in mice with depleted gut microbiota. Moreover, we observed a global shift in the abundance of several sphingolipids upon atorvastatin treatment which was absent in gut microbiota depleted mice. The regulatory effect of atorvastatin on the expression of distinct hepatic and intestinal cholesterol-regulating genes, including Ldlr, Srebp2 and Npc1l1 was altered upon depletion of gut microbiota. In response to HFD feeding, the relative abundance of the bacterial phyla Bacteroidetes decreased, while the abundance of Firmicutes increased. The altered ratio between Firmicutes to Bacteroidetes was partly reversed in HFD fed mice treated with atorvastatin. Conclusions: Our findings support a regulatory impact of atorvastatin on the gut microbial profile and, in turn, demonstrate a crucial role of the gut microbiome for atorvastatin-related effects on blood lipids. These results provide novel insights into potential microbiota-dependent mechanisms of lipid regulation by statins, which may account for variable response to statin treatment.
Cotter T.G., Rinella M.
Gastroenterology scimago Q1 wos Q1
2020-05-01 citations by CoLab: 859 Abstract  
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease, with a worldwide prevalence of 25%. In the United States, NAFLD and its subtype, nonalcoholic steatohepatitis, affect 30% and 5% of the population, respectively. Considering the ongoing obesity epidemic beginning in childhood, the rise in diabetes, and other factors, the prevalence of NAFLD along with the proportion of those with advanced liver disease is projected to continue to increase. This will have an important impact on public health reflected in health care costs, including impact on the need for liver transplantation, for which nonalcoholic steatohepatitis is already close to becoming the most common indication. NAFLD patients with evidence of nonalcoholic steatohepatitis and advanced fibrosis are at markedly increased risk of adverse outcomes, including overall mortality, and liver-specific morbidity and mortality, respectively. Identification of this cohort of NAFLD patients is paramount, given the associated poorer outcomes, in order to target resources to those who need it most. Various noninvasive tools have been developed in this regard. This review provides an update on the epidemiology, clinical and prognostic features, and diagnostic approach to patients with NAFLD.
Chong C.Y., Orr D., Plank L.D., Vatanen T., O'Sullivan J.M., Murphy R.
Nutrients scimago Q1 wos Q1 Open Access
2020-03-27 citations by CoLab: 47 PDF Abstract  
Background: Non-alcoholic fatty liver disease (NAFLD) can be ameliorated by weight loss although difficult to maintain. Emerging evidence indicates that prebiotics and antibiotics improve NAFLD. Aim: To determine whether inulin supplementation after brief metronidazole therapy is effective in reducing alanine aminotransferase (ALT) and maintaining weight loss achieved through a very-low-calorie diet (VLCD) among people with NAFLD. Methods: Sixty-two people with NAFLD commenced 4-week VLCD using Optifast meal replacements (600 kcal/day). Sixty were then randomised into a 12-week double-blind, placebo-controlled, parallel three-arm trial: (1) 400 mg metronidazole twice daily in Week 1 then inulin 4 g twice daily OR (2) placebo twice daily in week one then inulin OR (3) placebo-placebo. Main outcomes were ALT and body weight at 12 weeks. Fecal microbiota changes were also evaluated. Results: Mean body mass index (BMI) and ALT reduced after VLCD by 2.4 kg/m2 and 11 U/L, respectively. ALT further decreased after metronidazole-inulin compared to after placebo-placebo (mean ALT change −19.6 vs. −0.2 U/L, respectively; p = 0.026); however, weight loss maintenance did not differ. VLCD treatment decreased the ratio of Firmicutes/Bacteroidetes (p = 0.002). Conclusion: Brief metronidazole followed by inulin supplementation can reduce ALT beyond that achieved after VLCD in patients with NAFLD.
Aron-Wisnewsky J., Vigliotti C., Witjes J., Le P., Holleboom A.G., Verheij J., Nieuwdorp M., Clément K.
2020-03-09 citations by CoLab: 689 Abstract  
Gut microbiota dysbiosis has been repeatedly observed in obesity and type 2 diabetes mellitus, two metabolic diseases strongly intertwined with non-alcoholic fatty liver disease (NAFLD). Animal studies have demonstrated a potential causal role of gut microbiota in NAFLD. Human studies have started to describe microbiota alterations in NAFLD and have found a few consistent microbiome signatures discriminating healthy individuals from those with NAFLD, non-alcoholic steatohepatitis or cirrhosis. However, patients with NAFLD often present with obesity and/or insulin resistance and type 2 diabetes mellitus, and these metabolic confounding factors for dysbiosis have not always been considered. Patients with different NAFLD severity stages often present with heterogeneous lesions and variable demographic characteristics (including age, sex and ethnicity), which are known to affect the gut microbiome and have been overlooked in most studies. Finally, multiple gut microbiome sequencing tools and NAFLD diagnostic methods have been used across studies that could account for discrepant microbiome signatures. This Review provides a broad insight into microbiome signatures for human NAFLD and explores issues with disentangling these signatures from underlying metabolic disorders. More advanced metagenomics and multi-omics studies using system biology approaches are needed to improve microbiome biomarkers. The gut microbiota has been linked to non-alcoholic fatty liver disease (NAFLD), but metabolic confounding factors, such as obesity and diabetes, complicate analysis. This Review provides a broad insight into microbiome signatures for human NAFLD and explores issues with disentangling them from underlying metabolic disorders.
Qin D., Pan P., Lyu B., Chen W., Gao Y.
Biomedicine and Pharmacotherapy scimago Q1 wos Q1 Open Access
2024-08-01 citations by CoLab: 6 Abstract  
Metabolic dysfunction-associated steatotic liver disease (MASLD) has a multifactorial and complex pathogenesis. Notably, the disorder of Bile acid (BA) metabolism and lipid metabolism-induced lipotoxicity are the main risk factors of MASLD. Lupeol, traditional regional medicine from Xinjiang, has a long history of use for its anti-inflammatory, anti-tumor, and immune-modulating properties. Recent research suggests its potential as a therapeutic option for MASLD due to its proposed binding capacity to the nuclear BA receptor, Farnesoid X receptor (FXR), hence could represent a therapeutic option for MASLD. In this study, a natural triterpenoid drug lupeol improved BA metabolism and MASLD in mice through the FXR signaling pathway and the gut-liver axis. Furthermore, lupeol effectively restored gut healthiness and improved intestinal immunity, barrier integrity, and inflammation, as indicated by the reconstructed gut flora. Compared with fenofibrate (Feno), lupeol treatment significantly reduced weight gain, fat deposition, and liver injury, decreased serum total cholesterol (TC) and triglyceride (TG) levels, and alleviated hepatic steatosis and liver inflammation. BA analysis showed that lupeol treatment accelerated BA efflux and decreased uptake of BA by increasing hepatic FXR and bile salt export pump (BSEP) expression. Gut microbiota alterations could be related to enhanced fecal BA excretion in lupeol-treated mice. Therefore, consumption of lupeol may prevent HFD-induced MASLD and BA accumulation, possibly via the FXR signaling pathway and regulating the gut microbiota.

Top-30

Journals

1
1

Publishers

1
1
  • We do not take into account publications without a DOI.
  • Statistics recalculated only for publications connected to researchers, organizations and labs registered on the platform.
  • Statistics recalculated weekly.

Are you a researcher?

Create a profile to get free access to personal recommendations for colleagues and new articles.
Share
Cite this
GOST | RIS | BibTex
Found error?