101
|
Wong VWS, Zelber-Sagi S, Cusi K, Carrieri P, Wright E, Crespo J, Lazarus JV. Management of NAFLD in primary care settings. Liver Int 2022; 42:2377-2389. [PMID: 35986897 DOI: 10.1111/liv.15404] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/23/2022] [Accepted: 08/10/2022] [Indexed: 02/13/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects at least 25% of the general population and is an increasingly important cause of cirrhosis and hepatocellular carcinoma. Although it is the research focus of the hepatology field, it is clear that primary care physicians are seeing the majority of NAFLD patients and are in a pivotal position to provide quality care. In this article, we review the role of primary care in the management of NAFLD. NAFLD is common in patients with diabetes, obesity and other metabolic risk factors. Abdominal ultrasonography is the most commonly used method to diagnose fatty liver. Simple fibrosis scores have high negative predictive values in excluding advanced liver fibrosis and future liver-related events and can be used in primary care as initial evaluation. An abnormal result should be followed by subsequent workup or specialist referral. Primary care is the ideal setting to institute multidisciplinary care, especially the involvement of dietitians and physical activity trainers in lifestyle intervention, as well as initiating the discussion of bariatric surgery in patients with severe obesity. Although specific drug treatment for steatohepatitis would require a more precise diagnosis, metabolic drugs that improve both steatohepatitis and cardiovascular outcomes (e.g. glucagon-like peptide-1 receptor agonists) may be considered in patients with NAFLD.
Collapse
Affiliation(s)
- Vincent W S Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, Florida, USA
| | - Patrizia Carrieri
- Aix Marseille Univ, Inserm, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, ISSPAM, Marseille, France
| | - Eugene Wright
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Valdecilla Biomedical Research Institute (IDIVAL), Santander, University of Cantabria, Santander, Cantabria, Spain
| | - Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain.,Faculty of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
102
|
Phung HH, Lee CH. Mouse models of nonalcoholic steatohepatitis and their application to new drug development. Arch Pharm Res 2022; 45:761-794. [DOI: 10.1007/s12272-022-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
103
|
Clare K, Dillon JF, Brennan PN. Reactive Oxygen Species and Oxidative Stress in the Pathogenesis of MAFLD. J Clin Transl Hepatol 2022; 10:939-946. [PMID: 36304513 PMCID: PMC9547261 DOI: 10.14218/jcth.2022.00067] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/22/2022] [Accepted: 06/06/2022] [Indexed: 12/04/2022] Open
Abstract
The pathogenesis of metabolic-associated fatty liver disease (MAFLD) is complex and thought to be dependent on multiple parallel hits on a background of genetic susceptibility. The evidence suggests that MAFLD progression is a dynamic two-way process relating to repetitive bouts of metabolic stress and inflammation interspersed with endogenous anti-inflammatory reparative responses. In MAFLD, excessive hepatic lipid accumulation causes the production of lipotoxins that induce mitochondrial dysfunction, endoplasmic reticular stress, and over production of reactive oxygen species (ROS). Models of MAFLD show marked disruption of mitochondrial function and reduced oxidative capacitance with impact on cellular processes including mitophagy, oxidative phosphorylation, and mitochondrial biogenesis. In excess, ROS modify insulin and innate immune signaling and alter the expression and activity of essential enzymes involved in lipid homeostasis. ROS can also cause direct damage to intracellular structures causing hepatocyte injury and death. In select cases, the use of anti-oxidants and ROS scavengers have been shown to diminish the pro-apoptopic effects of fatty acids. Given this link, endogenous anti-oxidant pathways have been a target of interest, with Nrf2 activation showing a reduction in oxidative stress and inflammation in models of MAFLD. Thyroid hormone receptor β (THRβ) agonists and nuclear peroxisome proliferation-activated receptor (PPAR) family have also gained interest in reducing hepatic lipotoxicity and restoring hepatic function in models of MAFLD. Unfortunately, the true interplay between the clinical and molecular components of MAFLD progression remain only partly understood. Most recently, multiomics-based strategies are being adopted for hypothesis-free analysis of the molecular changes in MAFLD. Transcriptome profiling maps the unique genotype-phenotype associations in MAFLD and with various single-cell transcriptome-based projects underway, there is hope of novel physiological insights to MAFLD progression and uncover therapeutic targets.
Collapse
Affiliation(s)
- Kathleen Clare
- Royal Alexandra Hospital, Paisley, NHS Greater Glasgow and Clyde, PA2 9PN, UK
| | - John F. Dillon
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Paul N. Brennan
- University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
- University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, EH16 4UU, UK
- Correspondence to: Paul N. Brennan, University of Dundee, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK. ORCID: https://orcid.org/0000-0001-8368-1478. Tel: +44-7445308786, E-mail:
| |
Collapse
|
104
|
Ferroptosis: Shedding Light on Mechanisms and Therapeutic Opportunities in Liver Diseases. Cells 2022; 11:cells11203301. [PMID: 36291167 PMCID: PMC9600232 DOI: 10.3390/cells11203301] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/04/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
Cell death is a vital physiological or pathological phenomenon in the development process of the organism. Ferroptosis is a kind of newly-discovered regulated cell death (RCD), which is different from other RCD patterns, such as apoptosis, necrosis and autophagy at the morphological, biochemical and genetic levels. It is a kind of iron-dependent mode of death mediated by lipid peroxides and lipid reactive oxygen species aggregation. Noteworthily, the number of studies focused on ferroptosis has been increasing exponentially since ferroptosis was first found in 2012. The liver is the organ that stores the most iron in the human body. Recently, it was frequently found that there are different degrees of iron metabolism disorder and lipid peroxidation and other ferroptosis characteristics in various liver diseases. Numerous investigators have discovered that the progression of various liver diseases can be affected via the regulation of ferroptosis, which may provide a potential therapeutic strategy for clinical hepatic diseases. This review aims to summarize the mechanism and update research progress of ferroptosis, so as to provide novel promising directions for the treatment of liver diseases.
Collapse
|
105
|
Antioxidative Self-Assembling Nanoparticles Attenuate the Development of Steatohepatitis and Inhibit Hepatocarcinogenesis in Mice. Antioxidants (Basel) 2022; 11:antiox11101939. [PMID: 36290662 PMCID: PMC9598142 DOI: 10.3390/antiox11101939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/23/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress (OS) contributes to nonalcoholic steatohepatitis (NASH) and hepatocarcinogenesis. We investigated whether antioxidative self-assembling nanoparticles (SMAPoTN) could reduce the development of NASH and hepatocellular carcinoma (HCC) in p62/Sqstm1 and Nrf2 double knockout (DKO) mice and studied protective mechanisms. We measured disease development in male DKO mice fed a normal chow (NASH model) or a 60% high-fat diet (HFD; HCC model) with or without SMAPoTN administration for 26 weeks. SMAPoTN inhibited liver fibrosis in both groups and prevented HCC development (0% vs. 33%, p < 0.05) in the HFD group. SMAPoTN reduced OS, inflammatory cytokine signaling, and liver fibrosis. RNA-sequencing revealed that SMAPoTN decreased endoplasmic reticulum stress signaling genes in both groups, HCC driver genes, and cancer pathway genes, especially PI3K-AKT in the HFD groups. In the SMAPoTN treatment HFD group, serum lipopolysaccharide levels and liver lipopolysaccharide-binding protein expression were significantly lower compared with those in the nontreatment group. SMAPoTN improved the α-diversity of gut microbiota, and changed the microbiota composition. Oral SMAPoTN administration attenuated NASH development and suppressed hepatocarcinogenesis in DKO mice by improving endoplasmic reticulum stress in the liver and intestinal microbiota. SMAPoTN may be a new therapeutic option for NASH subjects and those with a high HCC risk.
Collapse
|
106
|
Luo Z, Gao Q, Li Y, Bai Y, Zhang J, Xu W, Xu J. Flammulina velutipes Mycorrhizae Attenuate High Fat Diet-Induced Lipid Disorder, Oxidative Stress and Inflammation in the Liver and Perirenal Adipose Tissue of Mice. Nutrients 2022; 14:nu14183830. [PMID: 36145203 PMCID: PMC9505303 DOI: 10.3390/nu14183830] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/11/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Flammulina velutipes (FV) is edible mushroom that has nutritional and medicinal values. FV mycorrhizae, the by-products of FV, are an abundant source and receive less attention. The objective of this study was to investigate the composition of FV mycorrhizae, and its effects on high fat diet (HFD)-induced lipid disorder, oxidative stress, and inflammatory cytokines, both in the liver and perirenal adipose tissue (PAT) of mice. The results showed that FV mycorrhizae contain abundant trace elements, polysaccharide, amino acids and derivatives, and organic compounds. It was found that 4% FV mycorrhizae (HFDFV) supplementation decreased HFD-induced liver weight and triglyceride (TG) in the plasma, liver and PAT, altered plasma and hepatic fatty acids profiles, promoted gene expression involved in lipid hydrolysis, fatty acid transportation and β-oxidation in the liver and reduced lipid synthesis in the liver and PAT. HFDFV attenuated HFD-induced oxidative stress and pro-inflammatory cytokine by increasing GSH/GSSG, and decreasing levels of MDA and IL6 both in the liver and PAT, while it differentially regulated gene expression of IL1β, IL6, and CCL2 in liver and PAT. The results indicated that FV mycorrhizae are effective to attenuate HFD-induced lipid disorder, oxidative stress and inflammation in the liver and PAT, indicating their promising constituents for functional foods and herbal medicine.
Collapse
Affiliation(s)
- Zhen Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qingying Gao
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuanfei Li
- Institute of Biological Technology, Nanchang Normal University, Nanchang 330032, China
| | - Yifei Bai
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Jing Zhang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weina Xu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianxiong Xu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
- Correspondence: ; Tel.: +86-021-34205767
| |
Collapse
|
107
|
E4orf1 Prevents Progression of Fatty Liver Disease in Mice on High Fat Diet. Int J Mol Sci 2022; 23:ijms23169286. [PMID: 36012550 PMCID: PMC9409018 DOI: 10.3390/ijms23169286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/05/2022] [Accepted: 08/14/2022] [Indexed: 11/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) covers a broad spectrum of liver diseases ranging from steatosis to cirrhosis. There are limited data on prevention of hepatic steatosis or its progression to liver disease. Here, we tested if either transgenic (Tg) doxycycline-induced expression in adipose tissue of E4orf1 (E4), an adenoviral protein, or dietary fat restriction attenuated hepatic steatosis or its progression in mice. Twelve to fourteen-week-old TgE4 mice (E4 group) and control mice were exposed to a 60% (Kcal) high fat diet (HFD) for 20 weeks, while another group of mice on HFD for 10 weeks were switched to a chow diet (chow group) for another 10 weeks. Glycemic control was determined at weeks 10 and 20. Tissues were collected for gene and protein analysis at sacrifice. Compared to control, diet reversal significantly reduced body weight in the chow group, whereas E4 expression attenuated weight gain, despite HFD. E4 mice evinced significantly improved glucose clearance, lower endogenous insulin secretion, reduced serum triglycerides, attenuated hepatic steatosis and inflammation. Interestingly, in spite of weight loss and lower liver fat, chow mice showed significant upregulation of hepatic genes involved in lipid metabolism. Despite HFD, E4 prevents hepatic lipid accumulation and progression of hepatic steatosis, while diet reversal maintains hepatic health, but is unable to improve molecular changes.
Collapse
|
108
|
Stierwalt HD, Morris EM, Maurer A, Apte U, Phillips K, Li T, Meers GME, Koch LG, Britton SL, Graf G, Rector RS, Mercer K, Shankar K, Thyfault JP. Rats with high aerobic capacity display enhanced transcriptional adaptability and upregulation of bile acid metabolism in response to an acute high-fat diet. Physiol Rep 2022; 10:e15405. [PMID: 35923133 PMCID: PMC9350427 DOI: 10.14814/phy2.15405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 06/09/2023] Open
Abstract
Rats selectively bred for the high intrinsic aerobic capacity runner (HCR) or low aerobic capacity runner (LCR) show pronounced differences in susceptibility for high-fat/high sucrose (HFHS) diet-induced hepatic steatosis and insulin resistance, replicating the protective effect of high aerobic capacity in humans. We have previously shown multiple systemic differences in energy and substrate metabolism that impacts steatosis between HCR and LCR rats. This study aimed to investigate hepatic-specific mechanisms of action via changes in gene transcription. Livers of HCR rats had a greater number of genes that significantly changed in response to 3-day HFHS compared with LCR rats (171 vs. 75 genes: >1.5-fold, p < 0.05). HCR and LCR rats displayed numerous baseline differences in gene expression while on a low-fat control diet (CON). A 3-day HFHS diet resulted in greater expression of genes involved in the conversion of excess acetyl-CoA to cholesterol and bile acid (BA) synthesis compared with the CON diet in HCR, but not LCR rats. These results were associated with higher fecal BA loss and lower serum BA concentrations in HCR rats. Exercise studies in rats and mice also revealed higher hepatic expression of cholesterol and BA synthesis genes. Overall, these results suggest that high aerobic capacity and exercise are associated with upregulated BA synthesis paired with greater fecal excretion of cholesterol and BA, an effect that may play a role in protection against hepatic steatosis in rodents.
Collapse
Affiliation(s)
- Harrison D. Stierwalt
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| | - E. Matthew Morris
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Adrianna Maurer
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and TherapeuticsUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | | | - Tiangang Li
- Department of PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Grace M. E. Meers
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
| | - Lauren G. Koch
- Physiology and PharmacologyThe University of ToledoToledoOhioUSA
| | | | - Greg Graf
- Department of Pharmaceutical SciencesSaha Cardiovascular Research Center, University of KentuckyLexingtonKentuckyUSA
| | - R. Scott Rector
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
- Research ServiceHarry S Truman Memorial VA HospitalColumbiaMissouriUSA
| | - Kelly Mercer
- Arkansas Children's Nutrition CenterUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Kartik Shankar
- Section of Nutrition, Department of PediatricsUniversity of Colorado School of Medicine Anschutz Medical CampusAuroraColoradoUSA
| | - John P. Thyfault
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| |
Collapse
|
109
|
Gart E, van Duyvenvoorde W, Caspers MPM, van Trigt N, Snabel J, Menke A, Keijer J, Salic K, Morrison MC, Kleemann R. Intervention with isoleucine or valine corrects hyperinsulinemia and reduces intrahepatic diacylglycerols, liver steatosis, and inflammation in Ldlr-/-.Leiden mice with manifest obesity-associated NASH. FASEB J 2022; 36:e22435. [PMID: 35830259 DOI: 10.1096/fj.202200111r] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 11/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is associated with a disturbed metabolism in liver, insulin resistance, and excessive accumulation of ectopic fat. Branched-chain amino acids (BCAAs) may beneficially modulate hepatic lipids, however, it remains unclear whether individual BCAAs can attenuate already established NASH and associated oxidative-inflammatory stress. After a 26 weeks run-in on fast food diet (FFD), obese Ldlr-/-.Leiden mice were treated for another 12 weeks with either valine or isoleucine (3% of FFD) and then compared to FFD controls. Valine and isoleucine did not affect obesity, dyslipidemia, gut permeability, or fecal fatty acid excretion, but significantly reduced hyperinsulinemia. Valine and isoleucine reduced ALT, CK18-M30, and liver steatosis with a particularly pronounced suppression of the microvesicular component (-61% by valine and -71% by isoleucine). Both BCAAs decreased intrahepatic diacylglycerols and 4-hydroxynonenal immunoreactivity, a marker for oxidative stress-induced lipid peroxidation. Functional genomics analysis demonstrated that valine and isoleucine affected BCAA metabolism genes, deactivated master regulators of anabolic pathways related to steatosis (e.g., SREBPF1), and activated master regulators of mitochondrial biogenesis (e.g., PPARGC1A) and lipid catabolism (e.g., ACOX1, AMPK). This correction of critical metabolic pathways on gene expression level was accompanied by a significant decrease in histological liver inflammation, and suppression of FFD-stimulated cytokine and chemokine proteins KC/CXCL1, MCP-1/CCL2, and MIP-2/CXCL2 and their pathways. In conclusion, dietary intervention with either valine or isoleucine corrected liver diacylglycerols, gene expression of multiple metabolic processes, and reduced NASH histology with profound hepatoprotective effects on oxidative stress and inflammatory proteins.
Collapse
Affiliation(s)
- Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - Nikki van Trigt
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Jessica Snabel
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Kanita Salic
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, the Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
110
|
Patel M. Evidence-based management of patients with nonalcoholic fatty liver disease. JAAPA 2022; 35:20-24. [PMID: 35881712 DOI: 10.1097/01.jaa.0000840492.53680.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ABSTRACT Over the past decade, fatty liver disease has become a forefront health issue. The clinical implication of this silent disease extends well beyond just the liver and is linked to a variety of health concerns, including cardiovascular disease, diabetes, and cancer. The prevalence of fatty liver disease in the United States is estimated to be 25% and increasing. This article reviews the pathophysiology of fatty liver disease, how clinicians can recognize contributing factors, and appropriate interventions based on the American Association for the Study of Liver Disease's guidelines.
Collapse
Affiliation(s)
- Minal Patel
- Minal Patel practices in gastroenterology/hepatology at Digestive Disease Associates in Wyomissing, Pa. The author has disclosed no potential conflicts of interest, financial or otherwise
| |
Collapse
|
111
|
Estimating global prevalence, incidence, and outcomes of non-alcoholic fatty liver disease from 2000 to 2021: systematic review and meta-analysis. Chin Med J (Engl) 2022; 135:1682-1691. [PMID: 36070463 PMCID: PMC9509027 DOI: 10.1097/cm9.0000000000002277] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The increasing burden of non-alcoholic fatty liver disease (NAFLD) worldwide imposes an emerging public health issue. We perform the current study to estimate the global prevalence, incidence, disease progression, and clinical outcomes of NAFLD. METHODS A systematic search was conducted in Medline, Embase, Web of Science, Google Scholar, and Cochrane CENTRAL that screened articles in English language published from January 2000 to December 2021. NAFLD prevalence, incidence, rate of disease progression, and outcomes were calculated with the DerSimonian-Laird random effects model with arcsine transformation. RESULTS Our search identified 59,156 records, of which 578 studies fulfilled our inclusion criteria. The overall prevalence of NAFLD was 29.38% (95% confidence interval [CI] 28.09-30.69) regardless of the diagnostic techniques. Looking at the group in which the diagnosis was made by ultrasound exclusively, the pooled prevalence was 30.49% (95% CI 29.55-31.43). NAFLD has become more prevalent during the year 2011-2021 (31.63%, 95% CI 30.23-33.04) compared with year 2000-2010 (27.94%, 95% CI 26.23-29.69). The pooled estimation of non-alcoholic steatohepatitis prevalence was 8.26% (95% CI 1.13-21.01), 46.49% (95% CI 35.93-57.20), and 46.72% (95% CI 37.57-55.98) in general population, NAFLD patients, and severe/morbidly obese patients, respectively. Based on a total of 110,142 newly developed NAFLD patients, the pooled incident rate was estimated as 46.24 cases per 1000 person-years (95% CI 43.21-49.30). In patients with NAFLD, the incident rate of hepatocellular carcinoma was 1.46 (95% CI 0.90-2.03) cases per 1000 person-years. The overall pooled estimate of NAFLD related mortality was 23.91 (95% CI 13.55-37.18) death per 1000 person-years. CONCLUSIONS The prevalence of NAFLD is increasing globally. It is contributing to poor clinical outcomes including hepatocellular carcinoma and death. Rising awareness and urgent actions are warranted to control the NAFLD pandemic across the globe. REGISTRATION PROSPERO, No. CRD42020171104.
Collapse
|
112
|
Pujia R, Mazza E, Montalcini T, Arturi F, Brunetti A, Aversa A, Romeo S, Perticone M, Sciacqua A, Pujia A. Liver Stiffness in Obese Hypothyroid Patients Taking Levothyroxine. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58070946. [PMID: 35888665 PMCID: PMC9316150 DOI: 10.3390/medicina58070946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
Background and Objectives: Thyroid dysfunction is associated with non-alcoholic fatty liver disease, but its role in the progression of liver damage in obese patients remains unclear. In addition, several case reports have suggested the existence of a levothyroxine-induced liver injury, which has been poorly investigated. Our aim was to verify whether a difference in the prevalence of liver fibrosis exists in a population of obese individuals taking Levothyroxine. Materials and Methods: We conducted a cross-sectional study on a population of 137 obese individuals, of which 49 were on replacement therapy with Levothyroxine. We excluded those who had hypertriglyceridemia and diabetes mellitus. All participants underwent a liver stiffness assessment by transient elastography as well as biochemical measurements. In subjects with liver fibrosis, other cause of liver fibrosis were ruled out. Results: Participants taking Levothyroxine had a higher prevalence of liver fibrosis than those not taking Levothyroxine (30.6% vs. 2.3%; p < 0.001), and these results were obtained after we made an adjustment for age (Exp(B) = 18.9; 95% CI = 4.1−87.4; p < 0.001). The liver stiffness value differed significantly between groups (6.0 ± 3.6 and 5.1 ± 1.2, p = 0.033). Of those subjects taking Levothyroxine, there were no significant differences in the dose of medication (1.21 ± 0.36 vs. 1.07 ± 0.42; p = 0.240) and treatment duration (13.7 ± 7.43 vs. 11.13 ± 6.23; p = 0.380) between those with and without liver fibrosis. Conclusions: We found, for the first time, a greater prevalence of liver fibrosis in obese individuals taking Levothyroxine than in those not taking this medication. This finding needs to be confirmed by longitudinal population studies as well as by cellular studies.
Collapse
Affiliation(s)
- Roberta Pujia
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
| | - Elisa Mazza
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy;
- Correspondence:
| | - Franco Arturi
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
| | - Antonio Brunetti
- Department of Health Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Antonio Aversa
- Department of Clinical and Experimental Medicine, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Stefano Romeo
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
- Department of Molecular and Clinical Medicine, The University of Gothenburg, 40530 Gothenburg, Sweden
| | - Maria Perticone
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
| | - Angela Sciacqua
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
| | - Arturo Pujia
- Department of Medical and Surgical Science, University Magna Grecia of Catanzaro, 88100 Catanzaro, Italy; (R.P.); (E.M.); (F.A.); (S.R.); (M.P.); (A.S.); (A.P.)
| |
Collapse
|
113
|
Cigrovski Berkovic M, Rezic T, Bilic-Curcic I, Mrzljak A. Semaglutide might be a key for breaking the vicious cycle of metabolically associated fatty liver disease spectrum? World J Clin Cases 2022; 10:6759-6768. [PMID: 36051145 PMCID: PMC9297405 DOI: 10.12998/wjcc.v10.i20.6759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/10/2022] [Accepted: 05/14/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolically associated fatty liver disease (MAFLD) is a liver manifestation of metabolic syndrome potentially related to unfavorable hepatic and extrahepatic outcomes and progression to cirrhosis. Up to date, there are no approved pharmacotherapies for the treatment of MAFLD, so management focused on lifestyle interventions to encourage weight loss, and treatment of coexisting conditions is the only available option. Unfortunately, the aforementioned is often not potent enough to offer reversal or slow down hepatic inflammation and fibrosis. Glucagon-like peptide-1 receptor agonists have a favorable effect on glycemic management and weight loss of patients with type 2 diabetes mellitus and recently published data suggest their potential in MAFLD treatment. In addition, some of the agents have proven cardiovascular and renal benefits in dedicated cardiovascular outcome trials, making them an interesting therapeutic option. In this opinion review, we discuss the role of semaglutide in MAFLD.
Collapse
Affiliation(s)
- Maja Cigrovski Berkovic
- Department of Endocrinology, Diabetes, Metabolism and Clinical Pharmacology, Clinical Hospital Dubrava, Zagreb 10000, Croatia
- Faculty of Kinesiology, University of Zagreb, Zagreb 10000, Croatia
| | - Tanja Rezic
- Department of Endocrinology and Diabetes, Clinical Hospital Dubrava, Zagreb 10000, Croatia
| | - Ines Bilic-Curcic
- Department of Endocrinology and Diabetes, University Hospital Centre Osijek, Osijek 31000, Croatia
| | - Anna Mrzljak
- Department of Gastroenterology and Hepatology, University Hospital Centre Zagreb, Zagreb 10000, Croatia
- School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| |
Collapse
|
114
|
Engin B, Willis SA, Malaikah S, Sargeant JA, Yates T, Gray LJ, Aithal GP, Stensel DJ, King JA. The effect of exercise training on adipose tissue insulin sensitivity: A systematic review and meta-analysis. Obes Rev 2022; 23:e13445. [PMID: 35319136 DOI: 10.1111/obr.13445] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
Abstract
This systematic review and meta-analysis determined the impact of exercise training on adipose tissue insulin sensitivity in adults. Its scope extended to studies measuring whole-body and localized subcutaneous adipose tissue insulin sensitivity using validated techniques. Consensus from four studies demonstrates that exercise training improved whole-body adipose tissue insulin sensitivity when measured via stable-isotope lipid tracers (rate of appearance suppression in response to hyperinsulinemia). Meta-analysis of 20 studies (26 intervention arms) employing the adipose tissue insulin resistance index (ADIPO-IR) supported these findings (-10.63 [-14.12 to -7.15] pmol·L-1 × mmol·L-1 ). With ADIPO-IR, this response was greater in studies documenting weight loss and shorter sampling time (≤48 h) post-training. Overall, exercise training did not affect whole-body adipose tissue insulin sensitivity in seven studies (11 intervention arms) measuring the suppression of circulating non-esterified fatty acids in response to insulin infusion (1.51 [-0.12 to 3.14]%); however, subgroup analysis identified an enhanced suppression post-training in trials reporting weight loss. From four microdialysis studies, consensus indicates no effect of exercise training on localized (abdominal/femoral) adipose tissue insulin sensitivity, potentially suggesting that enhanced whole-body responses are related to improvements in central adipose depots. However, heterogeneity within microdialysis protocols dictates that findings must be viewed with caution.
Collapse
Affiliation(s)
- Buket Engin
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK
| | - Scott A Willis
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK
| | - Sundus Malaikah
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK.,Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jack A Sargeant
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK.,Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Thomas Yates
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK.,Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Laura J Gray
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Guruprasad P Aithal
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and The University of Nottingham, Nottingham, UK
| | - David J Stensel
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK
| | - James A King
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, UK
| |
Collapse
|
115
|
Zhao J, Fan H, Wang T, Yu B, Mao S, Wang X, Zhang W, Wang L, Zhang Y, Ren Z, Liang B. TyG index is positively associated with risk of CHD and coronary atherosclerosis severity among NAFLD patients. Cardiovasc Diabetol 2022; 21:123. [PMID: 35778734 PMCID: PMC9250269 DOI: 10.1186/s12933-022-01548-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022] Open
Abstract
Background Insulin resistance (IR), endothelial dysfunction, inflammation, glucose and lipid metabolism disorders, and thrombosis are believed involved in coronary heart disease (CHD) and non-alcoholic fatty liver disease (NAFLD). Triglyceride-glucose (TyG) index, a new IR indicator, is correlated with NAFLD occurrence and severity, but its relationship with CHD risk remains unclear. This study investigated the correlation between TyG index and CHD risk among NAFLD patients. Methods This cross-sectional study included 424 patients with NAFLD and chest pain in the Department of Cardiology, The Second Hospital of Shanxi Medical University, from January 2021 to December 2021. The TyG index was calculated and coronary angiography performed. All individuals were divided into NAFLD + CHD and NAFLD groups and then by TyG index level. The t-test, Mann–Whitney U-test, or one-way analysis of variance compared differences in continuous variables, while the chi-square test or Fisher’s exact test compared differences in categorical variables. Logistic regression analysis determined the independent protective or hazardous factors of NAFLD with CHD. The receiver operating characteristic curve evaluated the ability of different TyG index rule-in thresholds to predict CHD. The relationship between Gensini score and TyG index was evaluated using linear correlation and multiple linear regression. Results CHD was detected in 255 of 424 patients. Compared to NAFLD group, multivariate logistic regression showed that TyG index was a risk factor for CHD among NAFLD patients after adjustment for age, sex, hypertension, and diabetes mellitus with the highest odds ratio (OR, 2.519; 95% CI, 1.559–4.069; P < 0.001). TG, low-density lipoprotein cholesterol, FBG and TYG–body mass index were also risk factors for CHD among NAFLD patients. High-density lipoprotein cholesterol level was a protective factor for CHD events in patients with NAFLD. In an in-depth analysis, multivariate logistic regression analysis showed that each 1-unit increase in TyG index was associated with a 2.06-fold increased risk of CHD (OR, 2.06; 95% CI, 1.16–3.65; P = 0.013). The multifactor linear regression analysis showed each 0.1-unit increase in TyG in the NAFLD-CHD group was associated with a 2.44 increase in Gensini score (β = 2.44; 95% CI, 0.97–3.91; P = 0.002). Conclusions The TyG index was positively correlated with CHD risk in NAFLD patients and reflected coronary atherosclerosis severity.
Collapse
Affiliation(s)
- Jianqi Zhao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Hongxuan Fan
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Ting Wang
- Department of Neurology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Bing Yu
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Shaobin Mao
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Xun Wang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Wenjing Zhang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Leigang Wang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Yao Zhang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Zhaoyu Ren
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
116
|
Lin L, Wang L, Du R, Hu C, Lu J, Wang T, Li M, Zhao Z, Xu Y, Xu M, Bi Y, Wang W, Ning G, Chen Y. Arterial Stiffness, Biomarkers of Liver Fat, and the Development of Metabolic Dysfunction in Metabolically Healthy Population: A Prospective Study. Front Cardiovasc Med 2022; 9:928782. [PMID: 35811692 PMCID: PMC9261979 DOI: 10.3389/fcvm.2022.928782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Background Metabolic dysfunction is known to be associated with arterial stiffness. However, the risks of metabolic syndrome and diabetes due to arterial stiffness and the potential mechanism remain unclear. We aimed to investigate the association of arterial stiffness with the risk of metabolic syndrome and diabetes, and determine whether this association is mediated by liver fat. Methods A prospective study was conducted with 4,139 Chinese adults who were metabolically healthy at baseline. Arterial stiffness was measured by brachial-ankle pulse wave velocity (baPWV). Obesity was defined as body mass index ≥25 kg/m2. The primary outcomes were incident metabolic syndrome and diabetes. Results During a median follow-up of 4.4 years, 1,022 (24.7%) and 354 (9.5%) participants developed metabolic syndrome and diabetes, respectively. Compared with those in the lowest quartile of baPWV, participants in the highest quartile had 85 and 91% higher risks of metabolic syndrome and diabetes [risk ratio (RR) 1.85, 95% confidence interval (CI) 1.41, 2.42 for metabolic syndrome; RR 1.91, 95% CI 1.16, 3.15 for diabetes]. Mediation analyses indicated that fatty liver significantly mediated the association of arterial stiffness with metabolic syndrome and diabetes risk. Specifically, 18.4% of metabolic syndrome and 12.6% of diabetes risk due to arterial stiffness were mediated through fatty liver. Conclusions Arterial stiffness was associated with higher risks of metabolic syndrome and diabetes in individuals with obesity. This association may be partially mediated by fatty liver.
Collapse
Affiliation(s)
- Lin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Du
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
117
|
Fang Q, Li X, Wang M, Qiao X, Huang F, Hu C, Xue Y, Zhao S, Lin Y. Walnut green husk ethanol extract improves gut microbiota and their metabolites associated with NLRP3 in non-alcoholic steatohepatitis. Food Funct 2022; 13:6387-6403. [PMID: 35616069 DOI: 10.1039/d2fo00012a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Increasing studies have shown that walnut green husk (WGH) has obvious effects on reducing lipid, resisting oxidation, and protecting the liver. However, the mechanism by which WGH can prevent high-fat diet (HFD)-induced non-alcoholic steatohepatitis (NASH) remains unclear. This study is aimed at investigating the effects of WGH ethanol extract (WGHE) on NLRP3-related biochemical indicators and the diversity and metabolism of gut microbiota in HFD-induced NASH rats. WGHE was administered to HFD-induced NASH rats for 6 weeks. The results showed that WGHE could decrease the levels of blood and liver TC, TG, LDL-C, AST, and ALT and the levels of liver indices, including IL-1β, IL-6, TNF-α, TGF-β, FFA, VLDL, caspase-1, ASC, and NLRP3, while it could increase the levels of HDL-C. The pathological damage to liver tissues was significantly reduced. Moreover, WGHE could reduce the Firmicutes/Bacteroidetes (F/B) ratio and the relative abundances of potentially harmful bacteria, such as Lachnospiraceae and Christensenellaceae, and increase that of potentially beneficial bacteria, such as norank_f__Muribaculaceae. These bacteria were associated with NASH and most of them were significantly associated. A total of 23 gut bacteria and 31 metabolites were significantly altered by HFD, which was reversed by WGHE. The common functional pathways, including lipid metabolism and steroid biosynthesis, were identified through the analysis of KEGG metabolic pathways. In addition, the changes in gut microbiota, such as unclassified_f__Lachnospiraceae, unclassified_g__Blautia, and unclassified_g__Desulfovibrio, were associated with the changes in key intestinal metabolites, such as arachidonoyl amine, xanthine, and 25,26-epoxy-1α-hydroxyvitamin D3. In conclusion, WGHE could mitigate HFD-induced NASH in rats by interfering with the NLRP3-related gut microbiota and their metabolites.
Collapse
Affiliation(s)
- Qionglian Fang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Xinping Li
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Mengmeng Wang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Xue Qiao
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Feng Huang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Chunyan Hu
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Yongmei Xue
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Shenglan Zhao
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| | - Yuping Lin
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Resource, Kunming 650500, P.R. China.
| |
Collapse
|
118
|
Yamaguchi T, Yoshida K, Murata M, Suwa K, Tsuneyama K, Matsuzaki K, Naganuma M. Smad3 Phospho-Isoform Signaling in Nonalcoholic Steatohepatitis. Int J Mol Sci 2022; 23:ijms23116270. [PMID: 35682957 PMCID: PMC9181097 DOI: 10.3390/ijms23116270] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis with insulin resistance, oxidative stress, lipotoxicity, adipokine secretion by fat cells, endotoxins (lipopolysaccharides) released by gut microbiota, and endoplasmic reticulum stress. Together, these factors promote NAFLD progression from steatosis to nonalcoholic steatohepatitis (NASH), fibrosis, and eventually end-stage liver diseases in a proportion of cases. Hepatic fibrosis and carcinogenesis often progress together, sharing inflammatory pathways. However, NASH can lead to hepatocarcinogenesis with minimal inflammation or fibrosis. In such instances, insulin resistance, oxidative stress, and lipotoxicity can directly lead to liver carcinogenesis through genetic and epigenetic alterations. Transforming growth factor (TGF)-β signaling is implicated in hepatic fibrogenesis and carcinogenesis. TGF-β type I receptor (TβRI) and activated-Ras/c-Jun-N-terminal kinase (JNK) differentially phosphorylate the mediator Smad3 to create two phospho-isoforms: C-terminally phosphorylated Smad3 (pSmad3C) and linker-phosphorylated Smad3 (pSmad3L). TβRI/pSmad3C signaling terminates cell proliferation, while constitutive Ras activation and JNK-mediated pSmad3L promote hepatocyte proliferation and carcinogenesis. The pSmad3L signaling pathway also antagonizes cytostatic pSmad3C signaling. This review addresses TGF-β/Smad signaling in hepatic carcinogenesis complicating NASH. We also discuss Smad phospho-isoforms as biomarkers predicting HCC in NASH patients with or without cirrhosis.
Collapse
Affiliation(s)
- Takashi Yamaguchi
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
- Correspondence: ; Tel.: +81-72-804-0101; Fax: +81-72-804-2524
| | - Katsunori Yoshida
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Miki Murata
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Kanehiko Suwa
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Koichi Tsuneyama
- Department of Pathology & Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto, Tokushima 770-8503, Japan;
| | - Koichi Matsuzaki
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Makoto Naganuma
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| |
Collapse
|
119
|
Abstract
Initially a condition that received limited recognition and whose clinical impact was controversial, non-alcoholic steatohepatitis (NASH) has become a leading cause of chronic liver disease. Although there are no approved therapies, major breakthroughs, which will be reviewed here, have paved the way for future therapeutic successes. The unmet medical need in NASH is no longer disputed, and progress in the understanding of its pathogenesis has resulted in the identification of many pharmacological targets. Key surrogate outcomes for therapeutic trials are now accepted by regulatory agencies, thus creating a path for drug registration. A set of non-invasive measurements enabled early-stage trials to be conducted expeditiously, thus providing early indications on the biological and possibly clinical actions of therapeutic candidates. This generated efficacy results for a number of highly promising compounds that are now in late-stage development. Intense research aimed at further improving the assessment of histological endpoints and in developing non-invasive predictive biomarkers is underway. This will help improve the design and feasibility of successful trials, ultimately providing patients with therapeutic options that can change the course of the disease.
Collapse
|
120
|
Ajah AA, Lembede BW, Nkomozepi P, Erlwanger KH, Nyakudya TT. Neonatal Oral Administration of Chrysin Prevents Long-Term Development of Non-Alcoholic Fatty Liver Disease in a Sexually Dimorphic Manner in Fructose Nurtured Sprague Dawley Rats. Life (Basel) 2022; 12:life12060790. [PMID: 35743821 PMCID: PMC9225280 DOI: 10.3390/life12060790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/30/2022] [Accepted: 05/23/2022] [Indexed: 12/03/2022] Open
Abstract
High-fructose diets are linked with the development of non-alcoholic fatty liver disease (NAFLD), the management of which is a burden to society. Interventions with phytochemicals in the early postnatal period may prevent fructose-induced NAFLD later in adulthood. We investigated the protective potential of chrysin against fructose-induced NAFLD. Four-day-old male and female suckling Sprague Dawley rats (N = 112) were randomly grouped and orally gavaged daily with distilled water (negative Control-Cn + W), chrysin(Chr-100 mg/kg), fructose-solution (Fr-20% w/v), and Chr + Fr between postnatal day (PND) 4 and 21 and then weaned onto normal rat chow and plain drinking water to PND 55. From PND 56 to 130, half of the rats continued on plain water, and the rest had Fr as drinking fluid. Terminally, the liver tissue was collected, and the lipid content was determined and histologically assessed for NAFLD. Dietary Fr induced an increased hepatic lipid content (p = 0.0001 vs. Cn + W) both sexes, and it was only attenuated by neonatal Chr in female rats (p < 0.05). Histologically, there was increased microvesicular steatosis (p = 0.0001 vs. Cn + W) in both sexes, and it was prevented by neonatal Chr (p > 0.05). Fr caused macrovesicular steatosis (p = 0.01 vs. Cn + W) in females only, and chrysin did not prevent it (p > 0.05). Fr induced hepatocellular hypertrophy, and inflammation was observed in females only (p = 0.01 vs. Cn + W), and this was prevented by Chr (p > 0.05). The collagen area fraction was increased by Fr (p = 0.02 (males) and p = 0.04 (females) vs. Cn + W, respectively; however, chrysin did not prevent this (p > 0.05). Neonatal chrysin prevented some of the deleterious effects of the high-fructose diet on the liver, suggesting that chrysin should be further explored as a strategic prophylactic neonatal intervention against high-fructose-diet-induced NAFLD.
Collapse
Affiliation(s)
- Austin A. Ajah
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (B.W.L.); (K.H.E.)
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, University of Port Harcourt, P.M.B. 5323, Choba, Port Harcourt 500102, Nigeria
- Correspondence:
| | - Busisani W. Lembede
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (B.W.L.); (K.H.E.)
| | - Pilani Nkomozepi
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Corner Beit and Siemert Street, Doornfontein, Johannesburg 2094, South Africa; (P.N.); (T.T.N.)
| | - Kennedy H. Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (B.W.L.); (K.H.E.)
| | - Trevor T. Nyakudya
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Corner Beit and Siemert Street, Doornfontein, Johannesburg 2094, South Africa; (P.N.); (T.T.N.)
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa
| |
Collapse
|
121
|
Shao J, Ge T, Wei Y, Zhou Y, Shi M, Liu H, Chen Z, Xia Y. Co-interventions with Clostridium butyricum and soluble dietary fiber targeting the gut microbiota improve MAFLD via the Acly/Nrf2/NF-κB signaling pathway. Food Funct 2022; 13:5807-5819. [PMID: 35543143 DOI: 10.1039/d1fo04224f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Purpose: The pathogenesis of metabolic associated fatty liver disease (MAFLD) is complex. Lipid metabolic disorder, chronic inflammation, and oxidative stress are the core events for MAFLD. Dietary intervention is an important treatment strategy for preventing the onset and progression of MAFLD. Clostridium butyricum (CB) and soluble dietary fiber (SDF) are often considered beneficial for health. We explored how two microbiota-targeted interventions (SDF and CB) influence the hepatic immune system, oxidative stress, and lipid metabolism in MAFLD mice. Methods: To explore the role of SDF and CB in MAFLD, we generated MAFLD mouse models by feeding C57BL/6 mice with a high-fat diet (HFD). After 8 weeks of intervention, we measured immune cell function, lipid metabolism, and oxidative stress levels in the livers of mice. Results: Single intervention with SDF or CB was not effective in improving MAFLD; however, co-interventions with SDF and CB increased microbiota diversity and decreased inflammation, oxidative stress, and lipid synthesis. Moreover, we determined that co-intervention with SDF and CB mediated fatty acid oxidation by activating the Acly/Nrf2/NF-κB signaling pathway. Most importantly, co-intervention exerted anti-inflammatory effects by inhibiting the differentiation of macrophages into pro-inflammatory M1 macrophages. Conclusion: This study show that co-intervention with SDF and CB can improve MAFLD, and co-intervention with SDF and CB are suggested to be potential gut microbiota modulators and therapeutic substances for MAFLD.
Collapse
Affiliation(s)
- Junwei Shao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, China.
| | - Tiantian Ge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, China.
| | - Yingliang Wei
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang, Liaoning, 110004, China
| | - Yuhan Zhou
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Mengyuan Shi
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Huiyuan Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, College of Medicine, Zhejiang University, China.
| | - Yang Xia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
122
|
Branković M, Jovanović I, Dukić M, Radonjić T, Oprić S, Klašnja S, Zdravković M. Lipotoxicity as the Leading Cause of Non-Alcoholic Steatohepatitis. Int J Mol Sci 2022; 23:ijms23095146. [PMID: 35563534 PMCID: PMC9105530 DOI: 10.3390/ijms23095146] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/30/2022] [Accepted: 04/30/2022] [Indexed: 12/11/2022] Open
Abstract
The emerging issues nowadays are non-alcoholic fatty liver disease (NAFLD) and its advanced stage non-alcoholic steatohepatitis (NASH), which further can be a predisposing factor for chronic liver complications, such as cirrhosis and/or development of hepatocellular carcinoma (HCC). Liver lipotoxicity can influence the accumulation of reactive oxygen species (ROS), so oxidative stress is also crucial for the progression of NASH. Moreover, NASH is in strong connection with metabolic disorders, and supporting evidence shows that insulin resistance (IR) is in a close relation to NAFLD, as it is involved in the progression to NASH and further progression to hepatic fibrosis. The major issue is that, at the moment, NASH treatment is based on lifestyle changes only due to the fact that no approved therapeutic options are available. The development of new therapeutic strategies should be conducted towards the potential NAFLD and NASH treatment by the modulation of IR but also by dietary antioxidants. As it seems, NASH is going to be the leading indication for liver transplantation as a consequence of increased disease prevalence and the lack of approved treatment; thus, an effective solution is needed as soon as possible.
Collapse
Affiliation(s)
- Marija Branković
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence:
| | - Igor Jovanović
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
| | - Marija Dukić
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
| | - Tijana Radonjić
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
| | - Svetlana Oprić
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
| | - Slobodan Klašnja
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
| | - Marija Zdravković
- University Hospital Medical Center Bežanijska kosa, Dr Žorža Matea bb, 11000 Belgrade, Serbia; (I.J.); (M.D.); (T.R.); (S.O.); (S.K.); (M.Z.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
123
|
Ren G, Hwang PTJ, Millican R, Shin J, Brott BC, van Groen T, Powell CM, Bhatnagar S, Young ME, Jun HW, Kim JA. Subcutaneous Administration of a Nitric Oxide-Releasing Nanomatrix Gel Ameliorates Obesity and Insulin Resistance in High-Fat Diet-Induced Obese Mice. ACS APPLIED MATERIALS & INTERFACES 2022; 14:19104-19115. [PMID: 35467831 PMCID: PMC9233978 DOI: 10.1021/acsami.1c24113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Nitric oxide (NO) is a gaseous signaling molecule, which plays crucial roles in various biological processes, including inflammatory responses, metabolism, cardiovascular functions, and cognitive function. NO bioavailability is reduced with aging and cardiometabolic disorders in humans and rodents. NO stimulates the metabolic rate by increasing the mitochondrial biogenesis and brown fat activation. Therefore, we propose a novel technology of providing exogenous NO to improve the metabolic rate and cognitive function by promoting the development of brown adipose tissue. In the present study, we demonstrate the effects of the peptide amphiphiles-NO-releasing nanomatrix gel (PANO gel) on high-fat diet-induced obesity, insulin resistance, and cognitive functions. Eight-week-old male C57BL/6 mice were subcutaneously injected in the brown fat area with the PANO gel or vehicle (PA gel) every 2 weeks for 12 weeks. The PANO gel-injected mice gained less body weight, improved glucose tolerance, and decreased fasting serum insulin and leptin levels compared with the PA gel-injected mice. Insulin signaling in the muscle, liver, and epididymal white adipose tissue was improved by the PANO gel injection. The PANO gel reduced inflammation, increased lipolysis in the epididymal white adipose tissue, and decreased serum lipids and liver triglycerides. Interestingly, the PANO gel stimulated uncoupled protein 1 gene expression in the brown and beige fat tissues. Furthermore, the PANO gel increased the cerebral blood flow and improved learning and memory abilities. Our results suggest that using the PANO gel to supply exogenous NO is a novel technology to treat metabolic disorders and cognitive dysfunctions.
Collapse
Affiliation(s)
- Guang Ren
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294
| | | | | | - Juhee Shin
- Department of Biomedical engineering, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Brigitta C. Brott
- Endomimetics, LLC, Birmingham, AL 35242
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Thomas van Groen
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Craig M. Powell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294
- UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Martin E. Young
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL 35294
- UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Ho-Wook Jun
- Endomimetics, LLC, Birmingham, AL 35242
- Department of Biomedical engineering, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jeong-a Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294
- UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
124
|
Ma R, Zhan Y, Zhang Y, Wu L, Wang X, Guo M. Schisandrin B ameliorates non-alcoholic liver disease through anti-inflammation activation in diabetic mice. Drug Dev Res 2022; 83:735-744. [PMID: 34927282 PMCID: PMC9299884 DOI: 10.1002/ddr.21905] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 11/14/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic risk factor associated with non-alcoholic liver disease (NAFLD). Schisandrin B (Sch B) is a promising agent for NAFLD. However, the actions of Sch B on diabetes-associated NAFLD and the underlying mechanisms are not characterized. This study aimed to assess whether Sch B has beneficial effects on T2DM-associated NAFLD. Sch B (50 mg/kg, gavage) was administrated to C57BL/KSJ db/db mice for 2 weeks. Body weight, liver weight, blood glucose, and insulin resistance were measured. Serum lipid level and liver function were detected using the biochemistry analyzer. Quantitative Real-Time PCR assay was used to evaluate mRNA levers of lipid metabolism genes. Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling (TUNEL) staining was performed to measure apoptosis in the liver. Pathological analysis and immunohistochemistry assessment were used to analyze hepatic steatosis and inflammatory infiltration. Sch B supplementation significantly decrease body weight, related liver weight, blood glucose, and serum insulin, and improved insulin resistance in db/db mice. Sch B obviously corrected NAFLD phenotypes including lipid deposition, steatohepatitis, and high levels of hepatic enzymes and serum lipid. In addition, mRNA levels of Sterol response element-bind protein 1c (SREBP-1c), fatty acid synthetase (Fasn), and acetyl-CoA carboxylase (ACC) were markedly downregulated by Sch B treatment. TUNEL-positive cells were also decreased by Sch B. Furthermore, Sch B inhibited the Kupffer cells, IL-1β, and TNF-α infiltration to the liver. Sch B ameliorated insulin resistance and lipid accumulation under high glucose conditions, which was partly associated with its inhibition of apoptosis and anti-inflammatory actions.
Collapse
Affiliation(s)
- Ruojia Ma
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Yike Zhan
- Department of CardiologyJieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat‐sen University)JieyangChina
| | - Yamei Zhang
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Liangan Wu
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Xing Wang
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| | - Ming Guo
- Department of CardiologyAffiliated Xiaoshan Hospital, Hangzhou Normal UniversityHangzhouChina
| |
Collapse
|
125
|
Gado TH, Asal SA, Abdelraouf FH, AbdelSamie R. Evaluating Serum Neuregulin 4 as a Noninvasive Biomarker in Patients with Non-alcoholic Fatty Liver Disease. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) spectrum ranges from simple hepatic steatosis to non-alcoholic steatohepatitis. Considering the restrictions of liver biopsy, various serological biomarkers have recently emerged for non-invasive diagnosis of NAFLD.
AIM: This study aimed to evaluate the association between serum neuregulin 4 (Nrg4) and NAFLD and the use of serum Nrg4 as a noninvasive marker for diagnosis of NAFLD.
METHODS: Sixty-three Egyptian NAFLD patients and 63 controls were enrolled and subjected to detailed history taking, thorough clinical examination including anthropometric measures (body mass index [BMI] and waist circumference). Laboratory investigations included complete blood count, lipid profile (total cholesterol, low-density lipoprotein cholesterol [LDL-C], high-density lipoprotein-cholesterol [HDL-C], triglycerides), serum albumin, transaminases, bilirubin levels, hepatitis markers (anti-HCV antibodies and hepatitis B surface antigen), anti-nuclear antibodies and anti-smooth muscle antibody. Measurement of serum Nrg4 by ELISA and non-invasive NAFLD scores such as NAFLD fibrosis score and FIB4 score were applied to all patients.
RESULTS: There was a statistically significant difference between cases and controls regarding the BMI, waist circumference, hemoglobin level, total leukocytic count, total cholesterol, LDL-C, HDL-C, and serum triglycerides levels. Nrg4 was significantly decreased in NAFLD patients as compared to controls. Moreover, Nrg4, total cholesterol, and LDL-C levels were statistically significant independent predictors of NAFLD. No significant differences were observed between Nrg4 level and the variable stages of hepatic fibrosis by NAFLD fibrosis score.
CONCLUSION: Decreased serum Nrg4 level is frequent in NAFLD patients and is an independent predictor of NAFLD, suggesting that Nrg4 might have a potential role in prevention and treatment of NAFLD.
Collapse
|
126
|
Lange NF, Graf V, Caussy C, Dufour JF. PPAR-Targeted Therapies in the Treatment of Non-Alcoholic Fatty Liver Disease in Diabetic Patients. Int J Mol Sci 2022; 23:ijms23084305. [PMID: 35457120 PMCID: PMC9028563 DOI: 10.3390/ijms23084305] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR), ligand-activated transcription factors of the nuclear hormone receptor superfamily, have been identified as key metabolic regulators in the liver, skeletal muscle, and adipose tissue, among others. As a leading cause of liver disease worldwide, non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) cause a significant burden worldwide and therapeutic strategies are needed. This review provides an overview of the evidence on PPAR-targeted treatment of NAFLD and NASH in individuals with type 2 diabetes mellitus. We considered current evidence from clinical trials and observational studies as well as the impact of treatment on comorbid metabolic conditions such as obesity, dyslipidemia, and cardiovascular disease. Future areas of research, such as possible sexually dimorphic effects of PPAR-targeted therapies, are briefly reviewed.
Collapse
Affiliation(s)
- Naomi F. Lange
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, 3012 Bern, Switzerland
- Correspondence: (N.F.L.); (J.-F.D.)
| | - Vanessa Graf
- Department of Diabetes, Endocrinology, Clinical Nutrition, and Metabolism, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
| | - Cyrielle Caussy
- Univ Lyon, CarMen Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, 69495 Pierre-Bénite, France;
- Département Endocrinologie, Diabète et Nutrition, Hôpital Lyon Sud, Hospices Civils de Lyon, 69495 Pierre-Bénite, France
| | - Jean-François Dufour
- Centre des Maladies Digestives, 1003 Lausanne, Switzerland
- Swiss NASH Foundation, 3011 Bern, Switzerland
- Correspondence: (N.F.L.); (J.-F.D.)
| |
Collapse
|
127
|
Association of Non-Alcoholic Fatty Liver Disease and Hepatic Fibrosis with Epicardial Adipose Tissue Volume and Atrial Deformation Mechanics in a Large Asian Population Free from Clinical Heart Failure. Diagnostics (Basel) 2022; 12:diagnostics12040916. [PMID: 35453964 PMCID: PMC9033151 DOI: 10.3390/diagnostics12040916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and cardiovascular disease share several cardiometabolic risk factors. Excessive visceral fat can manifest as ectopic fat depots over vital organs, such as the heart and liver. This study assessed the associations of NAFLD and liver fibrosis with cardiac structural and functional disturbances. We assessed 2161 participants using ultrasound, and categorized them as per the NAFLD Fibrosis Score into three groups: (1) non-fatty liver; (2) fatty liver with low fibrosis score; and (3) fatty liver with high fibrosis score. Epicardial fat volume (EFV) was measured through multidetector computed tomography. All participants underwent echocardiographic study, including tissue Doppler-based E/e’ ratio and speckle tracking-based left ventricular global longitudinal strain, peak atrial longitudinal strain (PALS), and atrial longitudinal strain rates during systolic, early and late-diastolic phases (ALSRsyst, ALSRearly. ALSRlate). Larger EFV, decreased e’ velocity, PALS, ALSRsyst, and ALSRearly, along with elevated E/e’ ratio, were seen in all groups, especially in those with high fibrosis scores. After multivariate adjustment for traditional risk factors and EFV, fibrosis scores remained significantly associated with elevated E/e’ ratio, LA stiffness, and decreased PALS (β: 0.06, 1.4, −0.01, all p < 0.05). Thus, NAFLD is associated with LV diastolic dysfunction and subclinical changes in LA contractile mechanics.
Collapse
|
128
|
Progression of Nonalcoholic Fatty Liver Disease-Associated Fibrosis in a Large Cohort of Patients with Type 2 Diabetes. Dig Dis Sci 2022; 67:1379-1388. [PMID: 33779880 DOI: 10.1007/s10620-021-06955-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/12/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) can progress to advanced fibrosis, especially in patients with type 2 diabetes. Small studies have shown that fibrosis can also regress. AIM We aimed to provide large-scale data on progression and regression of fibrosis in diabetics with NAFLD. METHODS Adult diabetic patients with the diagnosis of NAFLD based on ICD-9 codes were identified. We used scores from noninvasive tests to identify patients with advanced fibrosis, calculated at first assessment and last follow-up visit. Cutoff values for advanced fibrosis were AST: ALT ratio > 1.4, AST to platelet ratio index > 1.5, FIB-4 score > 2.67, and NAFLD fibrosis score > 0.676. RESULTS Our cohort included 50,695 diabetics with NAFLD (55.3% female; 71% Caucasian; mean age, 51.2 ± 11.6 y). During median follow-up of 84.4 months, 25.8% transitioned from no advanced fibrosis to advanced fibrosis (progression), 6.4% transitioned from advanced fibrosis to no advanced fibrosis (regression), and the rest remained stable. Factors associated with transition to advanced fibrosis were female sex, older age at first evaluation, African-American race, obesity, chronic kidney disease, or coronary artery disease. Use of insulin increased the risk of progression to advanced fibrosis (odds ratio,1.36; p < .001), whereas use of oral hypoglycemic agents, angiotensin 2 receptor blockers, and fibrates was associated with reduced risk (odds ratios, 0.92, 0.94 and 0.90, respectively; all p < .05). CONCLUSIONS In a large cohort of patients with type 2 diabetes and NAFLD, more than a quarter progressed to advanced fibrosis. These findings indicate the need for early detection and staging of NAFLD in diabetics.
Collapse
|
129
|
Barritt AS, Marshman E, Noureddin M. Review article: role of glucagon-like peptide-1 receptor agonists in non-alcoholic steatohepatitis, obesity and diabetes-what hepatologists need to know. Aliment Pharmacol Ther 2022; 55:944-959. [PMID: 35266164 PMCID: PMC9310586 DOI: 10.1111/apt.16794] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/13/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is characterised by hepatic lipid accumulation, cell injury, inflammation and fibrosis. Insulin resistance, a hallmark of type 2 diabetes (T2D) and obesity, is a key pathogenic driver of NASH. Other than difficult-to-maintain lifestyle changes, there are no approved treatments for NASH. Due to their effects on multiple pathophysiological processes, glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been tested in disorders related to insulin resistance and metabolic defects. AIMS To summarise studies of GLP-1RAs relevant to the treatment of NASH. METHODS PubMed searches were performed and results were compiled. RESULTS Large trials with GLP-1RAs in T2D demonstrate highly effective glucose lowering, with body weight loss, and in some cases, reduced cardiovascular events and improved liver transaminases. The GLP-1RAs, liraglutide and semaglutide, were associated with clinically relevant, sustained body weight reduction in individuals with overweight or obesity and without T2D. In a phase II trial in NASH, liraglutide reduced metabolic dysfunction, insulin resistance and lipotoxicity in key organs associated with NASH pathogenesis. Furthermore, liraglutide and semaglutide led to histological resolution of NASH in ~40% to 60% of patients, although a statistically significant effect on fibrosis has not been confirmed. Regarding safety, GLP-1RAs are associated with gastrointestinal and gallbladder-related adverse events, with the latter perhaps related to weight loss. Meta-analyses do not indicate increased risk of acute pancreatitis, pancreatic cancer or other malignancies with GLP-1RAs. CONCLUSIONS These studies support the use of GLP-1RAs for the improvement of underlying metabolic dysfunction observed in NASH and suggest further long-term phase III trials are warranted.
Collapse
Affiliation(s)
- A. Sidney Barritt
- Division of Gastroenterology and Hepatology, UNC Liver CenterUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | | | - Mazen Noureddin
- Department of MedicineCedars‐Sinai Medical CenterLos AngelesCAUSA
| |
Collapse
|
130
|
Cimifugin Ameliorates Lipotoxicity-Induced Hepatocyte Damage and Steatosis through TLR4/p38 MAPK- and SIRT1-Involved Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4557532. [PMID: 35355867 PMCID: PMC8958062 DOI: 10.1155/2022/4557532] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 12/21/2022]
Abstract
Objective. Hepatic metabolic disorder induced by lipotoxicity plays a detrimental role in metabolic fatty liver disease pathogenesis. Cimifugin (Cim), a coumarin derivative extracted from the root of Saposhnikovia divaricata, possesses multiple biological properties against inflammation, allergy, and oxidative stress. However, limited study has addressed the hepatoprotective role of Cim. Here, we investigate the protective effect of Cim against lipotoxicity-induced cytotoxicity and steatosis in hepatocytes and clarify its potential mechanisms. Methods. AML-12, a nontransformed mouse hepatocyte cell line, was employed in this study. The cells were incubated with palmitate or oleate to imitate hepatotoxicity or steatosis model, respectively. Results. Cim significantly reversed palmitate-induced hepatocellular injury in a dose-dependent manner, accompanied by improvements in oxidative stress and mitochondrial damage. Cim pretreatment reversed palmitate-stimulated TLR4/p38 MAPK activation and SIRT1 reduction without affecting JNK, ERK1/2, and AMPK pathways. The hepatoprotective effects of Cim were abolished either through activating TLR4/p38 by their pharmacological agonists or genetical silencing SIRT1 via special siRNA, indicating a mechanistic involvement. Moreover, Cim treatment improved oleate-induced hepatocellular lipid accumulation, which could be blocked by either TLR4 stimulation or SIRT1 knockdown. We observed that SIRT1 was a potential target of TLR4 in palmitate-treated hepatocytes, since TLR4 agonist LPS aggravated, whereas TLR4 antagonist CLI-095 alleviated palmitate-decreased SIRT1 expression. SIRT1 knockdown did not affect palmitate-induced TLR4. In addition, TLR4 activation by LPS significantly abolished Cim-protected SIRT1 reduction induced by palmitate. These results collaboratively indicated that TLR4-regulated SIRT1 pathways was mechanistically involved in the protective effects of Cim against lipotoxicity. Conclusion. In brief, we demonstrate the protective effects of Cim against lipotoxicity-induced cell death and steatosis in hepatocytes. TLR4-regulated p38 MAPK and SIRT1 pathways are involved in Cim-protected hepatic lipotoxicity. Cim is a potential candidate for improving hepatic metabolic disorders mediated by lipotoxicity.
Collapse
|
131
|
Balbi B, Benini F, Corda L, Corsico A, Ferrarotti I, Gatta N. An Italian expert consensus on the management of alpha1-antitrypsin deficiency: a comprehensive set of algorithms. Panminerva Med 2022; 64:215-227. [PMID: 35146988 DOI: 10.23736/s0031-0808.22.04592-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Alpha1-antitrypin deficiency (AATD) is a genetic-based risk condition, mainly affecting the lungs and liver. Despite its wide distribution, it is largely underdiagnosed, thus being considered a rare disease, and is consequently managed in ad hoc reference centers. Unfortunately, an easy-to-use algorithm for managing such a complex disease is still lacking. METHODS An expert consensus meeting was conducted among experts in the management of AATD to build a comprehensive algorithm, including diagnosis, monitoring, AAT therapy, rehabilitation and lung transplantation, and liver disease, that could serve as a guide for physicians and treating centers. A panel of AATD specialists evaluated the results of their work. RESULTS Diagnosis is the most delicate phase, and awareness about this condition should be raised among GPs. A set of recommendations has been written about the most suitable follow-up visits. Augmentation therapy with AAT may be useful to reduce the progression of emphysema and lung function decline in selected patients. Exercise capacity may be improved by pulmonary rehabilitation and, in selected cases, by lung volume reduction or lung transplantation. Support therapies are needed for those who develop liver disease, and, in selected cases, liver transplantation may be considered. Patients should be carefully educated about their lifestyle, including smoking cessation, body weight control, and reduced alcohol intake. CONCLUSIONS The proposed algorithm obtained the endorsement of the Italian Society of Pneumology (SIP). However, further studies and additional clinical data are required to confirm the validity of these recommendations.
Collapse
Affiliation(s)
- Bruno Balbi
- Pulmonary Rehabilitation of the Institute of Veruno, Istituti Clinici Scientifici Maugeri IRCCS, Novara, Italy -
| | - Federica Benini
- Center for diagnosis, monitoring and therapy of alpha1-antitrypsin deficiency, Gastroenterology Unit, Department of Medicine, Spedali Civili, Brescia, Italy
| | - Luciano Corda
- Center for diagnosis, monitoring and therapy of alpha1-antitrypsin deficiency. Respiratory, Medicine Unit, Department of Internal Medicine, Spedali Civili, Brescia, Italy
| | - Angelo Corsico
- Center for diagnosis of alpha1-antitrypsin hereditary deficiency, Chest Medicine Unit. I.R.C.C.S. Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Ilaria Ferrarotti
- Center for diagnosis of alpha1-antitrypsin hereditary deficiency, Chest Medicine Unit. I.R.C.C.S. Policlinico San Matteo Foundation, University of Pavia, Pavia, Italy
| | - Nuccia Gatta
- Patients' association Associazione Nazionale Alfa1-At per la tutela dei pazienti con Deficit di Alfa1-antitripsina, Sarezzo, Brescia, Italy
| | | |
Collapse
|
132
|
Sebastiani G, Patel K, Ratziu V, Feld JJ, Neuschwander-Tetri BA, Pinzani M, Petta S, Berzigotti A, Metrakos P, Shoukry N, Brunt EM, Tang A, Cobbold JF, Ekoe JM, Seto K, Ghali P, Chevalier S, Anstee QM, Watson H, Bajaj H, Stone J, Swain MG, Ramji A. Current considerations for clinical management and care of non-alcoholic fatty liver disease: Insights from the 1st International Workshop of the Canadian NASH Network (CanNASH). CANADIAN LIVER JOURNAL 2022; 5:61-90. [PMID: 35990786 PMCID: PMC9231423 DOI: 10.3138/canlivj-2021-0030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 08/30/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects approximately 8 million Canadians. NAFLD refers to a disease spectrum ranging from bland steatosis to non-alcoholic steatohepatitis (NASH). Nearly 25% of patients with NAFLD develop NASH, which can progress to liver cirrhosis and related end-stage complications. Type 2 diabetes and obesity represent the main risk factors for the disease. The Canadian NASH Network is a national collaborative organization of health care professionals and researchers with a primary interest in enhancing understanding, care, education, and research around NAFLD, with a vision of best practices for this disease state. At the 1st International Workshop of the CanNASH network in April 2021, a joint event with the single topic conference of the Canadian Association for the Study of the Liver (CASL), clinicians, epidemiologists, basic scientists, and community members came together to share their work under the theme of NASH. This symposium also marked the initiation of collaborations between Canadian and other key opinion leaders in the field representative of international liver associations. The main objective is to develop a policy framework that outlines specific targets, suggested activities, and evidence-based best practices to guide provincial, territorial, and federal organizations in developing multidisciplinary models of care and strategies to address this epidemic.
Collapse
Affiliation(s)
- Giada Sebastiani
- Division of Gastroenterology and Hepatology, Royal Victoria Hospital, McGill University Health Centre, Montréal, Québec, Canada
| | - Keyur Patel
- Toronto Centre for Liver Disease, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Vlad Ratziu
- Assistance Publique-Hôpitaux de Paris, Hôpital Pitié Salpêtrière, Sorbonne University, Paris, France
| | - Jordan J Feld
- Toronto Centre for Liver Disease, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine - Royal Free Hospital, London, UK
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, PROMISE, Università di Palermo, Palermo, Italy
| | - Annalisa Berzigotti
- Hepatology, University Clinic for Visceral Surgery and Medicine, Inselspital, University Hospital of Bern, University of Bern, Switzerland
| | - Peter Metrakos
- Cancer Research Program, McGill University Health Centre Research Institute, Montréal, Québec, Canada
| | - Naglaa Shoukry
- Centre de Recherche du Centre hospitalier de l'Université de Montréal, Montréal, Québec, Canada
| | | | - An Tang
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Québec, Canada
| | - Jeremy F Cobbold
- Department of Gastroenterology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Jean-Marie Ekoe
- Montreal Institute for Clinical Research, Division of Endocrinology and Department of Nutrition, Université de Montréal, Montréal, Québec, Canada
| | - Karen Seto
- Canadian Liver Foundation, Markham, Ontario, Canada
| | - Peter Ghali
- University of Florida, Gainesville, Florida, USA
| | | | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Harpreet Bajaj
- LMC Diabetes and Endocrinology, Brampton, Ontario, Canada
| | - James Stone
- Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Mark G Swain
- Calgary Liver Unit, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Alnoor Ramji
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
133
|
Jin M, Zheng L, Wei Y, Cheng J, Zhang D, Yan S, Qin H, Wang Q, Ci X, Feng H. Enterobacter cloacae aggravates metabolic disease by inducing inflammation and lipid accumulation. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 90:103819. [PMID: 35077907 DOI: 10.1016/j.etap.2022.103819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 01/03/2022] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
It is well known that gut microbiota imbalance can promote the development of metabolic disease. Enterobacter cloacae (E. cloacae) is a kind of opportunistic pathogen in the intestine. Therefore, we hypothesized that E. cloacae accelerated the development of metabolic disease. To answer this question, we used E. cloacae to induce disease in guinea pigs. We used H&E staining to detect the pathological changes of liver and aorta and used Oil Red O staining to evaluate the lipid accumulation in the liver. And that we used a kit to detect AST content and used Western blot to detect protein levels in the liver. We found that E. cloacae could induce liver pathological changes and lipid accumulation as well as aortic wall pathological changes in guinea pigs. And E. cloacae increased the liver index to 5.94% and the serum AST level to 41.93 U/L. Importantly, E. cloacae activated liver high mobility group protein (HMGB1)/toll-like receptor 4 (TLR4)/myeloiddifferentiationfactor88 (MYD88)/nuclear factor-kappa B (NF-κB) signal and sterol regulatory element-binding protein 1c (SREBP-1c) and inhibited AMP-activated protein kinase (AMPK). We conclude that E. cloacae promote nonalcoholic fatty liver disease (NAFLD) by inducing inflammation and lipid accumulation, and E. cloacae also promote atherosclerosis. These findings are important for study on the pathogenesis and drug screening of NAFLD and atherosclerosis.
Collapse
Affiliation(s)
- Meiyu Jin
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Lianwen Zheng
- Reproductive Medical Center, the Second Hospital of Jilin University, Changchun, PR China
| | - Yunfei Wei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Jiaqi Cheng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Di Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Siru Yan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Haiyan Qin
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Qi Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun 130001, PR China
| | - Haihua Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, PR China.
| |
Collapse
|
134
|
Bustamante-Bernal MA, Chavez LO, Zuckerman MJ. Endoscopic Bariatric Interventions in Patients with Chronic Liver Disease. Clin Liver Dis 2022; 26:139-148. [PMID: 34802660 DOI: 10.1016/j.cld.2021.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity and its associated comorbidities are rapidly increasing in the US population. Therefore, metabolic associated fatty liver disease (MAFLD), previously known as nonalcoholic fatty liver disease (NAFLD), has become a leading indication for liver transplantation. Lifestyle modifications as a sole therapy have been insufficient to reduce the burden of chronic liver disease secondary to MAFLD. Endoscopic bariatric interventions (EBI) appear to be safe and effective therapies for obesity and chronic liver disease secondary to MAFLD. Gastric EBI include endoscopic sleeve gastroplasty (ESG) and intragastric balloons (IGB). Small bowel EBI are also evolving in the field of bariatric endoscopy.
Collapse
Affiliation(s)
- Marco A Bustamante-Bernal
- Division of Gastroenterology and Hepatology, Texas Tech University Health Sciences Center, 4800 Alberta Avenue, El Paso, TX 79905, USA.
| | - Luis O Chavez
- Division of Gastroenterology and Hepatology, Texas Tech University Health Sciences Center, 4800 Alberta Avenue, El Paso, TX 79905, USA
| | - Marc J Zuckerman
- Division of Gastroenterology and Hepatology, Texas Tech University Health Sciences Center, 4800 Alberta Avenue, El Paso, TX 79905, USA
| |
Collapse
|
135
|
Handelsman Y, Anderson JE, Bakris GL, Ballantyne CM, Beckman JA, Bhatt DL, Bloomgarden ZT, Bozkurt B, Budoff MJ, Butler J, Dagogo-Jack S, de Boer IH, DeFronzo RA, Eckel RH, Einhorn D, Fonseca VA, Green JB, Grunberger G, Guerin C, Inzucchi SE, Jellinger PS, Kosiborod MN, Kushner P, Lepor N, Mende CW, Michos ED, Plutzky J, Taub PR, Umpierrez GE, Vaduganathan M, Weir MR. DCRM Multispecialty Practice Recommendations for the management of diabetes, cardiorenal, and metabolic diseases. J Diabetes Complications 2022; 36:108101. [PMID: 34922811 PMCID: PMC9803322 DOI: 10.1016/j.jdiacomp.2021.108101] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 11/27/2021] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D), chronic kidney disease (CKD), atherosclerotic cardiovascular disease (ASCVD), and heart failure (HF)-along with their associated risk factors-have overlapping etiologies, and two or more of these conditions frequently occur in the same patient. Many recent cardiovascular outcome trials (CVOTs) have demonstrated the benefits of agents originally developed to control T2D, ASCVD, or CKD risk factors, and these agents have transcended their primary indications to confer benefits across a range of conditions. This evolution in CVOT evidence calls for practice recommendations that are not constrained by a single discipline to help clinicians manage patients with complex conditions involving diabetes, cardiorenal, and/or metabolic (DCRM) diseases. The ultimate goal for these recommendations is to be comprehensive yet succinct and easy to follow by the nonexpert-whether a specialist or a primary care clinician. To meet this need, we formed a volunteer task force comprising leading cardiologists, nephrologists, endocrinologists, and primary care physicians to develop the DCRM Practice Recommendations, a multispecialty consensus on the comprehensive management of the patient with complicated metabolic disease. The task force recommendations are based on strong evidence and incorporate practical guidance that is clinically relevant and simple to implement, with the aim of improving outcomes in patients with DCRM. The recommendations are presented as 18 separate graphics covering lifestyle therapy, patient self-management education, technology for DCRM management, prediabetes, cognitive dysfunction, vaccinations, clinical tests, lipids, hypertension, anticoagulation and antiplatelet therapy, antihyperglycemic therapy, hypoglycemia, nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH), ASCVD, HF, CKD, and comorbid HF and CKD, as well as a graphical summary of medications used for DCRM.
Collapse
Affiliation(s)
| | | | | | | | | | - Deepak L Bhatt
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Javed Butler
- University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | | - Robert H Eckel
- University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Daniel Einhorn
- Scripps Whittier Institute for Diabetes, San Diego, CA, USA
| | | | | | - George Grunberger
- Grunberger Diabetes Institute, Bloomfield Hills, MI, USA, Wayne State University School of Medicine, Detroit, MI, USA, Oakland University William Beaumont School of Medicine, Rochester, MI, USA, Charles University, Prague, Czech Republic
| | - Chris Guerin
- University of California San Diego School of Medicine, San Diego, CA, USA
| | | | - Paul S Jellinger
- The Center for Diabetes & Endocrine Care, University of Miami Miller School of Medicine, Hollywood, FL, USA
| | - Mikhail N Kosiborod
- Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, MO, USA
| | | | - Norman Lepor
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Christian W Mende
- University of California San Diego School of Medicine, San Diego, CA, USA
| | - Erin D Michos
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jorge Plutzky
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pam R Taub
- University of California San Diego School of Medicine, San Diego, CA, USA
| | | | | | - Matthew R Weir
- University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
136
|
The multifaceted role of ferroptosis in liver disease. Cell Death Differ 2022; 29:467-480. [PMID: 35075250 PMCID: PMC8901678 DOI: 10.1038/s41418-022-00941-0] [Citation(s) in RCA: 269] [Impact Index Per Article: 134.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of non-apoptotic cell death characterized by excessive lipid peroxidation and associated with a plethora of pathological conditions in the liver. Emerging evidence supports the notion that dysregulated metabolic pathways and impaired iron homeostasis play a role in the progression of liver disease via ferroptosis. Although the molecular mechanisms by which ferroptosis causes disease are poorly understood, several ferroptosis-associated genes and pathways have been implicated in liver disease. Here, we review the physiological role of the liver in processing nutrients, our current understanding of iron metabolism, the characteristics of ferroptosis, and the mechanisms that regulate ferroptosis. In addition, we summarize the role of ferroptosis in the pathogenesis of liver disease, including liver injury, non-alcoholic steatohepatitis, liver fibrosis, liver cirrhosis, and hepatocellular carcinoma. Finally, we discuss the therapeutic potential of targeting ferroptosis for managing liver disease.
Collapse
|
137
|
Wooten JS, Poole KE, Harris MP, Guilford BL, Schaller ML, Umbaugh D, Seija A. The effects of voluntary wheel running during weight-loss on biomarkers of hepatic lipid metabolism and inflammation in C57Bl/6J mice. Curr Res Physiol 2022; 5:63-72. [PMID: 35141529 PMCID: PMC8814598 DOI: 10.1016/j.crphys.2022.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/23/2021] [Accepted: 01/18/2022] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to determine the effect of voluntary wheel running (VWR) during weight-loss on hepatic lipid and inflammatory biomarkers using a murine model. To induce obesity, male C57Bl/6 mice were fed a 60% high-fat diet (HF) for 10 weeks. At 10 weeks, weight-loss was promoted by randomizing HF-fed mice to a normal diet (ND) either with (WL + VWR) or without (WL) access to running wheels for 8 weeks. Age-matched dietary control mice were fed either a ND or HF for 18 weeks. Following weight-loss, WL + VWR had a lower body mass compared to all groups despite an average weekly caloric consumption comparable to HF mice. WL + VWR had an increased adiponectin concentration when compared to WL, but no difference between WL and WL + VWR was observed for plasma glucose and lipid biomarkers. When compared to HF, the lower hepatic total lipids in both WL and WL + VWR were associated with increased pAMPK:AMPK and reduced pACC-1:ACC-1 ratios. When compared to WL, WL + VWR resulted in lower hepatic cholesterol and trended to lower hepatic triglyceride. In both WL and WL + VWR, pNF-κB p65:NF-κB p65 ratio was lower than HF and comparable to ND. TGFβ1 and BAMBI protein levels were evaluated as biomarkers for hepatic fibrosis. No differences in TGFβ1 was observed between groups; however, WL and WL + VWR had BAMBI protein levels comparable to ND. Overall, the addition of voluntary exercise resulted in greater weight-loss and improvements in hepatic cholesterol and triglyceride levels; however, limited improvements in hepatic inflammation were observed when compared to weight-loss by diet alone.
Collapse
Affiliation(s)
- Joshua S. Wooten
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Kaylee E. Poole
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Matthew P. Harris
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Brianne L. Guilford
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Megan L. Schaller
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - David Umbaugh
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Andrew Seija
- Department of Applied Health, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
138
|
Kim J, Mondaca-Ruff D, Singh S, Wang Y. SIRT1 and Autophagy: Implications in Endocrine Disorders. Front Endocrinol (Lausanne) 2022; 13:930919. [PMID: 35909524 PMCID: PMC9331929 DOI: 10.3389/fendo.2022.930919] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/20/2022] [Indexed: 11/26/2022] Open
Abstract
Autophagy is a cellular process involved in the selective degradation and recycling of dysfunctional intracellular components. It plays a crucial role in maintaining cellular homeostasis and survival by removing damaged and harmful proteins, lipids, and organelles. SIRT1, an NAD+-dependent multifunctional enzyme, is a key regulator of the autophagy process. Through its deacetylase activity, SIRT1 participates in the regulation of different steps of autophagy, from initiation to degradation. The levels and function of SIRT1 are also regulated by the autophagy process. Dysregulation in SIRT1-mediated autophagy hinders the proper functioning of the endocrine system, contributing to the onset and progression of endocrine disorders. This review provides an overview of the crosstalk between SIRT1 and autophagy and their implications in obesity, type-2 diabetes mellitus, diabetic cardiomyopathy, and hepatic steatosis.
Collapse
|
139
|
Mutamba AK, He X, Wang T. Therapeutic advances in overcoming intrauterine growth restriction induced metabolic syndrome. Front Pediatr 2022; 10:1040742. [PMID: 36714657 PMCID: PMC9875160 DOI: 10.3389/fped.2022.1040742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Intrauterine growth restriction (IUGR) remains a great public health challenge as it affects neonatal survival and influences their normal biological development and metabolism. Several clinical researches have revealed the occurrence of metabolic syndrome, such as insulin resistance, obesity, type 2 diabetes mellitus, oxidative stress, dyslipidemia, as direct results of IUGR. Therefore, it is essential to understand its underlying mechanism, impact and develop effective therapies. The purpose of this work is to review the current knowledge on IUGR induced metabolic syndrome and relevant therapies. Here in, we elaborate on the characteristics and causes of IUGR by pointing out recent research findings. Furthermore, we discuss the impact of IUGR on different organs of the body, followed by preclinical studies on IUGR using suitable animal models. Additionally, various metabolic disorders with their genetic implications, such as insulin resistance, type 2 diabetes mellitus, dyslipidemia, obesity are detailed. Finally, the current therapeutic options used in the treatment of IUGR are summarized with some prospective therapies highlighted.
Collapse
Affiliation(s)
- Alpha Kalonda Mutamba
- Department of Pediatrics, Neonatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaori He
- Department of Pediatrics, Neonatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Tao Wang
- Laboratory of Neonatal Disease, Institute of Pediatrics, Central South University, Changsha, China
| |
Collapse
|
140
|
Gluais‐Dagorn P, Foretz M, Steinberg GR, Batchuluun B, Zawistowska‐Deniziak A, Lambooij JM, Guigas B, Carling D, Monternier P, Moller DE, Bolze S, Hallakou‐Bozec S. Direct AMPK Activation Corrects NASH in Rodents Through Metabolic Effects and Direct Action on Inflammation and Fibrogenesis. Hepatol Commun 2022; 6:101-119. [PMID: 34494384 PMCID: PMC8710801 DOI: 10.1002/hep4.1799] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
No approved therapies are available for nonalcoholic steatohepatitis (NASH). Adenosine monophosphate-activated protein kinase (AMPK) is a central regulator of cell metabolism; its activation has been suggested as a therapeutic approach to NASH. Here we aimed to fully characterize the potential for direct AMPK activation in preclinical models and to determine mechanisms that could contribute to efficacy for this disease. A novel small-molecule direct AMPK activator, PXL770, was used. Enzyme activity was measured with recombinant complexes. De novo lipogenesis (DNL) was quantitated in vivo and in mouse and human primary hepatocytes. Metabolic efficacy was assessed in ob/ob and high-fat diet-fed mice. Liver histology, biochemical measures, and immune cell profiling were assessed in diet-induced NASH mice. Direct effects on inflammation and fibrogenesis were assessed using primary mouse and human hepatic stellate cells, mouse adipose tissue explants, and human immune cells. PXL770 directly activated AMPK in vitro and reduced DNL in primary hepatocytes. In rodent models with metabolic syndrome, PXL770 improved glycemia, dyslipidemia, and insulin resistance. In mice with NASH, PXL770 reduced hepatic steatosis, ballooning, inflammation, and fibrogenesis. PXL770 exhibited direct inhibitory effects on pro-inflammatory cytokine production and activation of primary hepatic stellate cells. Conclusion: In rodent models, direct activation of AMPK is sufficient to produce improvements in all core components of NASH and to ameliorate related hyperglycemia, dyslipidemia, and systemic inflammation. Novel properties of direct AMPK activation were also unveiled: improved insulin resistance and direct suppression of inflammation and fibrogenesis. Given effects also documented in human cells (reduced DNL, suppression of inflammation and stellate cell activation), these studies support the potential for direct AMPK activation to effectively treat patients with NASH.
Collapse
Affiliation(s)
| | - Marc Foretz
- Université de ParisInstitut CochinCNRSINSERMParisFrance
| | - Gregory R. Steinberg
- Centre for Metabolism, Obesity and Diabetes Research and Division of Endocrinology and MetabolismDepartment of MedicineMcMaster UniversityHamiltonONCanada
| | - Battsetseg Batchuluun
- Centre for Metabolism, Obesity and Diabetes Research and Division of Endocrinology and MetabolismDepartment of MedicineMcMaster UniversityHamiltonONCanada
| | | | - Joost M. Lambooij
- Department of ParasitologyLeiden University Medical CenterLeidenthe Netherlands
| | - Bruno Guigas
- Department of ParasitologyLeiden University Medical CenterLeidenthe Netherlands
| | - David Carling
- Cellular Stress GroupMedical Research CouncilLondon Institute of Medical SciencesHammersmith HospitalImperial CollegeLondonUnited Kingdom
| | | | | | | | | |
Collapse
|
141
|
Yang Y, Li S, Xu Y, Ke J, Zhao D. The Perirenal Fat Thickness Was Associated with Nonalcoholic Fatty Liver Disease in Patients with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2022; 15:1505-1515. [PMID: 35586202 PMCID: PMC9109981 DOI: 10.2147/dmso.s350579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 04/27/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Obesity is an important risk factor for nonalcoholic fatty liver disease (NAFLD). Perirenal fat and paranephric fat were seldom studied in NAFLD. We aimed to explore the relationship between perirenal fat thickness (PrFT) and paranephric fat thickness (PnFT) and NAFLD in patients with type 2 diabetes mellitus (T2DM). PATIENTS AND METHODS A total of 493 diabetic patients including 231 NAFLD patients were enrolled in our study from September 2019 to December 2020. Patients with NAFLD were categorized into three subgroups according to the severity and fibrosis risk of NAFLD. Anthropometric indices and clinical characteristics were collected from clinical records. PrFT and PnFT were measured via ultrasound. Multivariate logistic regression analysis was used to assess the association between PrFT, PnFT and presence, severity and advanced fibrosis risk of NAFLD. RESULTS Compared with non-NAFLD patients, those with NAFLD had significantly higher PrFT and PnFT. The PrFT and PnFT were independently associated with presence of NAFLD and the PrFT was independently associated with the advanced fibrosis risk of NAFLD after adjusting confounding factors. CONCLUSION The PrFT was independently associated with the presence and advanced fibrosis risk of NAFLD in patients with T2DM.
Collapse
Affiliation(s)
- Yuxian Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Shuting Li
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yuechao Xu
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Ke
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Jing Ke; Dong Zhao, Email ;
| | - Dong Zhao
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
142
|
Ma W, Wu W, Wen W, Xu F, Han D, Lyu J, Huang Y. Association of NAFLD with cardiovascular disease and all-cause mortality: a large-scale prospective cohort study based on UK Biobank. Ther Adv Chronic Dis 2022; 13:20406223221122478. [PMID: 36159632 PMCID: PMC9493675 DOI: 10.1177/20406223221122478] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Nonalcoholic fatty liver disease (NAFLD) is considered as the hepatic manifestation of metabolic syndrome, sharing the similar cardiometabolic risk factors with cardiovascular disease (CVD). Whether NAFLD by itself is associated with increased cardiovascular events and death remain an issue to debate. This study aimed to further investigate the association between NAFLD and adverse CVD outcomes. Methods Participants were followed up until the end of 2020 in current analysis. NAFLD is defined using fatty liver index (FLI). Cox proportional hazard model was used to analyze the association between NAFLD and all-cause mortality, major adverse cardiovascular events (MACEs), CVD mortality, fatal/nonfatal acute myocardial infarction (AMI), and fatal/nonfatal stroke. C-index was calculated to evaluate the model enhancement when adding NAFLD factor. Results After screening the data of 502,492 participants in the original cohort, 215,245 eligible participants were included in this study for MACEs outcome. Compared with non-NAFLD participants, the multivariable adjusted hazard ratios of NAFLD group was 1.25 (1.14-1.36) for MACEs; 1.14 (1.08-1.20) for all-cause mortality; 1.61(1.42-1.82) for CVD mortality; 1.58(1.19-2.11) for AMI mortality; and 1.18 (0.85-1.64) for stroke mortality. When adding FLI, C-index of NAFLD model improved for all-cause mortality, MACEs, and CVD mortality compared with that in the traditional CVD risk factor model. Conclusion NAFLD is an independent risk factor for all-cause mortality and adverse CVD outcomes. Based on the traditional CVD risk factor model, additionally screening NAFLD could improve the prediction efficiency for adverse CVD outcomes.
Collapse
Affiliation(s)
- Wen Ma
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, P.R. China.,Department of Clinical Research, The First Affiliated Hospital, Jinan University, Guangzhou, P.R. China.,School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Wentao Wu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, P.R. China
| | - Fengshuo Xu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Didi Han
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital, Jinan University, Guangzhou 510630, P.R. China.,Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization (2021B1212040007), Guangzhou, P.R. China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, Jiazi Road, Lunjiao, Shunde, Foshan 528300, P.R. China.,The George Institute for Global Health, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| |
Collapse
|
143
|
Sex differences in white adipose tissue expansion: emerging molecular mechanisms. Clin Sci (Lond) 2021; 135:2691-2708. [PMID: 34908104 DOI: 10.1042/cs20210086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/15/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022]
Abstract
The escalating prevalence of individuals becoming overweight and obese is a rapidly rising global health problem, placing an enormous burden on health and economic systems worldwide. Whilst obesity has well described lifestyle drivers, there is also a significant and poorly understood component that is regulated by genetics. Furthermore, there is clear evidence for sexual dimorphism in obesity, where overall risk, degree, subtype and potential complications arising from obesity all differ between males and females. The molecular mechanisms that dictate these sex differences remain mostly uncharacterised. Many studies have demonstrated that this dimorphism is unable to be solely explained by changes in hormones and their nuclear receptors alone, and instead manifests from coordinated and highly regulated gene networks, both during development and throughout life. As we acquire more knowledge in this area from approaches such as large-scale genomic association studies, the more we appreciate the true complexity and heterogeneity of obesity. Nevertheless, over the past two decades, researchers have made enormous progress in this field, and some consistent and robust mechanisms continue to be established. In this review, we will discuss some of the proposed mechanisms underlying sexual dimorphism in obesity, and discuss some of the key regulators that influence this phenomenon.
Collapse
|
144
|
Fouqueray P, Bolze S, Dubourg J, Hallakou-Bozec S, Theurey P, Grouin JM, Chevalier C, Gluais-Dagorn P, Moller DE, Cusi K. Pharmacodynamic effects of direct AMP kinase activation in humans with insulin resistance and non-alcoholic fatty liver disease: A phase 1b study. Cell Rep Med 2021; 2:100474. [PMID: 35028615 PMCID: PMC8714938 DOI: 10.1016/j.xcrm.2021.100474] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/14/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
AMPK is an energy sensor modulating metabolism, inflammation, and a target for metabolic disorders. Metabolic dysfunction results in lower AMPK activity. PXL770 is a direct AMPK activator, inhibiting de novo lipogenesis (DNL) and producing efficacy in preclinical models. We aimed to assess pharmacokinetics, safety, and pharmacodynamics of PXL770 in humans with metabolic syndrome-associated fatty liver disease. In a randomized, double-blind four-week trial, 12 overweight/obese patients with non-alcoholic fatty liver disease (NAFLD) and insulin resistance received PXL770 500 mg QD; 4 subjects received matching placebo. Endpoints included pharmacokinetics, hepatic fractional DNL, oral glucose tolerance testing, additional pharmacodynamic parameters, and safety. PK parameters show adequate plasma exposure in NAFLD patients for daily oral dosing. PXL770 decreases DNL—both peak and AUC are reduced versus baseline—and improves glycemic parameters and indices of insulin sensitivity versus baseline. Assessment of specific lipids reveals decrease in diacyglycerols/triacylglycerols. Safety/tolerability are similar to placebo. These results unveil initial human translation of AMPK activation and support this therapeutic strategy for metabolic disorders. AMPK is impaired in metabolic diseases—PXL770 is a direct AMPK activator In humans with NAFLD, PXL770 inhibits hepatic de novo lipogenesis (target engagement) PXL770 also improves glucose tolerance and insulin sensitivity PXL770 reduces plasma levels of di- and triglycerides
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| |
Collapse
|
145
|
Kamolvisit S, Chirnaksorn S, Nimitphong H, Sungkanuparph S. Pioglitazone for the Treatment of Metabolic-Associated Fatty Liver Disease in People Living With HIV and Prediabetes. Cureus 2021; 13:e19046. [PMID: 34858740 PMCID: PMC8613454 DOI: 10.7759/cureus.19046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Background Metabolic-associated fatty liver disease (MAFLD) is increasingly common among people living with the human immunodeficiency virus (PLHIV) and can progress to cirrhosis and cirrhotic-related complications. Pioglitazone is known to improve insulin sensitivity that results in decreasing serum fatty acids and resolution of non-alcoholic steatohepatitis. This study was aimed to evaluate the efficacy of pioglitazone for the treatment of MAFLD in PLHIV and prediabetes. Methods A randomized controlled trial was conducted in HIV-positive individuals with prediabetes who had evidence of a fatty liver by abdominal ultrasonography or controlled attenuation parameter (CAP) ≥ 238 decibels per meter (dB/m) through using transient elastography. Participants were randomized to take pioglitazone, 30 mg/day, (pioglitazone group) or placebo (control group) and were followed up and assessed for 48 weeks. Results A total of 98 participants were enrolled, 49 in each group. The mean age was 50.8 years and 66.3% were males. All participants had received antiretroviral therapy with undetectable HIV ribonucleic acid (RNA) and the mean CD4 cell count was 463.2 cells/mm3. The mean baseline CAP and liver stiffness were 285.7 dB/m and 5.4 kilopascals (kPa), respectively. At 24 weeks, the mean change of the CAP level was -25.7 dB/m in the pioglitazone group and -5.6 dB/m in the control group (p = 0.040); the mean change of liver stiffness was 0.014 kPa in the pioglitazone group and 0.403 kPa in the control group (p = 0.199). At 48 weeks, the mean change of the CAP level was -23.5 dB/m in the pioglitazone group and 10.2 dB/m in the control group (p < 0.001); the mean change of liver stiffness was -0.184 kPa in the pioglitazone group and 0.554 kPa in the control group (p = 0.016). The mean changes of fasting plasma glucose (FPG) at 24 and 48 weeks were -14.9 and -17.5 mg/dL in the pioglitazone group, respectively, and -3.6 and 4.5 mg/dL in the control group, respectively (p < 0.05). The mean change of the body mass index, lipid profiles, and liver enzymes were not different between the two groups at both time points (p > 0.05). No serious adverse effects were observed in either group. Conclusions Pioglitazone significantly reduces CAP, liver stiffness, and FPG in PLHIV with prediabetes and MAFLD. Further studies with long-term follow-up duration are warranted to determine the role of pioglitazone for clinical use in this population.
Collapse
Affiliation(s)
- Sarunporn Kamolvisit
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, THA
| | - Supphamat Chirnaksorn
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, THA
| | - Hataikarn Nimitphong
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, THA
| | - Somnuek Sungkanuparph
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, THA
| |
Collapse
|
146
|
Yu X, Chen C, Guo Y, Tong Y, Zhao Y, Wu L, Sun X, Wu X, Song Z. High NAFLD fibrosis score in non-alcoholic fatty liver disease as a predictor of carotid plaque development: a retrospective cohort study based on regular health check-up data in China. Ann Med 2021; 53:1621-1631. [PMID: 34498502 PMCID: PMC8439219 DOI: 10.1080/07853890.2021.1974081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/23/2021] [Indexed: 10/31/2022] Open
Abstract
PURPOSES There is increasing concern regarding cardiovascular risk in non-alcoholic fatty liver disease (NAFLD) patients with liver fibrosis. This study aims: (1) to assess the association between NAFLD and liver fibrosis status and the development of carotid plaque (CP), and (2) to identify CP risk factors among general population with different baseline NAFLD and liver fibrosis status. METHODS This retrospective cohort study included 14,288 adult participants who went for regular health check-ups between 2014 and 2019, in one hospital in Zhejiang, China. NAFLD was diagnosed by abdominal ultrasound and the NAFLD fibrosis score (NFS) was calculated to reflect the extent of liver fibrosis. Cox proportional hazards analyses were applied to assess the risk of CP development across groups with different baseline NAFLD and NFS status. RESULTS NAFLD participants with high NFS had higher risk of CP compared to non-NAFLD participants (adjusted hazard ratio 1.68, 95% confidence interval [CI] 1.43-1.96, p < .001). Progression from NAFLD free and NAFLD with low NFS to NAFLD with high NFS are associated with 1.56-fold (95% CI 1.21-2.01, p = .001) and 1.43-fold (95% CI 1.11-1.84, p = .006) increased risk of CP, respectively. Risk factors associated with CP vary based on baseline NAFLD and NFS status. Among NAFLD participants with high NFS, hypertension is the only significant risk factor after adjustment for other potential influencing factors. CONCLUSIONS NAFLD and liver fibrosis status can be an independent predictor for CP development regardless of metabolic abnormalities. Hypertension is a major risk factor for CP development among NAFLD patients with high NFS.KEY MESSAGESNon-alcoholic fatty liver disease (NAFLD) and liver fibrosis status can be an independent predictor for development of carotid plaque.Progression from NAFLD free and NAFLD with low NAFLD fibrosis score (NFS) to NAFLD with high NFS are associated with increased risk of carotid plaque.Risk factors associated with carotid plaque vary based on baseline NAFLD and NFS status, and hypertension plays the most important role among patients with NAFLD and high NFS.
Collapse
Affiliation(s)
- Xinyan Yu
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chen Chen
- Department of Big Data in Health Science, School of Public Health, Zhejiang University, Hangzhou, China
- Center for Biostatistics, Bioinformatics, and Big Data, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Guo
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuling Tong
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zhao
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingyan Wu
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xue Sun
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xifeng Wu
- Department of Big Data in Health Science, School of Public Health, Zhejiang University, Hangzhou, China
- Center for Biostatistics, Bioinformatics, and Big Data, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhenya Song
- Department of General Practice and Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
147
|
Han T, Fan Y, Gao J, Fatima M, Zhang Y, Ding Y, Bai L, Wang C. Sodium glucose cotransporter 2 inhibitor dapagliflozin depressed adiposity and ameliorated hepatic steatosis in high-fat diet induced obese mice. Adipocyte 2021; 10:446-455. [PMID: 34550043 PMCID: PMC8475578 DOI: 10.1080/21623945.2021.1979277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
With the increasing obesity prevalence, the rates of obesity-related diseases, including type 2 diabetes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases, have increased dramatically. Dapagliflozin, one of the sodium glucose cotransporter inhibitors, not only exerts hypoglycaemic effects through increasing urinary glucose excretion but alsoreprograms the metabolic system, leading to benefits in metabolic and cardiovascular diseases. In this study, pre-established obese mice on a high-fat diet were given dapagliflozin by gavage for fourweeks. It showed that dapagliflozin can enhance fat utilization and browning of adipose tissue and improve local oxidative stress, thus inhibiting fat accumulation and hepatic steatosis without disturbance in body weight or plasma glycolipid level. Overall, our study highlights the potential clinical application of SGLT2 inhibition in the prevention of obesity and related metabolic diseases, such as insulin resistance, NAFLD, and diabetes.
Collapse
Affiliation(s)
- Tuo Han
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, China
| | - Yajie Fan
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jie Gao
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, China
| | - Mahreen Fatima
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, China
| | - Yali Zhang
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, China
| | - Yiming Ding
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, China
| | - Liang Bai
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Centre, Xi’an, Shaanxi, China
| | - Congxia Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
148
|
Král J, Machytka E, Horká V, Selucká J, Doleček F, Špičák J, Kovářová V, Haluzík M, Bužga M. Endoscopic Treatment of Obesity and Nutritional Aspects of Bariatric Endoscopy. Nutrients 2021; 13:nu13124268. [PMID: 34959819 PMCID: PMC8703836 DOI: 10.3390/nu13124268] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/14/2021] [Accepted: 11/26/2021] [Indexed: 11/25/2022] Open
Abstract
Obesity is a significant problem worldwide. Several serious diseases that decrease patient quality of life and increase mortality (high blood pressure, dyslipidaemia, type 2 diabetes etc.) are associated with obesity. Obesity treatment is a multidisciplinary and complex process that requires maximum patient compliance. Change of lifestyle is fundamental in the treatment of obesity. While pharmacotherapeutic options are available, their efficacy is limited. Surgical treatment though highly effective, carries the risk of complications and is thus indicated mostly in advanced stages of obesity. Endoscopic treatments of obesity are less invasive than surgical options, and are associated with fewer complications and nutritional deficits. Currently, there is a large spectrum of endoscopic methods based on the principles of gastric volume reduction, size restriction and gastric or small bowel bypass being explored with only few available in routine practice. The aim of this publication is to present an up-to-date summary of available endoscopic methods for the treatment of obesity focusing on their efficacy, safety and nutritional aspects.
Collapse
Affiliation(s)
- Jan Král
- Department of Hepatogastroenterology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 14021 Prague, Czech Republic; (E.M.); (J.S.); (J.Š.)
- Correspondence:
| | - Evžen Machytka
- Department of Hepatogastroenterology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 14021 Prague, Czech Republic; (E.M.); (J.S.); (J.Š.)
- Faculty of Medicine and Dentistry, Palacký University Olomouc, 77147 Olomouc, Czech Republic
- Department of Internal Medicine-Gastroenterology and Geriatrics, University Hospital Olomouc, 77900 Olomouc, Czech Republic
| | - Veronika Horká
- Research Obesity Centre, Department of Human Movement Studies, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic;
| | - Jana Selucká
- Department of Hepatogastroenterology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 14021 Prague, Czech Republic; (E.M.); (J.S.); (J.Š.)
| | - Filip Doleček
- Department of Surgical Studies, Faculty of Medicine, University of Ostrava, 70300 Ostrava, Czech Republic;
| | - Julius Špičák
- Department of Hepatogastroenterology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 14021 Prague, Czech Republic; (E.M.); (J.S.); (J.Š.)
| | - Viktorie Kovářová
- Department of Diabetology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 14021 Prague, Czech Republic; (V.K.); (M.H.)
| | - Martin Haluzík
- Department of Diabetology, Institute for Clinical and Experimental Medicine, Vídeňská 1958/9, 14021 Prague, Czech Republic; (V.K.); (M.H.)
| | - Marek Bužga
- Department of Physiology and Pathophysiology, Faculty of Medicine, University of Ostrava, 70103 Ostrava, Czech Republic;
- Institute of Laboratory Medicine, University Hospital Ostrava, 70800 Ostrava, Czech Republic
| |
Collapse
|
149
|
Wang H, Zhang S, Guo J. Lipotoxic Proximal Tubular Injury: A Primary Event in Diabetic Kidney Disease. Front Med (Lausanne) 2021; 8:751529. [PMID: 34760900 PMCID: PMC8573085 DOI: 10.3389/fmed.2021.751529] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/27/2021] [Indexed: 01/23/2023] Open
Abstract
The pathogenesis of diabetic nephropathy is a complex process that has a great relationship with lipotoxicity. Since the concept of “nephrotoxicity” was proposed, many studies have confirmed that lipotoxicity plays a significant role in the progression of diabetic nephropathy and causes various renal dysfunction. This review will make a brief summary of renal injury caused by lipotoxicity that occurs primarily and predominantly in renal tubules during diabetic progression, further leading to glomerular dysfunction. The latest research suggests that lipotoxicity-mediated tubular injury may be a major event in diabetic nephropathy.
Collapse
Affiliation(s)
- Hua Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shu Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Nephrology, Nephropathy Research Institutes of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
150
|
Fengler VH, Macheiner T, Goessler W, Ratzer M, Haybaeck J, Sargsyan K. Hepatic Response of Magnesium-Restricted Wild Type Mice. Metabolites 2021; 11:metabo11110762. [PMID: 34822420 PMCID: PMC8625093 DOI: 10.3390/metabo11110762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/26/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Magnesium-deficiency is implicated in many metabolic disorders, e.g., type 2 diabetes and metabolic syndrome, representing risk factors for non-alcoholic fatty liver disease (NAFLD). This study aims to investigate the contribution of magnesium-restriction to the development of NAFLD. Magnesium-deficiency was induced in C57BL/6 mice by feeding a magnesium-deficient-diet. Metabolic markers as well as markers of inflammation and liver function were assessed. Furthermore, liver tissue was examined histopathologically and compared with specimens from high-fat-diet fed and control mice. Finally, the hepatic inflammatory response was quantified by determining hepatic IL-6, TNFα, and MCP-1. Magnesium-restriction resulted in at least a 2-fold significant reduction of serum magnesium levels compared to the high-fat-diet fed and control mice, whereas the hepatic magnesium content was decreased due to high-fat-diet feeding. No changes in metabolic markers in magnesium-restricted mice were observed, while the cholesterol content was elevated in high-fat-diet fed mice. Magnesium-restricted mice additionally featured inflammation and enlarged hepatocytes in liver histology. Furthermore, magnesium-restricted and high-fat-diet fed mice exhibited elevated hepatic TNFα levels compared to control mice. Accordingly, our data suggest that magnesium is involved in hepatic inflammatory processes and hepatocyte enlargement, key histological features of human NAFLD, and may therefore contribute to development and progression of the disease.
Collapse
Affiliation(s)
- Vera H. Fengler
- Biobank Graz, Medical University of Graz, 8036 Graz, Austria;
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Tanja Macheiner
- International Biobank and Education, Medical University of Graz, 8036 Graz, Austria;
| | - Walter Goessler
- Institute of Chemistry, University of Graz, 8010 Graz, Austria;
| | - Maria Ratzer
- Institute for Biomedicine and Health Sciences, Joanneum Research, 8010 Graz, Austria;
| | - Johannes Haybaeck
- Department of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
- Diagnostic & Research Center for Molecular BioMedicine, Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Karine Sargsyan
- International Biobank and Education, Medical University of Graz, 8036 Graz, Austria;
- Correspondence: ; Tel.: +43-316-385-72718
| |
Collapse
|