1
|
El-Hammadi MM, Martín-Navarro L, Berrocoso E, Álvarez-Fuentes J, Crespo-Facorro B, Suárez-Pereira I, Vázquez-Bourgon J, Martín-Banderas L. Enhanced Metabolic Syndrome Management Through Cannabidiol-Loaded PLGA Nanoparticles: Development and In Vitro Evaluation. J Biomed Mater Res A 2025; 113:e37916. [PMID: 40277882 DOI: 10.1002/jbm.a.37916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025]
Abstract
Cannabidiol (CBD) holds promise for managing metabolic diseases, yet enhancing its oral bioavailability and efficacy remains challenging. To address this, we developed polymeric nanoparticles (NPs), using poly(lactic-co-glycolic acid) (PLGA), encapsulating CBD using nanoprecipitation, aiming to create an effective CBD-nanoformulation for metabolic disorder treatment. These NPs (135-265 nm) demonstrated high encapsulation efficiency (EE% ≈ 100%) and sustained release kinetics. Their therapeutic potential was evaluated in an in vitro metabolic syndrome model employing sodium palmitate-induced HepG2 cells. Key assessment parameters included cell viability (MTT assay), glucose uptake, lipid accumulation (Oil Red O staining), triglycerides, cholesterol, HDL-c levels, and gene expression of metabolic regulators. Results showed an IC50 of 9.85 μg/mL for free CBD and 11.26 μg/mL for CBD-loaded NPs. CBD-loaded NPs significantly enhanced glucose uptake, reduced lipid content, lowered triglycerides and total cholesterol, and increased HDL-c levels compared to free CBD. Gene analysis indicated reduced gluconeogenesis via downregulation of PPARγ, FOXO-1, PEPCK, and G6Pase and enhanced fatty acid oxidation through CPT-1 upregulation. These findings suggest that CBD-loaded NPs may serve as a novel therapeutic strategy for the management of metabolic disorders, warranting further in vivo studies.
Collapse
Affiliation(s)
- Mazen M El-Hammadi
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Lucía Martín-Navarro
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
| | - Esther Berrocoso
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Neuropsychopharmacology & Psychobiology Research Group, Department of Psychology, University of Cadiz, Cádiz, Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
| | - Josefa Álvarez-Fuentes
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Benedicto Crespo-Facorro
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain
- Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio, Sevilla, Spain
| | - Irene Suárez-Pereira
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Biomedical Research and Innovation Institute of Cádiz (INiBICA) Research Unit, Puerta del Mar University Hospital, University of Cádiz, Cádiz, Spain
- Neuropsychopharmacology & Psychobiology Research Group, Department of Neuroscience, University of Cadiz, Cádiz, Spain
| | - Javier Vázquez-Bourgon
- Centro de Investigación Biomédica en Red en Salud Mental (CIBERSAM), Madrid, Spain
- Department of Psychiatry, University Hospital Marqués de Valdecilla. Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
- Departamento de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Spain
| | - Lucía Martín-Banderas
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBIS)-Campus Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
2
|
Ding FF, Li M, Wang T, Zhou NN, Qiao F, Du ZY, Zhang ML. Influence of dietary sodium taurocholate on the growth performance and liver health of Nile tilapia (Oreochromis niloticus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:319-330. [PMID: 36044098 DOI: 10.1007/s10695-022-01116-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/20/2022] [Indexed: 06/15/2023]
Abstract
Bile acids (BAs) are a class of cholesterol-derived amphipathic molecules approved as new animal feed additives. However, the functional researches mainly focused on BAs mixture, and the influence of the individual BA on fishes was still limited. In the present study, Nile tilapia were fed basal diet with three levels of sodium taurocholate at 0 mg/kg (CON), 300 mg/kg (TCAL), and 600 mg/kg (TCAH) for 8 weeks. The results indicated that addition of sodium taurocholate did not significantly influence the growth performance. Instead, TCAH group had higher cholesterol accumulation with liver fibrosis. In TCAH group, the level of nuclear factor E2-related factor 2 (nrf2) signaling-associated oxidative stress factors significantly increased in the liver. Additionally, fish in TCAH group had the highest expression level of genes encoding endoplasmic reticulum (ER) stress and inflammatory cytokines in the liver. In conclusion, 300 mg/kg of sodium taurocholate did not significantly influence the growth performance of fish, while 600 mg/kg of sodium taurocholate markedly induced cholesterol accumulation and liver injury, suggesting that the application of taurocholic acid in aquafeed should be re-evaluated.
Collapse
Affiliation(s)
- Fei-Fei Ding
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Miao Li
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Tong Wang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Nan-Nan Zhou
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Fang Qiao
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
3
|
Zhou T, Cao L, Du Y, Qin L, Lu Y, Zhang Q, He Y, Tan D. Gypenosides ameliorate high-fat diet-induced nonalcoholic fatty liver disease in mice by regulating lipid metabolism. PeerJ 2023; 11:e15225. [PMID: 37065701 PMCID: PMC10103699 DOI: 10.7717/peerj.15225] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 03/23/2023] [Indexed: 04/18/2023] Open
Abstract
Gypenosides (GP), extracted from the traditional Chinese herb Gynostemma pentaphyllum (Thunb.) Makino, have been used to treat metabolic disorders, including lipid metabolism disorders and diabetes. Although recent studies have confirmed their beneficial effects in nonalcoholic fatty liver disease (NAFLD), the underlying therapeutic mechanism remains unclear. In this study, we explored the protective mechanism of GP against NAFLD in mice and provided new insights into the prevention and treatment of NAFLD. Male C57BL6/J mice were divided into three experimental groups: normal diet, high-fat diet (HFD), and GP groups. The mice were fed an HFD for 16 weeks to establish an NAFLD model and then treated with GP for 22 weeks. The transcriptome and proteome of the mice livers were profiled using RNA sequencing and high-resolution mass spectrometry, respectively. The results showed that GP decreased serum lipid levels, liver index, and liver fat accumulation in mice. Principal component and heatmap analyses indicated that GP significantly modulated the changes in the expression of genes associated with HFD-induced NAFLD. The 164 differentially expressed genes recovered using GP were enriched in fatty acid and steroid metabolism pathways. Further results showed that GP reduced fatty acid synthesis by downregulating the expression of Srebf1, Fasn, Acss2, Acly, Acaca, Fads1, and Elovl6; modulated glycerolipid metabolism by inducing the expression of Mgll; promoted fatty acid transportation and degradation by inducing the expression of Slc27a1, Cpt1a, and Ehhadh; and reduced hepatic cholesterol synthesis by downregulating the expression of Tm7sf2, Ebp, Sc5d, Lss, Fdft1, Cyp51, Nsdhl, Pmvk, Mvd, Fdps, and Dhcr7. The proteomic data further indicated that GP decreased the protein expression levels of ACACA, ACLY, ACSS2, TM7SF2, EBP, FDFT1, NSDHL, PMVK, MVD, FDPS, and DHCR7 and increased those of MGLL, SLC27A1, and EHHADH. In conclusion, GP can regulate the key genes involved in hepatic lipid metabolism in NAFLD mice, providing initial evidence for the mechanisms underlying the therapeutic effect of GP in NAFLD.
Collapse
Affiliation(s)
- Tingting Zhou
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ligang Cao
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yimei Du
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Lin Qin
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yanliu Lu
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qianru Zhang
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuqi He
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Daopeng Tan
- Guizhou Engineering Research Center of Industrial Key-technology for Dendrobium Nobile, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
4
|
Attenuation of hepatic fibrosis by p-Coumaric acid via modulation of NLRP3 inflammasome activation in C57BL/6 mice. J Nutr Biochem 2023; 112:109204. [PMID: 36400112 DOI: 10.1016/j.jnutbio.2022.109204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 09/28/2022] [Accepted: 11/02/2022] [Indexed: 11/17/2022]
Abstract
A prolonged high-fat and high-sucrose (HFHS) diet induces hepatic inflammation and mediates hepatic stellate cell (HSC) activation, which result in hepatic fibrosis. Aberrant activation of the innate immune system components, such as the NOD-like receptor protein 3 (NLRP3) inflammasome, has been implicated in HSC activation and hepatic fibrosis. We have previously shown that p-coumaric acid (PCA)-enriched peanut sprout extracts exert anti-inflammatory effects. However, it is unknown whether PCA reduces hepatic fibrosis by modulating innate immunity and HSC activation. To test this hypothesis, C57BL/6 male mice were randomly assigned to three groups and fed low-fat (LF) diet (11% calories from fat), high-fat (HF) diet (60% calories from fat, 0.2% cholesterol) with sucrose drink (20% sucrose, HFHS), or HFHS diet with PCA treatment (HFHS+PCA, 50 mg/kg body weight, intraperitoneally) for 13 weeks. The results showed that PCA treatment (1) partly improved systemic insulin sensitivity without altering adiposity, (2) attenuated hepatic signaling pathways associated with NLRP3 inflammasome activation, including toll-like receptor 4 (TLR4)/nuclear factor kappa B (NFκB), and endoplasmic reticulum/oxidative stress, and (3) reduced circulating interleukin (IL)-1β levels. More importantly, PCA ameliorated hepatic fibrosis compared to that in the HFHS group, and the anti-fibrogenic effects of PCA were confirmed in vitro in transforming growth factor β (TGFβ) treated-LX-2 HSCs. The role of PCA in decreased NLRP3 activation and caspase-1 cleavage was recapitulated in primary bone marrow‒derived macrophages. These findings indicate that PCA contributes to the prevention of HFHS diet‒mediated liver fibrosis, partly by attenuating the activation of the NLRP3 inflammasome.
Collapse
|
5
|
Arruda AP, Parlakgül G. Endoplasmic Reticulum Architecture and Inter-Organelle Communication in Metabolic Health and Disease. Cold Spring Harb Perspect Biol 2023; 15:a041261. [PMID: 35940911 PMCID: PMC9899651 DOI: 10.1101/cshperspect.a041261] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The endoplasmic reticulum (ER) is a key organelle involved in the regulation of lipid and glucose metabolism, proteostasis, Ca2+ signaling, and detoxification. The structural organization of the ER is very dynamic and complex, with distinct subdomains such as the nuclear envelope and the peripheral ER organized into ER sheets and tubules. ER also forms physical contact sites with all other cellular organelles and with the plasma membrane. Both form and function of the ER are highly adaptive, with a potent capacity to respond to transient changes in environmental cues such as nutritional fluctuations. However, under obesity-induced chronic stress, the ER fails to adapt, leading to ER dysfunction and the development of metabolic pathologies such as insulin resistance and fatty liver disease. Here, we discuss how the remodeling of ER structure and contact sites with other organelles results in diversification of metabolic function and how perturbations to this structural flexibility by chronic overnutrition contribute to ER dysfunction and metabolic pathologies in obesity.
Collapse
Affiliation(s)
- Ana Paula Arruda
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Güneş Parlakgül
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
6
|
ZHOU T, CAO L, QIN L, LU Y, HE Y, TAN D. Differential effects of medium- and long-term high-fat diets on the expression of genes or proteins related to nonalcoholic fatty liver disease in mice. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.117522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
7
|
Taskinen JH, Ruhanen H, Matysik S, Käkelä R, Olkkonen VM. Global effects of pharmacologic inhibition of OSBP in human umbilical vein endothelial cells. Steroids 2022; 185:109053. [PMID: 35623602 DOI: 10.1016/j.steroids.2022.109053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/14/2022] [Accepted: 05/23/2022] [Indexed: 11/26/2022]
Abstract
Oxysterol-binding protein (OSBP) is a cholesterol/PI4P exchanger at contacts of the endoplasmic reticulum (ER) with trans-Golgi network (TGN) and endosomes. Several central endothelial cell (EC) functions depend on adequate cholesterol distribution in cellular membranes. Here we elucidated the effects of pharmacologic OSBP inhibition on the lipidome and transcriptome of human umbilical vein endothelial cells (HUVECs). OSBP was inhibited for 24 h with 25 nM Schweinfurthin G (SWG) or Orsaponin (OSW-1), followed by analyses of cellular cholesterol, 27-hydroxy-cholesterol, and triacylglycerol concentration, phosphatidylserine synthesis rate, the lipidome, as well as lipid droplet staining and western analysis of OSBP protein. Next-generation RNA sequencing of the SWG-treated and control HUVECs and angiogenesis assays were performed. Protein-normalized lipidomes of the inhibitor-treated cells revealed decreases in glycerophospholipids, the most pronounced effect being on phosphatidylserines and the rate of their synthesis, as well as increases in cholesteryl esters, triacylglycerols and lipid droplet number. Transcriptome analysis of SWG-treated cells suggested ER stress responses apparently caused by disturbed cholesterol exit from the ER, as indicated by suppression of cholesterol biosynthetic genes. OSBP was associated with the TGN in the absence of inhibitors and disappeared therefrom in inhibitor-treated cells in a time-dependent manner, coinciding with OSBP reduction on western blots. Prolonged treatment with SWG or OSW-1 inhibited angiogenesis in vitro. To conclude, inhibition of OSBP in primary endothelial cells induced multiple effects on the lipidome, transcriptome changes suggesting ER stress, and disruption of in vitro angiogenic capacity. Thus, OSBP is a crucial regulator of EC lipid homeostasis and angiogenic capacity.
Collapse
Affiliation(s)
- Juuso H Taskinen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland.
| | - Hanna Ruhanen
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Molecular and Integrative Biosciences Research Programme, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland.
| | - Silke Matysik
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany.
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Molecular and Integrative Biosciences Research Programme, University of Helsinki, Viikinkaari 1, PO BOX 65, 00014 University of Helsinki, Finland.
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland; Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
8
|
Torres S, Segalés P, García-Ruiz C, Fernández-Checa JC. Mitochondria and the NLRP3 Inflammasome in Alcoholic and Nonalcoholic Steatohepatitis. Cells 2022; 11:1475. [PMID: 35563780 PMCID: PMC9105698 DOI: 10.3390/cells11091475] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/18/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Alcoholic (ASH) and nonalcoholic steatohepatitis (NASH) are advanced stages of fatty liver disease and two of the most prevalent forms of chronic liver disease. ASH and NASH are associated with significant risk of further progression to cirrhosis and hepatocellular carcinoma (HCC), the most common type of liver cancer, and a major cause of cancer-related mortality. Despite extensive research and progress in the last decades to elucidate the mechanisms of the development of ASH and NASH, the pathogenesis of both diseases is still poorly understood. Mitochondrial damage and activation of inflammasome complexes have a role in inducing and sustaining liver damage. Mitochondrial dysfunction produces inflammatory factors that activate the inflammasome complexes. NLRP3 inflammasome (nucleotide-binding oligomerization domain-like receptor protein 3) is a multiprotein complex that activates caspase 1 and the release of pro-inflammatory cytokines, including interleukin-1β (IL-1β) and interleukin-18 (IL-18), and contributes to inflammatory pyroptotic cell death. The present review, which is part of the issue "Mitochondria in Liver Pathobiology", provides an overview of the role of mitochondrial dysfunction and NLRP3 activation in ASH and NASH.
Collapse
Affiliation(s)
- Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
| | - Paula Segalés
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
| | - Carmen García-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - José C. Fernández-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (S.T.); (P.S.)
- Liver Unit, Hospital Clinic I Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, 28029 Madrid, Spain
- Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
9
|
Sabapathy T, Helmerhorst E, Ellison G, Bridgeman SC, Mamotte CD. High-fat diet induced alterations in plasma membrane cholesterol content impairs insulin receptor binding and signalling in mouse liver but is ameliorated by atorvastatin. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166372. [PMID: 35248691 DOI: 10.1016/j.bbadis.2022.166372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 10/18/2022]
Abstract
A high-fat diet (HFD) impairs insulin binding and signalling and may contribute to the development of insulin resistance. In addition, in vitro studies have shown that alterations in plasma membrane cholesterol influence ligand binding and downstream signalling for several receptor-tyrosine kinases (RTKs), including the insulin receptor. Using an ex vivo approach, we explored the effects of a HFD on insulin binding and signalling in mouse liver and relate these to observed changes in plasma membrane cholesterol. Mice fed a HFD demonstrated decreased insulin signalling compared to mice fed a normal chow diet (ND), indicated by a 3-fold decrease in insulin binding (P < 0.001) and a similar decrease in insulin receptor phosphorylation (~2.5-fold; P < 0.0001). Interestingly, we also observed a marked decrease in the cholesterol content of liver plasma membranes in the HFD fed mice (P < 0.0001). These effects of the HFD were found to be ameliorated by atorvastatin treatment (P < 0.0001). However, in ND mice, atorvastatin had no influence on membrane cholesterol content or insulin binding and signalling. The influence of membrane cholesterol on insulin binding and signalling was also corroborated in HepG2 cells. To the best of our knowledge, this is the first demonstration of the effects of a HFD and atorvastatin treatment on changes in plasma membrane cholesterol content and the consequent effects on insulin binding and signalling. Collectively, these findings suggest that changes in membrane cholesterol content could be an important underlying reason for the long-known effects of a HFD on the development of insulin resistance.
Collapse
Affiliation(s)
- Thiru Sabapathy
- School of Medicine, Curtin University, Bentley, Western Australia 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia 6102, Australia
| | - Erik Helmerhorst
- School of Medicine, Curtin University, Bentley, Western Australia 6102, Australia
| | - Gaewyn Ellison
- School of Medicine, Curtin University, Bentley, Western Australia 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia 6102, Australia; School of Molecular and Life Sciences, Curtin University, Bentley, Western Australia 6102, Australia.
| | - Stephanie C Bridgeman
- School of Medicine, Curtin University, Bentley, Western Australia 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia 6102, Australia
| | - Cyril D Mamotte
- School of Medicine, Curtin University, Bentley, Western Australia 6102, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia 6102, Australia.
| |
Collapse
|
10
|
Zou J, Yan C, Wan JB. Red yeast rice ameliorates non-alcoholic fatty liver disease through inhibiting lipid synthesis and NF-κB/NLRP3 inflammasome-mediated hepatic inflammation in mice. Chin Med 2022; 17:17. [PMID: 35078487 PMCID: PMC8788078 DOI: 10.1186/s13020-022-00573-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Red yeast rice (RYR), a nutraceutical with a profound cholesterol-lowering effect, was found to attenuate non-alcoholic fatty liver disease (NAFLD) in mice. Despite monacolin K in RYR being a specific inhibitor of hydroxymethylglutaryl-coenzyme A reductase (HMCGR), the mechanisms underlying the protective effects of RYR against NAFLD are not fully elucidated. METHODS Using a mouse model of high-fat diet (HFD) feeding and a cellular model of HepG2 cells challenged by lipopolysaccharide (LPS) and palmitic acid (PA), the possible molecular mechanisms were exploited in the aspects of NF-κB/NLRP3 inflammasome and mTORC1-SREBPs signaling pathways by examining the relevant gene/protein expressions. Subsequently, the correlation between these two signals was also verified using cellular experiments. RESULTS RYR ameliorated lipid accumulation and hepatic inflammation in vivo and in vitro. RYR improved lipid metabolism through modulating mTORC1-SREBPs and their target genes related to triglyceride and cholesterol synthesis. Furthermore, RYR suppressed hepatic inflammation by inhibiting the NF-κB/NLRP3 inflammasome signaling. Interestingly, the treatment with RYR or MCC950, a specific NLRP3 inhibitor, resulted in the reduced lipid accumulation in HepG2 cells challenged by LPS plus PA, suggesting that the inhibitory effects of RYR on NLRP3 inflammasome-mediated hepatic inflammation may partially, in turn, contribute to the lipid-lowering effect of RYR. CONCLUSIONS The modulation of NF-κB/NLRP3 inflammasome and lipid synthesis may contribute to the ameliorative effects of RYR against HFD-induced NAFLD.
Collapse
Affiliation(s)
- Jian Zou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, Taipa, China
| | - Chunyan Yan
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, Taipa, China.
| |
Collapse
|
11
|
Zhang Y, Ge X, Li Y, Zhang B, Wang P, Hao M, Gao P, Zhao Y, Sun T, Lu S, Ma W. TWIST2 and the PPAR signaling pathway are important in the progression of nonalcoholic steatohepatitis. Lipids Health Dis 2021; 20:39. [PMID: 33879188 PMCID: PMC8059034 DOI: 10.1186/s12944-021-01458-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To investigate the roles of the transcription factors twist family bHLH transcription factor 1 (TWIST1), twist family bHLH transcription factor 2 (TWIST2), and peroxisome proliferator activated receptor gamma (PPARγ) in the progression of nonalcoholic steatohepatitis. METHODS The protein levels of TWIST1, TWIST2 and PPARγ were determined in the serum of nonalcoholic fatty liver disease (NAFLD) patients and healthy controls by enzyme-linked immunosorbent assay (ELISA). An in vivo model for fatty liver was established by feeding C57BL/6 J mice a high-fat diet (HFD). An in vitro model of steatosis was established by treating LO-2 cells with oleic acid (OA). RNA sequencing was performed on untreated and OA-treated LO-2 cells followed by TWIST1, TWIST2 and PPARγ gene mRNA levels analysis, Gene Ontology (GO) enrichment and pathway analysis. RESULTS The TWIST2 serum protein levels decreased significantly in all fatty liver groups (P < 0.05), while TWIST1 varied. TWIST2 tended to be lower in mice fed an HFD and was significantly lower at 3 months. Similarly, in the in vitro model, the TWIST2 protein level was downregulated significantly at 48 and 72 h after OA treatment. RNA sequencing of LO-2 cells showed an approximately 2.3-fold decrease in TWIST2, with no obvious change in TWIST1 and PPARγ. The PPAR signaling pathway was enriched, with 4 genes upregulated in OA-treated cells (P = 0.0018). The interleukin (IL)-17 and tumor necrosis factor (TNF) signaling pathways were enriched in OA-treated cells. CONCLUSIONS The results provide evidence that the TWIST2 and PPAR signaling pathways are important in NAFLD and shed light on a potential mechanism of steatosis.
Collapse
Affiliation(s)
- Yanmei Zhang
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
- Department of Clinical Laboratory, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, P.R. China
| | - Xiaoxiao Ge
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
| | - Yongqing Li
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
| | - Bingyang Zhang
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
| | - Peijun Wang
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
| | - Mingju Hao
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, P. R. China
| | - Peng Gao
- Medical Research Center, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, P. R. China
| | - Yueyi Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, P. R. China
| | - Tao Sun
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, P. R. China
| | - Sumei Lu
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China.
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, P. R. China.
| | - Wanshan Ma
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, 250014, P. R. China.
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, P. R. China.
| |
Collapse
|
12
|
Signaling Nodes Associated with Endoplasmic Reticulum Stress during NAFLD Progression. Biomolecules 2021; 11:biom11020242. [PMID: 33567666 PMCID: PMC7915814 DOI: 10.3390/biom11020242] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/19/2022] Open
Abstract
Excess and sustained endoplasmic reticulum (ER) stress, paired with a failure of initial adaptive responses, acts as a critical trigger of nonalcoholic fatty liver disease (NAFLD) progression. Unfortunately, there is no drug currently approved for treatment, and the molecular basis of pathogenesis by ER stress remains poorly understood. Classical ER stress pathway molecules have distinct but inter-connected functions and complicated effects at each phase of the disease. Identification of the specific molecular signal mediators of the ER stress-mediated pathogenesis is, therefore, a crucial step in the development of new treatments. These signaling nodes may be specific to the cell type and/or the phase of disease progression. In this review, we highlight the recent advancements in knowledge concerning signaling nodes associated with ER stress and NAFLD progression in various types of liver cells.
Collapse
|
13
|
Charles-Messance H, Mitchelson KA, De Marco Castro E, Sheedy FJ, Roche HM. Regulating metabolic inflammation by nutritional modulation. J Allergy Clin Immunol 2020; 146:706-720. [DOI: 10.1016/j.jaci.2020.08.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
|
14
|
Oczkowicz M, Szmatoła T, Świątkiewicz M, Koseniuk A, Smołucha G, Witarski W, Wierzbicka A. 3'quant mRNA-Seq of Porcine Liver Reveals Alterations in UPR, Acute Phase Response, and Cholesterol and Bile Acid Metabolism in Response to Different Dietary Fats. Genes (Basel) 2020; 11:genes11091087. [PMID: 32961898 PMCID: PMC7565913 DOI: 10.3390/genes11091087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022] Open
Abstract
Animal fats are considered to be unhealthy, in contrast to vegetable fats, which are rich in unsaturated fatty acids. However, the use of some fats, such as coconut oil, is still controversial. In our experiment, we divided experimental animals (domestic pigs) into three groups differing only in the type of fat used in the diet: group R: rapeseed oil (n = 5); group B: beef tallow (n = 5); group C: coconut oil (n = 6). After transcriptomic analysis of liver samples, we identified 188, 93, and 53 DEGs (differentially expressed genes) in R vs. B, R vs. C, and B vs. C comparisons, respectively. Next, we performed a functional analysis of identified DEGs with String and IPA software. We observed the enrichment of genes engaged in the unfolded protein response (UPR) and the acute phase response among genes upregulated in B compared to R. In contrast, cholesterol biosynthesis and cholesterol efflux enrichments were observed among genes downregulated in B when compared to R. Moreover, activation of the UPR and inhibition of the sirtuin signaling pathway were noted in C when compared to R. The most striking difference in liver transcriptomic response between C and B was the activation of the acute phase response and inhibition of bile acid synthesis in the latest group. Our results suggest that excessive consumption of animal fats leads to the activation of a cascade of mutually propelling processes harmful to the liver: inflammation, UPR, and imbalances in the biosynthesis of cholesterol and bile acids via altered organelle membrane composition. Nevertheless, these studies should be extended with analysis at the level of proteins and their function.
Collapse
Affiliation(s)
- Maria Oczkowicz
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland; (T.S.); (A.K.); (G.S.); (W.W.); (A.W.)
- Correspondence: ; Tel.: +48666081109
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland; (T.S.); (A.K.); (G.S.); (W.W.); (A.W.)
- Centre of Experimental and Innovative Medicine, University of Agriculture in Kraków, Al. Mickiewicza 24/28, 30-059 Kraków, Poland
| | - Małgorzata Świątkiewicz
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland;
| | - Anna Koseniuk
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland; (T.S.); (A.K.); (G.S.); (W.W.); (A.W.)
| | - Grzegorz Smołucha
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland; (T.S.); (A.K.); (G.S.); (W.W.); (A.W.)
| | - Wojciech Witarski
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland; (T.S.); (A.K.); (G.S.); (W.W.); (A.W.)
| | - Alicja Wierzbicka
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul Krakowska 1, 32-083 Balice, Poland; (T.S.); (A.K.); (G.S.); (W.W.); (A.W.)
| |
Collapse
|
15
|
Ng DS. Emerging insight into the role of cellular cholesterol in the pathogenesis of nonalcoholic steatohepatitis progression. Curr Opin Lipidol 2020; 31:262-263. [PMID: 32692037 DOI: 10.1097/mol.0000000000000693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Dominic S Ng
- Department of Medicine, St Michael's Hospital
- Department of Physiology, Faculty of Medicine
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Norum KR, Remaley AT, Miettinen HE, Strøm EH, Balbo BEP, Sampaio CATL, Wiig I, Kuivenhoven JA, Calabresi L, Tesmer JJ, Zhou M, Ng DS, Skeie B, Karathanasis SK, Manthei KA, Retterstøl K. Lecithin:cholesterol acyltransferase: symposium on 50 years of biomedical research from its discovery to latest findings. J Lipid Res 2020; 61:1142-1149. [PMID: 32482717 PMCID: PMC7397740 DOI: 10.1194/jlr.s120000720] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/21/2020] [Indexed: 01/04/2023] Open
Abstract
LCAT converts free cholesterol to cholesteryl esters in the process of reverse cholesterol transport. Familial LCAT deficiency (FLD) is a genetic disease that was first described by Kaare R. Norum and Egil Gjone in 1967. This report is a summary from a 2017 symposium where Dr. Norum recounted the history of FLD and leading experts on LCAT shared their results. The Tesmer laboratory shared structural findings on LCAT and the close homolog, lysosomal phospholipase A2. Results from studies of FLD patients in Finland, Brazil, Norway, and Italy were presented, as well as the status of a patient registry. Drs. Kuivenhoven and Calabresi presented data from carriers of genetic mutations suggesting that FLD does not necessarily accelerate atherosclerosis. Dr. Ng shared that LCAT-null mice were protected from diet-induced obesity, insulin resistance, and nonalcoholic fatty liver disease. Dr. Zhou presented multiple innovations for increasing LCAT activity for therapeutic purposes, whereas Dr. Remaley showed results from treatment of an FLD patient with recombinant human LCAT (rhLCAT). Dr. Karathanasis showed that rhLCAT infusion in mice stimulates cholesterol efflux and suggested that it could also enhance cholesterol efflux from macrophages. While the role of LCAT in atherosclerosis remains elusive, the consensus is that a continued study of both the enzyme and disease will lead toward better treatments for patients with heart disease and FLD.
Collapse
Affiliation(s)
- Kaare R Norum
- Department of Nutrition, University of Oslo, Oslo, Norway
| | | | - Helena E Miettinen
- Department of Medicine, University of Helsinki and University Central Hospital, Helsinki, Finland
| | - Erik H Strøm
- Departments of Pathology Oslo University Hospital, Oslo, Norway
| | - Bruno E P Balbo
- Division of Nephrology and Molecular Medicine Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Carlos A T L Sampaio
- Division of Nephrology and Molecular Medicine Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Ingrid Wiig
- Centre for Rare Disorders, Oslo University Hospital, Oslo, Norway
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Laura Calabresi
- Center E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - John J Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette, IN
| | - Mingyue Zhou
- Cardiometabolic Disorder Research, AMGEN, San Francisco, CA
| | - Dominic S Ng
- Department of Medicine, University of Toronto and Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Bjørn Skeie
- Anesthesiology, Oslo University Hospital, Oslo, Norway
| | | | - Kelly A Manthei
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Kjetil Retterstøl
- Department of Nutrition, University of Oslo, Oslo, Norway .,Department of Endocrinology, Morbid Obesity, and Preventive Medicine, Lipid Clinic, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Dia VP, Bradwell J, Pangloli P. Sorghum Phenolics Inhibits Inflammasomes in Lipopolysaccharide (LPS)-Primed and Adenosine Triphosphate (ATP)-Activated Macrophages. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2019; 74:307-315. [PMID: 31104201 DOI: 10.1007/s11130-019-00736-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sorghum contains phenolic compounds with reported biological activities. The objective was to evaluate the ability of sorghum phenolic extract to inhibit inflammasomes in THP-1 human macrophages. THP-1 human macrophages was pre-treated with sorghum phenolics and the inflammasome was activated by lipopolysaccharide and adenosine triphosphate treatment. Treatment of macrophages with 50 μg sorghum extract/mL reduced IL-1β and IL-18 secretion by 59.7 and 32.0%, respectively, associated with caspase-1 activity reduction. Moreover, the production of intracellular reactive oxygen species was reduced. Our data showed the potential role of sorghum phenolics in diseases associated with aberrant inflammasomes activation.
Collapse
Affiliation(s)
- Vermont P Dia
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA.
| | - Jordan Bradwell
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA
| | - Philipus Pangloli
- Department of Food Science, The University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
18
|
Püschel GP, Henkel J. Dietary cholesterol does not break your heart but kills your liver. Porto Biomed J 2019; 3:e12. [PMID: 31595236 PMCID: PMC6726297 DOI: 10.1016/j.pbj.0000000000000012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
It is increasingly accepted that dietary cholesterol has a much lower impact on the progression of cardiovascular disease than previously assumed. However, both animal experiments and human studies seem to support the view that dietary cholesterol may contribute to the transition from benign steatosis to the potentially fatal non-alcoholic steatohepatitis. Cholesterol esters and cholesterol accumulate in the hepatocyte and impair its function. This leads to oxidative stress and endoplasmic reticulum stress triggering the release of pro-inflammatory cytokines and rendering the hepatocyte more susceptible to apoptotic or necrotic cell death. Kupffer cells group around dying hepatocytes and phagocytose the hepatocyte debris and lipids. In addition, they are exposed to lipid peroxidation products released from hepatocytes. Kupffer cells, thus activated, release pro-inflammatory, chemotactic and profibrotic cytokines that promote inflammation and fibrosis. Therefore, dietary cholesterol may be harmful to the liver, in particular when administered in combination with polyunsaturated fatty acids that favor lipid peroxidation.
Collapse
Affiliation(s)
- Gerhard P Püschel
- Department of Nutritional Biochemistry, University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Janin Henkel
- Department of Nutritional Biochemistry, University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| |
Collapse
|
19
|
Platko K, Lebeau PF, Byun JH, Poon SV, Day EA, MacDonald ME, Holzapfel N, Mejia-Benitez A, Maclean KN, Krepinsky JC, Austin RC. GDF10 blocks hepatic PPARγ activation to protect against diet-induced liver injury. Mol Metab 2019; 27:62-74. [PMID: 31288993 PMCID: PMC6717799 DOI: 10.1016/j.molmet.2019.06.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Growth differentiation factors (GDFs) and bone-morphogenic proteins (BMPs) are members of the transforming growth factor β (TGFβ) superfamily and are known to play a central role in the growth and differentiation of developing tissues. Accumulating evidence, however, demonstrates that many of these factors, such as BMP-2 and -4, as well as GDF15, also regulate lipid metabolism. GDF10 is a divergent member of the TGFβ superfamily with a unique structure and is abundantly expressed in brain and adipose tissue; it is also secreted by the latter into the circulation. Although previous studies have demonstrated that overexpression of GDF10 reduces adiposity in mice, the role of circulating GDF10 on other tissues known to regulate lipid, like the liver, has not yet been examined. METHODS Accordingly, GDF10-/- mice and age-matched GDF10+/+ control mice were fed either normal control diet (NCD) or high-fat diet (HFD) for 12 weeks and examined for changes in liver lipid homeostasis. Additional studies were also carried out in primary and immortalized human hepatocytes treated with recombinant human (rh)GDF10. RESULTS Here, we show that circulating GDF10 levels are increased in conditions of diet-induced hepatic steatosis and, in turn, that secreted GDF10 can prevent excessive lipid accumulation in hepatocytes. We also report that GDF10-/- mice develop an obese phenotype as well as increased liver triglyceride accumulation when fed a NCD. Furthermore, HFD-fed GDF10-/- mice develop increased steatosis, endoplasmic reticulum (ER) stress, fibrosis, and injury of the liver compared to HFD-fed GDF10+/+ mice. To explain these observations, studies in cultured hepatocytes led to the observation that GDF10 attenuates nuclear peroxisome proliferator-activated receptor γ (PPARγ) activity; a transcription factor known to induce de novo lipogenesis. CONCLUSION Our work delineates a hepatoprotective role of GDF10 as an adipokine capable of regulating hepatic lipid levels by blocking de novo lipogenesis to protect against ER stress and liver injury.
Collapse
Affiliation(s)
- Khrystyna Platko
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Paul F Lebeau
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Jae Hyun Byun
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Samantha V Poon
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Emily A Day
- The Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8N 4A6, Canada
| | - Melissa E MacDonald
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Nicholas Holzapfel
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Aurora Mejia-Benitez
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Kenneth N Maclean
- The Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Joan C Krepinsky
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada
| | - Richard C Austin
- Department of Medicine, McMaster University, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, Hamilton, Ontario, L8N 4A6, Canada.
| |
Collapse
|
20
|
Spradley FT, Smith JA, Alexander BT, Anderson CD. Developmental origins of nonalcoholic fatty liver disease as a risk factor for exaggerated metabolic and cardiovascular-renal disease. Am J Physiol Endocrinol Metab 2018; 315:E795-E814. [PMID: 29509436 PMCID: PMC6293166 DOI: 10.1152/ajpendo.00394.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intrauterine growth restriction (IUGR) is linked to increased risk for chronic disease. Placental ischemia and insufficiency in the mother are implicated in predisposing IUGR offspring to metabolic dysfunction, including hypertension, insulin resistance, abnormalities in glucose homeostasis, and nonalcoholic fatty liver disease (NAFLD). It is unclear whether these metabolic disturbances contribute to the developmental origins of exaggerated cardiovascular-renal disease (CVRD) risk accompanying IUGR. IUGR impacts the pancreas, adipose tissue, and liver, which are hypothesized to program for hepatic insulin resistance and subsequent NAFLD. NAFLD is projected to become the major cause of chronic liver disease and contributor to uncontrolled type 2 diabetes mellitus, which is a leading cause of chronic kidney disease. While NAFLD is increased in experimental models of IUGR, lacking is a full comprehension of the mechanisms responsible for programming of NAFLD and whether this potentiates susceptibility to liver injury. The use of well-established and clinically relevant rodent models, which mimic the clinical characteristics of IUGR, metabolic disturbances, and increased blood pressure in the offspring, will permit investigation into mechanisms linking adverse influences during early life and later chronic health. The purpose of this review is to propose mechanisms, including those proinflammatory in nature, whereby IUGR exacerbates the pathogenesis of NAFLD and how these adverse programmed outcomes contribute to exaggerated CVRD risk. Understanding the etiology of the developmental origins of chronic disease will allow investigators to uncover treatment strategies to intervene in the mother and her offspring to halt the increasing prevalence of metabolic dysfunction and CVRD.
Collapse
Affiliation(s)
- Frank T Spradley
- Department of Surgery, Division of Transplant and Hepatobiliary Surgery, School of Medicine, The University of Mississippi Medical Center , Jackson, Mississippi
- Cardiovascular-Renal Research Center, The University of Mississippi Medical Center , Jackson, Mississippi
- Department of Physiology and Biophysics, The University of Mississippi Medical Center , Jackson, Mississippi
| | - Jillian A Smith
- Department of Surgery, Division of Transplant and Hepatobiliary Surgery, School of Medicine, The University of Mississippi Medical Center , Jackson, Mississippi
| | - Barbara T Alexander
- Cardiovascular-Renal Research Center, The University of Mississippi Medical Center , Jackson, Mississippi
- Department of Physiology and Biophysics, The University of Mississippi Medical Center , Jackson, Mississippi
| | - Christopher D Anderson
- Department of Surgery, Division of Transplant and Hepatobiliary Surgery, School of Medicine, The University of Mississippi Medical Center , Jackson, Mississippi
- Cardiovascular-Renal Research Center, The University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
21
|
Lebeaupin C, Vallée D, Hazari Y, Hetz C, Chevet E, Bailly-Maitre B. Endoplasmic reticulum stress signalling and the pathogenesis of non-alcoholic fatty liver disease. J Hepatol 2018; 69:927-947. [PMID: 29940269 DOI: 10.1016/j.jhep.2018.06.008] [Citation(s) in RCA: 631] [Impact Index Per Article: 90.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/22/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022]
Abstract
The global epidemic of obesity has been accompanied by a rising burden of non-alcoholic fatty liver disease (NAFLD), with manifestations ranging from simple steatosis to non-alcoholic steatohepatitis, potentially developing into hepatocellular carcinoma. Although much attention has focused on NAFLD, its pathogenesis remains largely obscure. The hallmark of NAFLD is the hepatic accumulation of lipids, which subsequently leads to cellular stress and hepatic injury, eventually resulting in chronic liver disease. Abnormal lipid accumulation often coincides with insulin resistance in steatotic livers and is associated with perturbed endoplasmic reticulum (ER) proteostasis in hepatocytes. In response to chronic ER stress, an adaptive signalling pathway known as the unfolded protein response is triggered to restore ER proteostasis. However, the unfolded protein response can cause inflammation, inflammasome activation and, in the case of non-resolvable ER stress, the death of hepatocytes. Experimental data suggest that the unfolded protein response influences hepatic tumour development, aggressiveness and response to treatment, offering novel therapeutic avenues. Herein, we provide an overview of the evidence linking ER stress to NAFLD and discuss possible points of intervention.
Collapse
Affiliation(s)
| | - Deborah Vallée
- Université Côte d'Azur, INSERM, U1065, C3M, 06200 Nice, France
| | - Younis Hazari
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, 02115 Boston, MA, USA
| | - Eric Chevet
- "Chemistry, Oncogenesis, Stress, Signaling", Inserm U1242, Université de Rennes, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | | |
Collapse
|
22
|
Kim Y, Natarajan SK, Chung S. Gamma-Tocotrienol Attenuates the Hepatic Inflammation and Fibrosis by Suppressing Endoplasmic Reticulum Stress in Mice. Mol Nutr Food Res 2018; 62:e1800519. [DOI: 10.1002/mnfr.201800519] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/26/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Yongeun Kim
- Department of Nutrition and Health Sciences; University of Nebraska-Lincoln; Lincoln NE 68583 USA
| | - Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences; University of Nebraska-Lincoln; Lincoln NE 68583 USA
| | - Soonkyu Chung
- Department of Nutrition and Health Sciences; University of Nebraska-Lincoln; Lincoln NE 68583 USA
| |
Collapse
|
23
|
Boeckmans J, Natale A, Buyl K, Rogiers V, De Kock J, Vanhaecke T, Rodrigues RM. Human-based systems: Mechanistic NASH modelling just around the corner? Pharmacol Res 2018; 134:257-267. [PMID: 29964161 DOI: 10.1016/j.phrs.2018.06.029] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease characterized by excessive triglyceride accumulation in the liver accompanied by inflammation, cell stress and apoptosis. It is the tipping point to the life-threatening stages of non-alcoholic fatty liver disease (NAFLD). Despite the high prevalence of NASH, up to five percent of the global population, there are currently no approved drugs to treat this disease. Animal models, mostly based on specific diets and genetic modifications, are often employed in anti-NASH drug development. However, due to interspecies differences and artificial pathogenic conditions, they do not represent the human situation accurately and are inadequate for testing the efficacy and safety of potential new drugs. Human-based in vitro models provide a more legitimate representation of the human NASH pathophysiology and can be used to investigate the dysregulation of cellular functions associated with the disease. Also in silico methodologies and pathway-based approaches using human datasets, may contribute to a more accurate representation of NASH, thereby facilitating the quest for new anti-NASH drugs. In this review, we describe the molecular components of NASH and how human-based tools can contribute to unraveling the pathogenesis of this disease and be used in anti-NASH drug development. We also propose a roadmap for the development and application of human-based approaches for future investigation of NASH.
Collapse
Affiliation(s)
- Joost Boeckmans
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Alessandra Natale
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Karolien Buyl
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Vera Rogiers
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Joery De Kock
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Tamara Vanhaecke
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| | - Robim M Rodrigues
- Department of In VitroToxicology & Dermato-Cosmetology (IVTD) Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
24
|
Andersen CJ. Impact of Dietary Cholesterol on the Pathophysiology of Infectious and Autoimmune Disease. Nutrients 2018; 10:E764. [PMID: 29899295 PMCID: PMC6024721 DOI: 10.3390/nu10060764] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/02/2018] [Accepted: 06/11/2018] [Indexed: 01/02/2023] Open
Abstract
Cellular cholesterol metabolism, lipid raft formation, and lipoprotein interactions contribute to the regulation of immune-mediated inflammation and response to pathogens. Lipid pathways have been implicated in the pathogenesis of bacterial and viral infections, whereas altered lipid metabolism may contribute to immune dysfunction in autoimmune diseases, such as systemic lupus erythematosus, multiple sclerosis, and rheumatoid arthritis. Interestingly, dietary cholesterol may exert protective or detrimental effects on risk, progression, and treatment of different infectious and autoimmune diseases, although current findings suggest that these effects are variable across populations and different diseases. Research evaluating the effects of dietary cholesterol, often provided by eggs or as a component of Western-style diets, demonstrates that cholesterol-rich dietary patterns affect markers of immune inflammation and cellular cholesterol metabolism, while additionally modulating lipoprotein profiles and functional properties of HDL. Further, cholesterol-rich diets appear to differentially impact immunomodulatory lipid pathways across human populations of variable metabolic status, suggesting that these complex mechanisms may underlie the relationship between dietary cholesterol and immunity. Given the Dietary Guidelines for Americans 2015⁻2020 revision to no longer include limitations on dietary cholesterol, evaluation of dietary cholesterol recommendations beyond the context of cardiovascular disease risk is particularly timely. This review provides a comprehensive and comparative analysis of significant and controversial studies on the role of dietary cholesterol and lipid metabolism in the pathophysiology of infectious disease and autoimmune disorders, highlighting the need for further investigation in this developing area of research.
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Lecithin cholesterol acyltyransferase (LCAT) deficiency is a rare monogenic disorder causing lipoprotein dysregulation and multiple organ dysfunctions, including renal impairment. LCAT knockout mice have been shown informative in elucidating mechanisms of many major clinical morbid phenotypes. Extended characterization of the LDL receptor/LCAT double knockout (Ldlr/Lcat-DKO or DKO) mice had led to the discovery of a number of novel protective metabolic phenotypes, including resistance to obesity, nonalcoholic steatohepatitis (NASH) and insulin resistance. We seek to integrate the findings to explore novel pathogenic pathways. RECENT FINDINGS The chow fed DKO mice were found more insulin sensitive than their Ldlr-KO controls. Joint analyses of the three strains (DKO, Ldlr-KO and wild-type) revealed differential metabolic responses to a high cholesterol diet (HCD) vs. high-fat diet (HFD). DKO mice are protected from HFD-induced obesity, hepatic endoplasmic reticulum (ER) stress, insulin resistance, ER cholesterol and NASH markers (steatosis and inflammasomes). Joint analysis revealed the HFD-induced NASH is dependent on de-novo hepatic cholesterol biosynthesis. DKO mice are protected from HCD-induced hepatic ER stress, ER cholesterol, but not NASH, the latter likely due to cholesterol crystal accumulation. DKO mice were found to develop ectopic brown adipose tissue (BAT) in skeletal muscle. Ectopic BAT derived in part from myoblast in utero and from adult satellite cells. Primed expression of PRDM16 and UCP in quiescent satellite cell caused by LCAT deficiency synergizes with cell cholesterol depletion to induce satellite cell-to-BAT transdifferentiation. SUMMARY Metabolic phenotyping of selective LCAT null mice led to the discovery of novel metabolically protective pathways.
Collapse
|
26
|
Bellanti F, Villani R, Facciorusso A, Vendemiale G, Serviddio G. Lipid oxidation products in the pathogenesis of non-alcoholic steatohepatitis. Free Radic Biol Med 2017; 111:173-185. [PMID: 28109892 DOI: 10.1016/j.freeradbiomed.2017.01.023] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/11/2017] [Accepted: 01/15/2017] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the major public health challenge for hepatologists in the twenty-first century. NAFLD comprises a histological spectrum ranging from simple steatosis or fatty liver, to steatohepatitis, fibrosis, and cirrhosis. It can be categorized into two principal phenotypes: (1) non-alcoholic fatty liver (NAFL), and (2) non-alcoholic steatohepatitis (NASH). The mechanisms of NAFLD progression consist of lipid homeostasis alterations, redox unbalance, insulin resistance, and inflammation in the liver. Even though several studies show an association between the levels of lipid oxidation products and disease state, experimental evidence suggests that compounds such as reactive aldehydes and cholesterol oxidation products, in addition to representing hallmarks of hepatic oxidative damage, may behave as active players in liver dysfunction and the development of NAFLD. This review summarizes the processes that contribute to the metabolic alterations occurring in fatty liver that produce fatty acid and cholesterol oxidation products in NAFLD, with a focus on inflammation, the control of insulin signalling, and the transcription factors involved in lipid metabolism.
Collapse
Affiliation(s)
- Francesco Bellanti
- C.U.R.E. Centre for Liver Diseases Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Rosanna Villani
- C.U.R.E. Centre for Liver Diseases Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Antonio Facciorusso
- C.U.R.E. Centre for Liver Diseases Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Gianluigi Vendemiale
- C.U.R.E. Centre for Liver Diseases Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Gaetano Serviddio
- C.U.R.E. Centre for Liver Diseases Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| |
Collapse
|
27
|
Morris EM, McCoin CS, Allen JA, Gastecki ML, Koch LG, Britton SL, Fletcher JA, Fu X, Ding WX, Burgess SC, Rector RS, Thyfault JP. Aerobic capacity mediates susceptibility for the transition from steatosis to steatohepatitis. J Physiol 2017; 595:4909-4926. [PMID: 28504310 DOI: 10.1113/jp274281] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS Low intrinsic aerobic capacity is associated with increased all-cause and liver-related mortality in humans. Low intrinsic aerobic capacity in the low capacity runner (LCR) rat increases susceptibility to acute and chronic high-fat/high-sucrose diet-induced steatosis, without observed increases in liver inflammation. Addition of excess cholesterol to a high-fat/high-sucrose diet produced greater steatosis in LCR and high capacity runner (HCR) rats. However, the LCR rat demonstrated greater susceptibility to increased liver inflammatory and apoptotic markers compared to the HCR rat. The progressive non-alcoholic fatty liver disease observed in the LCR rats following western diet feeding was associated with further declines in liver fatty acid oxidation and mitochondrial respiratory capacity compared to HCR rats. ABSTRACT Low aerobic capacity increases risk for non-alcoholic fatty liver disease and liver-related disease mortality, but mechanisms mediating these effects remain unknown. We recently reported that rats bred for low aerobic capacity (low capacity runner; LCR) displayed susceptibility to high fat diet-induced steatosis in association with reduced hepatic mitochondrial fatty acid oxidation (FAO) and respiratory capacity compared to high aerobic capacity (high capacity runner; HCR) rats. Here we tested the impact of aerobic capacity on susceptibility for progressive liver disease following a 16-week 'western diet' (WD) high in fat (45% kcal), cholesterol (1% w/w) and sucrose (15% kcal). Unlike previously with a diet high in fat and sucrose alone, the inclusion of cholesterol in the WD induced hepatomegaly and steatosis in both HCR and LCR rats, while producing greater cholesterol ester accumulation in LCR compared to HCR rats. Importantly, WD-fed low-fitness LCR rats displayed greater inflammatory cell infiltration, serum alanine transaminase, expression of hepatic inflammatory markers (F4/80, MCP-1, TLR4, TLR2 and IL-1β) and effector caspase (caspase 3 and 7) activation compared to HCR rats. Further, LCR rats had greater WD-induced decreases in complete FAO and mitochondrial respiratory capacity. Intrinsic aerobic capacity had no impact on WD-induced hepatic steatosis; however, rats bred for low aerobic capacity developed greater hepatic inflammation, which was associated with reduced hepatic mitochondrial FAO and respiratory capacity and increased accumulation of cholesterol esters. These results confirm epidemiological reports that aerobic capacity impacts progression of liver disease and suggest that these effects are mediated through alterations in hepatic mitochondrial function.
Collapse
Affiliation(s)
- E Matthew Morris
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Colin S McCoin
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Julie A Allen
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michelle L Gastecki
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Lauren G Koch
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Justin A Fletcher
- Advanced Imaging Research Service, University of Texas Southwestern, Dallas, TX, USA
| | - Xiarong Fu
- Advanced Imaging Research Service, University of Texas Southwestern, Dallas, TX, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shawn C Burgess
- Advanced Imaging Research Service, University of Texas Southwestern, Dallas, TX, USA
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA.,Harry S. Truman Memorial Veterans Hospital-Research Service, Columbia, MO, USA
| | - John P Thyfault
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,Kansas City VA Medical Center-Research Service, Kansas City, MO, USA
| |
Collapse
|
28
|
The Combination of Blueberry Juice and Probiotics Ameliorate Non-Alcoholic Steatohepatitis (NASH) by Affecting SREBP-1c/PNPLA-3 Pathway via PPAR-α. Nutrients 2017; 9:nu9030198. [PMID: 28264426 PMCID: PMC5372861 DOI: 10.3390/nu9030198] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 12/19/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is liver inflammation and a major threat to public health. Several pharmaceutical agents have been used for NASH therapy but their high-rate side effects limit the use. Blueberry juice and probiotics (BP) have anti-inflammation and antibacterial properties, and may be potential candidates for NASH therapy. To understand the molecular mechanism, Sprague Dawley rats were used to create NASH models and received different treatments. Liver tissues were examined using HE (hematoxylin and eosin) and ORO (Oil Red O) stain, and serum biochemical indices were measured. The levels of peroxisome proliferators-activated receptor (PPAR)-α, sterol regulatory element binding protein-1c (SREBP-1c), Patatin-like phospholipase domain-containing protein 3 (PNPLA-3), inflammatory cytokines and apoptosis biomarkers in liver tissues were measured by qRT-PCR and Western blot. HE and ORO analysis indicated that the hepatocytes were seriously damaged with more and larger lipid droplets in NASH models while BP reduced the number and size of lipid droplets (p < 0.05). Meanwhile, BP increased the levels of SOD (superoxide dismutase), GSH (reduced glutathione) and HDL-C (high-density lipoprotein cholesterol), and reduced the levels of AST (aspartate aminotransferase), ALT (alanine aminotransferase), TG (triglycerides), LDL-C (low-density lipoprotein cholesterol) and MDA (malondialdehyde) in NASH models (p < 0.05). BP increased the level of PPAR-α (Peroxisome proliferator-activated receptor α), and reduced the levels of SREBP-1c (sterol regulatory element binding protein-1c) and PNPLA-3 (Patatin-like phospholipase domain-containing protein 3) (p < 0.05). BP reduced hepatic inflammation and apoptosis by affecting IL-6 (interleukin 6), TNF-α (Tumor necrosis factor α), caspase-3 and Bcl-2 in NASH models. Furthermore, PPAR-α inhibitor increased the level of SREBP-1c and PNPLA-3. Therefore, BP prevents NASH progression by affecting SREBP-1c/PNPLA-3 pathway via PPAR-α.
Collapse
|
29
|
Role of Uric Acid Metabolism-Related Inflammation in the Pathogenesis of Metabolic Syndrome Components Such as Atherosclerosis and Nonalcoholic Steatohepatitis. Mediators Inflamm 2016; 2016:8603164. [PMID: 28070145 PMCID: PMC5192336 DOI: 10.1155/2016/8603164] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/03/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
Uric acid (UA) is the end product of purine metabolism and can reportedly act as an antioxidant. However, recently, numerous clinical and basic research approaches have revealed close associations of hyperuricemia with several disorders, particularly those comprising the metabolic syndrome. In this review, we first outline the two molecular mechanisms underlying inflammation occurrence in relation to UA metabolism; one is inflammasome activation by UA crystallization and the other involves superoxide free radicals generated by xanthine oxidase (XO). Importantly, recent studies have demonstrated the therapeutic or preventive effects of XO inhibitors against atherosclerosis and nonalcoholic steatohepatitis, which were not previously considered to be related, at least not directly, to hyperuricemia. Such beneficial effects of XO inhibitors have been reported for other organs including the kidneys and the heart. Thus, a major portion of this review focuses on the relationships between UA metabolism and the development of atherosclerosis, nonalcoholic steatohepatitis, and related disorders. Although further studies are necessary, XO inhibitors are a potentially novel strategy for reducing the risk of many forms of organ failure characteristic of the metabolic syndrome.
Collapse
|
30
|
Carbohydrate and protein intake and risk of ulcerative colitis: Systematic review and dose-response meta-analysis of epidemiological studies. Clin Nutr 2016; 36:1259-1265. [PMID: 27776925 DOI: 10.1016/j.clnu.2016.10.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIM Dietary carbohydrate and protein intake is generally thought as risk factors for onset of ulcerative colitis (UC), while epidemiological data had been controversial. This study aimed to evaluate the role of carbohydrate and protein intake in the development of UC. METHODS Comprehensive search in PubMed and Embase was conducted to identify all relevant studies, and the role of carbohydrate and protein intake in the development of UC was quantitatively assessed by dose-response meta-analysis. RESULTS Nine studies (5 case-control and 4 prospective cohort) were identified with a total of 975 UC cases and 239352 controls. The summary relative risks (RR) for per 10 g increment/day were 1.005 (95%CI: 0.991-1.019, I2 = 31.5%, n = 5) for total carbohydrate intake, 1.001 (95%CI: 0.971-1.032, I2 = 0.0%, n = 7) for the subtype of fiber intake, 1.029 (95%CI: 0.962-1.101, I2 = 68.9%, n = 2) for the subtype of sugar intake, and 1.010 (95%CI: 0.975-1.047, I2 = 12.4%, n = 7) for total protein intake. Among sugar subtypes, only sucrose intake was found positively related with UC risk (RR for per 10 g increment/day: 1.098, 95%CI: 1.024-1.177, I2 = 0.0%, n = 3). No evidence of a non-linear dose-response association was found between the nutrient intake and UC risk, except for the subtype of sucrose (P for non-linear trend = 0.032). Subgroup analyses showed consistent results. CONCLUSIONS This meta-analysis suggested a lack of association between dietary carbohydrate or protein intake and the risk of UC, except for the subtype of sucrose which played a significant role in the development of UC. Large-scale prospective designed studies are needed to confirm our findings.
Collapse
|
31
|
Ng DS. Exploring cellular cholesterol-focused strategies in the treatment of cardiometabolic diseases. Curr Opin Lipidol 2016; 27:426-8. [PMID: 27383284 DOI: 10.1097/mol.0000000000000324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Dominic S Ng
- aKeenan Research Center, Li Ka Shing Knowledge Institute, St Michael's Hospital bDepartment of Medicine cDepartment of Physiology and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|