351
|
Abstract
Mammals undergo regular cycles of fasting and feeding that engage dynamic transcriptional responses in metabolic tissues. Here we review advances in our understanding of the gene regulatory networks that contribute to hepatic responses to fasting and feeding. The advent of sequencing and -omics techniques have begun to facilitate a holistic understanding of the transcriptional landscape and its plasticity. We highlight transcription factors, their cofactors, and the pathways that they impact. We also discuss physiological factors that impinge on these responses, including circadian rhythms and sex differences. Finally, we review how dietary modifications modulate hepatic gene expression programs.
Collapse
Affiliation(s)
- Lara Bideyan
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Rohith Nagari
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA.,Department of Biological Chemistry, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
352
|
Negi CK, Khan S, Dirven H, Bajard L, Bláha L. Flame Retardants-Mediated Interferon Signaling in the Pathogenesis of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms22084282. [PMID: 33924165 PMCID: PMC8074384 DOI: 10.3390/ijms22084282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a growing concern worldwide, affecting 25% of the global population. NAFLD is a multifactorial disease with a broad spectrum of pathology includes steatosis, which gradually progresses to a more severe condition such as nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and eventually leads to hepatic cancer. Several risk factors, including exposure to environmental toxicants, are involved in the development and progression of NAFLD. Environmental factors may promote the development and progression of NAFLD by various biological alterations, including mitochondrial dysfunction, reactive oxygen species production, nuclear receptors dysregulation, and interference in inflammatory and immune-mediated signaling. Moreover, environmental contaminants can influence immune responses by impairing the immune system’s components and, ultimately, disease susceptibility. Flame retardants (FRs) are anthropogenic chemicals or mixtures that are being used to inhibit or delay the spread of fire. FRs have been employed in several household and outdoor products; therefore, human exposure is unavoidable. In this review, we summarized the potential mechanisms of FRs-associated immune and inflammatory signaling and their possible contribution to the development and progression of NAFLD, with an emphasis on FRs-mediated interferon signaling. Knowledge gaps are identified, and emerging pharmacotherapeutic molecules targeting the immune and inflammatory signaling for NAFLD are also discussed.
Collapse
Affiliation(s)
- Chander K. Negi
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
- Correspondence: or
| | - Sabbir Khan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA;
| | - Hubert Dirven
- Department of Environmental Health, Section for Toxicology and Risk Assessment, Norwegian Institute of Public Health, 0456 Oslo, Norway;
| | - Lola Bajard
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
| | - Luděk Bláha
- Faculty of Science, RECETOX, Masaryk University, Kamenice 5, CZ62500 Brno, Czech Republic; (L.B.); (L.B.)
| |
Collapse
|
353
|
Raffaele M, Kovacovicova K, Frohlich J, Lo Re O, Giallongo S, Oben JA, Faldyna M, Leva L, Giannone AG, Cabibi D, Vinciguerra M. Mild exacerbation of obesity- and age-dependent liver disease progression by senolytic cocktail dasatinib + quercetin. Cell Commun Signal 2021; 19:44. [PMID: 33832488 PMCID: PMC8034117 DOI: 10.1186/s12964-021-00731-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is increasingly prevalent and represents a growing challenge in terms of prevention and treatment. A minority of affected patients develops inflammation, subsequently fibrosis, cirrhosis and hepatocellular carcinoma (HCC). HCC is a leading cause of cancer-related death. An increased number of senescent cells correlate with age-related tissue degeneration during NAFLD-induced HCC. Senolytics are promising agents that target selectively senescent cells. Previous studies showed that whereas a combination of the senolytic drugs dasatinib and quercetin (D + Q) reduced NAFLD in mice, D + Q lacked efficacy in removing doxorubicin-induced β-gal-positive senescent cells in human HCC xenografted mice. Whether D + Q has an effect on the age-associated spectrum of NAFLD-inflammation-HCC remains unknown. METHODS Here, we utilized an established model of age- and obesity-associated HCC, the low dose diethylnitrosamine (DEN)/high fat diet (HFD), a regimen promoting liver inflammation and tumorigenesis over a long period of 9 months. Four groups of mice each were created: group 1 included control untreated mice; group 2 included mice treated with D + Q; group 3 included mice undergoing the DEN/HFD protocol; group 4 included mice undergoing the DEN/HFD protocol with the administration of D + Q. At the end of the chemical/dietary regimen, we analyzed liver damage and cell senescence by histopathology, qPCR and immunoblotting approaches. RESULTS Unexpectedly, D + Q worsened liver disease progression in the DEN/HFD mouse model, slightly increasing histological damage and tumorigenesis, while having no effect on senescent cells removal. CONCLUSIONS In summary, using an animal model that fully recapitulates NAFLD, we demonstrate that these compounds are ineffective against age-associated NAFLD-induced HCC. Video Abstract.
Collapse
Affiliation(s)
- Marco Raffaele
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Kristina Kovacovicova
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Jan Frohlich
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Oriana Lo Re
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Sebastiano Giallongo
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jude A. Oben
- Institute for Liver and Digestive Health (ILDH), Division of Medicine, University College London (UCL), London, UK
| | | | - Lenka Leva
- Veterinary Research Institute, Brno, Czech Republic
| | - Antonino Giulio Giannone
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Pathologic Anatomy Unit-University of Palermo, Palermo, Italy
| | - Daniela Cabibi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Pathologic Anatomy Unit-University of Palermo, Palermo, Italy
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Institute for Liver and Digestive Health (ILDH), Division of Medicine, University College London (UCL), London, UK
- ERA Chair in Translational Stem Cell Biology, Medical University of Varna, Varna, Bulgaria
| |
Collapse
|
354
|
Ghosh S, Börsch A, Ghosh S, Zavolan M. The transcriptional landscape of a hepatoma cell line grown on scaffolds of extracellular matrix proteins. BMC Genomics 2021; 22:238. [PMID: 33823809 PMCID: PMC8025518 DOI: 10.1186/s12864-021-07532-2] [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: 08/24/2020] [Accepted: 03/14/2021] [Indexed: 11/10/2022] Open
Abstract
Background The behavior of cells in vivo is complex and highly dynamic, as it results from an interplay between intercellular matrix proteins with surface receptors and other microenvironmental cues. Although the effects of the cellular niche have been investigated for a number of cell types using different molecular approaches, comprehensive assessments of how the global transcriptome responds to 3D scaffolds composed of various extracellular matrix (ECM) constituents at different concentrations are still lacking. Results In this study, we explored the effects of two diverse extracellular matrix (ECM) components, Collagen I and Matrigel, on the transcriptional profile of cells in a cell culture system. Culturing Huh-7 cells on traditional cell culture plates (Control) or on the ECM components at different concentrations to modulate microenvironment properties, we have generated transcriptomics data that may be further explored to understand the differentiation and growth potential of this cell type for the development of 3D cultures. Our analysis infers transcription factors that are most responsible for the transcriptome response to the extracellular cues. Conclusion Our data indicates that the Collagen I substrate induces a robust transcriptional response in the Huh-7 cells, distinct from that induced by Matrigel. Enhanced hepatocyte markers (ALB and miR-122) reveal a potentially robust remodelling towards primary hepatocytes. Our results aid in defining the appropriate culture and transcription pathways while using hepatoma cell lines. As systems mimicking the in vivo structure and function of liver cells are still being developed, our study could potentially circumvent bottlenecks of limited availability of primary hepatocytes for preclinical studies of drug targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07532-2.
Collapse
Affiliation(s)
- Souvik Ghosh
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Anastasiya Börsch
- Biozentrum, University of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Mihaela Zavolan
- Biozentrum, University of Basel, Basel, Switzerland. .,Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| |
Collapse
|
355
|
NIMAKO C, IKENAKA Y, OKAMATSU-OGURA Y, BARIUAN JV, KOBAYASHI A, YAMAZAKI R, TAIRA K, HOSHI N, HIRANO T, NAKAYAMA SMM, ISHIZUKA M. Chronic low-dose exposure to imidacloprid potentiates high fat diet-mediated liver steatosis in C57BL/6J male mice. J Vet Med Sci 2021; 83:487-500. [PMID: 33487623 PMCID: PMC8025430 DOI: 10.1292/jvms.20-0479] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Hepatic steatosis is known to precede a continuum of events that lead to hepatic metabolic dysfunction, inflammation and carcinogenesis. Recently, studies have linked xenobiotic exposures to hepatic steatogenesis and its associated metabolic disorders; however, the underlying mechanisms remain elusive. This study aimed to elucidate the mechanistic role of imidacloprid in the prevalence of high fat diet (HFD)-induced liver steatosis, using a C57BL/6J mice model. Mice (3 weeks old) were fed with HFD and treated with 0.6 mg/kg bw/day (one-tenth of the NOAEL) of imidacloprid through water or diet, for 24 weeks. In a controlled group, mice were fed with only HFD. At the end of the study, imidacloprid treatment significantly potentiated HFD-induced body weight gain in mice. Also, imidacloprid increased the liver weights of mice, with complimentary reductions in mesenteric and gonadal white adipose tissue weights. Histopathological analysis of liver revealed a drastic steatosis in imidacloprid treated mice. Following a real-time qPCR analysis, imidacloprid upregulated transcriptions of hepatic fatty acid biosynthesis-related transcription factors and genes. Imidacloprid also induced hepatic expression of the gene encoding pregnane X receptor; but had no significant effect on hepatic expressions of liver X receptor and aryl hydrocarbon receptor. The imidacloprid treatment further enhanced serum alanine aminotransferase levels but downregulated hepatic antioxidant mRNA expressions. Ultimately, this study suggested an imidacloprid-potentiation effects on prevalence of HFD-induced liver steatosis via transcriptional modulations of the hepatic FA biosynthesis pathway.
Collapse
Affiliation(s)
- Collins NIMAKO
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University,
Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Yoshinori IKENAKA
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University,
Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, 11 Hoffman Street, Potchefstroom
2531, South Africa
| | - Yuko OKAMATSU-OGURA
- Laboratory of Biochemistry, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18,
Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Jussiaea V. BARIUAN
- Laboratory of Biochemistry, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18,
Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Atsushi KOBAYASHI
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita
18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Ryo YAMAZAKI
- Laboratory of Comparative Pathology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita
18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Kumiko TAIRA
- Department of Anesthesiology, Tokyo Women’s Medical University Center East, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666,
Japan
| | - Nobuhiko HOSHI
- Department of Animal Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, Hyogo
657-8501, Japan
| | - Tetsushi HIRANO
- Division of Drug and Structure Research, Life Science Research Center, University of Toyama, Sugitani 2630, Toyama 930-0194,
Japan
| | - Shouta M. M. NAKAYAMA
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University,
Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| | - Mayumi ISHIZUKA
- Laboratory of Toxicology, Department of Environmental Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University,
Kita 18, Nishi 9, Kita-ku, Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
356
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common liver pathology worldwide due to the rising prevalence of obesity. This term includes changes from simple steatosis to steatohepatitis and fibrosis. It was previously thought to be a hepatic manifestation of metabolic syndrome, but recent literature describes this relation as much more complex and bi-directional. Development of NAFLD is associated with other metabolic syndrome components but it can also exacerbate insulin resistance and increase cardiovascular risk. Recently a lot of attention is brought to the role of lipids and lipotoxicity in pathogenesis and progression of non-alcoholic fatty disease. It seems that some lipid classes can be protective against liver injury while others are harmful in excessive amounts. This study presents an overview of the main lipids involved in the pathogenesis of non-alcoholic fatty liver disease and summarizes their association with lipotoxicity, insulin resistance, oxidative stress and other processes responsible for its progression.
Collapse
|
357
|
Bissig-Choisat B, Alves-Bezerra M, Zorman B, Ochsner SA, Barzi M, Legras X, Yang D, Borowiak M, Dean AM, York RB, Galvan NTN, Goss J, Lagor WR, Moore DD, Cohen DE, McKenna NJ, Sumazin P, Bissig KD. A human liver chimeric mouse model for non-alcoholic fatty liver disease. JHEP Rep 2021; 3:100281. [PMID: 34036256 PMCID: PMC8138774 DOI: 10.1016/j.jhepr.2021.100281] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Background & Aims The accumulation of neutral lipids within hepatocytes underlies non-alcoholic fatty liver disease (NAFLD), which affects a quarter of the world's population and is associated with hepatitis, cirrhosis, and hepatocellular carcinoma. Despite insights gained from both human and animal studies, our understanding of NAFLD pathogenesis remains limited. To better study the molecular changes driving the condition we aimed to generate a humanised NAFLD mouse model. Methods We generated TIRF (transgene-free Il2rg -/-/Rag2 -/-/Fah -/-) mice, populated their livers with human hepatocytes, and fed them a Western-type diet for 12 weeks. Results Within the same chimeric liver, human hepatocytes developed pronounced steatosis whereas murine hepatocytes remained normal. Unbiased metabolomics and lipidomics revealed signatures of clinical NAFLD. Transcriptomic analyses showed that molecular responses diverged sharply between murine and human hepatocytes, demonstrating stark species differences in liver function. Regulatory network analysis indicated close agreement between our model and clinical NAFLD with respect to transcriptional control of cholesterol biosynthesis. Conclusions These NAFLD xenograft mice reveal an unexpected degree of evolutionary divergence in food metabolism and offer a physiologically relevant, experimentally tractable model for studying the pathogenic changes invoked by steatosis. Lay summary Fatty liver disease is an emerging health problem, and as there are no good experimental animal models, our understanding of the condition is poor. We here describe a novel humanised mouse system and compare it with clinical data. The results reveal that the human cells in the mouse liver develop fatty liver disease upon a Western-style fatty diet, whereas the mouse cells appear normal. The molecular signature (expression profiles) of the human cells are distinct from the mouse cells and metabolic analysis of the humanised livers mimic the ones observed in humans with fatty liver. This novel humanised mouse system can be used to study human fatty liver disease.
Collapse
Key Words
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- CBPEGs, cholesterol biosynthesis pathway enzyme genes
- CE, cholesteryl ester
- CER, ceramide
- CHHs, chimeric human hepatocytes
- CMHs, chimeric mouse hepatocytes
- CT, confidence transcript
- DAG, diacylglycerol
- DCER, dihydroceramide
- DEG, differentially expressed gene
- FA, fatty acid
- FAH, fumarylacetoacetate hydrolase
- FFA, free fatty acid
- GGT, gamma-glutamyl transpeptidase
- HCC, hepatocellular carcinoma
- HCER, hexosylceramide
- HCT, high confidence transcriptional target
- Human disease modelling
- Humanised mice
- LCER, lactosylceramide
- LPC, lysophosphatidylcholine
- LPE, lysophosphatidylethanolamine
- Lipid metabolism
- MAG, monoacylglycerol
- MUFA, monounsaturated fatty acid
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- NC, normal chow
- NTBC, nitisinone
- Non-alcoholic fatty liver disease
- PC, phosphatidylcholine
- PE, phosphatidylethanolamine
- PI, phosphatidylinositol
- PNPLA3, patatin-like-phospholipase domain-containing protein 3
- PUFA, polyunsaturated free FA
- SM, sphingomyelin
- SREBP, sterol regulatory element-binding protein
- Steatosis
- TAG, triacylglycerol
- TIRF, transgene-free Il2rg-/-/Rag2-/-/Fah-/-
- WD, Western-type diet
- hALB, human albumin
Collapse
Affiliation(s)
| | - Michele Alves-Bezerra
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Barry Zorman
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Mercedes Barzi
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Xavier Legras
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Diane Yang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Malgorzata Borowiak
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Institute for Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz Universtiy, Poznan, Poland
| | - Adam M. Dean
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Robert B. York
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - John Goss
- Department of Surgery, Texas Children’s Hospital, Houston, TX, USA
| | - William R. Lagor
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - David D. Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David E. Cohen
- Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Karl-Dimiter Bissig
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA
- Y.T. and Alice Chen Pediatric Genetics and Genomics Research Center, Duke University, Durham, NC, USA
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
- Corresponding author. Address: Duke University, Division of Medical Genetics, 905 South LaSalle street, Durham, NC-27708, USA. Tel.: +1 919 660 0761; fax: +1 919 660 0762.
| |
Collapse
|
358
|
Kalyesubula M, Mopuri R, Asiku J, Rosov A, Yosefi S, Edery N, Bocobza S, Moallem U, Dvir H. High-dose vitamin B1 therapy prevents the development of experimental fatty liver driven by overnutrition. Dis Model Mech 2021; 14:dmm.048355. [PMID: 33608323 PMCID: PMC7988776 DOI: 10.1242/dmm.048355] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Fatty liver is an abnormal metabolic condition of excess intrahepatic fat. This condition, referred to as hepatic steatosis, is tightly associated with chronic liver disease and systemic metabolic morbidity. The most prevalent form in humans, i.e. non-alcoholic fatty liver, generally develops due to overnutrition and sedentary lifestyle, and has as yet no approved drug therapy. Previously, we have developed a relevant large-animal model in which overnourished sheep raised on a high-calorie carbohydrate-rich diet develop hyperglycemia, hyperinsulinemia, insulin resistance, and hepatic steatosis. Here, we tested the hypothesis that treatment with thiamine (vitamin B1) can counter the development of hepatic steatosis driven by overnutrition. Remarkably, the thiamine-treated animals presented with completely normal levels of intrahepatic fat, despite consuming the same amount of liver-fattening diet. Thiamine treatment also decreased hyperglycemia and increased the glycogen content of the liver, but it did not improve insulin sensitivity, suggesting that steatosis can be addressed independently of targeting insulin resistance. Thiamine increased the catalytic capacity for hepatic oxidation of carbohydrates and fatty acids. However, at gene-expression levels, more-pronounced effects were observed on lipid-droplet formation and lipidation of very-low-density lipoprotein, suggesting that thiamine affects lipid metabolism not only through its known classic coenzyme roles. This discovery of the potent anti-steatotic effect of thiamine may prove clinically useful in managing fatty liver-related disorders.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Mugagga Kalyesubula
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel.,Department of Animal Science, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Ramgopal Mopuri
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Jimmy Asiku
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel.,Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel
| | - Alexander Rosov
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Sara Yosefi
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Nir Edery
- Pathology Laboratory, Kimron Veterinary Institute, Veterinary Services, Rishon LeZion 50250, Israel
| | - Samuel Bocobza
- Institute of Plant Sciences, Volcani Center - ARO, Rishon LeZion 7528809, Israel
| | - Uzi Moallem
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| | - Hay Dvir
- Institute of Animal Science, Volcani Center - Agricultural Research Organization (ARO), Rishon LeZion 7528809, Israel
| |
Collapse
|
359
|
Che L, Ren B, Jia Y, Dong Y, Wang Y, Shan J, Wang Y. Feprazone Displays Antiadipogenesis and Antiobesity Capacities in in Vitro 3 T3-L1 Cells and in Vivo Mice. ACS OMEGA 2021; 6:6674-6680. [PMID: 33748580 PMCID: PMC7970497 DOI: 10.1021/acsomega.0c05470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/09/2021] [Indexed: 05/14/2023]
Abstract
BACKGROUND AND PURPOSE Excessive lipid accumulation in adipose tissues and deregulation of adipogenesis-induced obesity affect millions of people worldwide. Feprazone, a nonsteroidal anti-inflammatory drug, has a wide clinical use. However, it is unknown whether Feprazone possesses an antiadipogenic ability. The aim of this study is to investigate whether Feprazone possesses an antiadipogenic ability in 3 T3-L1 cells and an antiobesity capacity in mouse models. METHODS An MTT assay was used to determine the optimized incubation concentrations of Feprazone in 3 T3-L1 cells. The lipid accumulation was evaluated using Oil Red O staining. The concentrations of triglyceride and glycerol release were detected to check the lipolysis during 3 T3-L1 adipogenesis. A quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the expressions of sterol regulatory element-binding protein-1C (SREBP-1C) and fatty acid binding protein 4 (FABP4) in treated cells. The expressions of peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT/enhancer-binding protein α (C/EBP-α), adipose triglyceride lipase (ATGL), and aquaporin-7 (AQP-7) were detected using qRT-PCR and Western blot analysis. After the high-fat diet (HFD) mice were treated with Feprazone, the pathological state of adipocyte tissues was evaluated using HE staining. The adipocyte size, visceral adipocyte tissue weight, and bodyweights were recorded. RESULTS According to the proliferation result, 30 and 60 μM Feprazone were used as the optimized concentrations of Feprazone. In the in vitro study, lipid accumulation, elevated production of triglycerides, the release of glycerol, upregulated SREBP-1C, FABP4, PPAR-γ, and C/EBP-α and downregulated ATGL and AQP-7 in the 3 T3-L1 adipocytes induced by the adipocyte differentiation cocktail medium were significantly reversed by treatment with Feprazone. In the in vivo experiment, we found that the increased adipocyte size, visceral adipocyte tissue weight, and body weights induced by HFD feeding in mice were significantly suppressed by the administration of Feprazone. CONCLUSION Feprazone might display anti-adipogenic and antiobesity capacities in in vitro 3 T3-L1 cells and in vivo mice.
Collapse
Affiliation(s)
- Liqun Che
- Department
of Endocrinology Ward 3, The Third Affiliated
Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Bo Ren
- Department
of Endocrinology Ward 3, The Third Affiliated
Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Yuanyuan Jia
- Department
of Endocrinology Ward 3, The Third Affiliated
Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Yujia Dong
- Department
of Endocrinology Ward 3, The Third Affiliated
Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Yanbing Wang
- Department
of Endocrinology Ward 3, The Third Affiliated
Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Jie Shan
- Department
of Endocrinology Ward 3, The Third Affiliated
Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Yuchun Wang
- Department
of pharmacology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
- . Tel.: +86-452-2663370
| |
Collapse
|
360
|
Zhang J, Ling N, Lei Y, Peng M, Hu P, Chen M. Multifaceted Interaction Between Hepatitis B Virus Infection and Lipid Metabolism in Hepatocytes: A Potential Target of Antiviral Therapy for Chronic Hepatitis B. Front Microbiol 2021; 12:636897. [PMID: 33776969 PMCID: PMC7991784 DOI: 10.3389/fmicb.2021.636897] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/18/2021] [Indexed: 12/17/2022] Open
Abstract
Hepatitis B virus (HBV) is considered a “metabolic virus” and affects many hepatic metabolic pathways. However, how HBV affects lipid metabolism in hepatocytes remains uncertain yet. Accumulating clinical studies suggested that compared to non-HBV-infected controls, chronic HBV infection was associated with lower levels of serum total cholesterol and triglycerides and a lower prevalence of hepatic steatosis. In patients with chronic HBV infection, high ALT level, high body mass index, male gender, or old age was found to be positively correlated with hepatic steatosis. Furthermore, mechanisms of how HBV infection affected hepatic lipid metabolism had also been explored in a number of studies based on cell lines and mouse models. These results demonstrated that HBV replication or expression induced extensive and diverse changes in hepatic lipid metabolism, by not only activating expression of some critical lipogenesis and cholesterolgenesis-related proteins but also upregulating fatty acid oxidation and bile acid synthesis. Moreover, increasing studies found some potential targets to inhibit HBV replication or expression by decreasing or enhancing certain lipid metabolism-related proteins or metabolites. Therefore, in this article, we comprehensively reviewed these publications and revealed the connections between clinical observations and experimental findings to better understand the interaction between hepatic lipid metabolism and HBV infection. However, the available data are far from conclusive, and there is still a long way to go before clarifying the complex interaction between HBV infection and hepatic lipid metabolism.
Collapse
Affiliation(s)
- Jiaxuan Zhang
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ning Ling
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Lei
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mingli Peng
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Chen
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
361
|
Rasineni K, Jordan CW, Thomes PG, Kubik JL, Staab EM, Sweeney SA, Talmon GA, Donohue TM, McNiven MA, Kharbanda KK, Casey CA. Contrasting Effects of Fasting on Liver-Adipose Axis in Alcohol-Associated and Non-alcoholic Fatty Liver. Front Physiol 2021; 12:625352. [PMID: 33746771 PMCID: PMC7966527 DOI: 10.3389/fphys.2021.625352] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/02/2021] [Indexed: 01/15/2023] Open
Abstract
Background: Fatty liver, a major health problem worldwide, is the earliest pathological change in the progression of alcohol-associated (AFL) and non-alcoholic fatty liver disease (NAFL). Though the causes of AFL and NAFL differ, both share similar histological and some common pathophysiological characteristics. In this study, we sought to examine mechanisms responsible for lipid dynamics in liver and adipose tissue in the setting of AFL and NAFL in response to 48 h of fasting. Methods: Male rats were fed Lieber-DeCarli liquid control or alcohol-containing diet (AFL model), chow or high-fat pellet diet (NAFL model). After 6-8 weeks of feeding, half of the rats from each group were fasted for 48 h while the other half remained on their respective diets. Following sacrifice, blood, adipose, and the liver were collected for analysis. Results: Though rats fed AFL and NAFL diets both showed fatty liver, the physiological mechanisms involved in the development of each was different. Here, we show that increased hepatic de novo fatty acid synthesis, increased uptake of adipose-derived free fatty acids, and impaired triglyceride breakdown contribute to the development of AFL. In the case of NAFL, however, increased dietary fatty acid uptake is the major contributor to hepatic steatosis. Likewise, the response to starvation in the two fatty liver disease models also varied. While there was a decrease in hepatic steatosis after fasting in ethanol-fed rats, the control, chow and high-fat diet-fed rats showed higher levels of hepatic steatosis than pair-fed counterparts. This diverse response was a result of increased adipose lipolysis in all experimental groups except fasted ethanol-fed rats. Conclusion: Even though AFL and NAFL are nearly histologically indistinguishable, the physiological mechanisms that cause hepatic fat accumulation are different as are their responses to starvation.
Collapse
Affiliation(s)
- Karuna Rasineni
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Clayton W. Jordan
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Paul G. Thomes
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Jacy L. Kubik
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
| | - Elizabeth M. Staab
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sarah A. Sweeney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Geoffrey A. Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Terrence M. Donohue
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology and the Center for Digestive Diseases, Mayo Clinic, Rochester, MN, United States
| | - Kusum K. Kharbanda
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Carol A. Casey
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
362
|
Chleilat F, Schick A, Reimer RA. Microbiota Changes in Fathers Consuming a High Prebiotic Fiber Diet Have Minimal Effects on Male and Female Offspring in Rats. Nutrients 2021; 13:820. [PMID: 33801321 PMCID: PMC8001975 DOI: 10.3390/nu13030820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Consuming a diet high in prebiotic fiber has been associated with improved metabolic and gut microbial parameters intergenerationally, although studies have been limited to maternal intake with no studies examining this effect in a paternal model. METHOD Male Sprague Dawley rats were allocated to either (1) control or (2) oligofructose-supplemented diet for nine weeks and then mated. Offspring consumed control diet until 16 weeks of age. Bodyweight, body composition, glycemia, hepatic triglycerides, gastrointestinal hormones, and gut microbiota composition were measured in fathers and offspring. RESULTS Paternal energy intake was reduced, while satiety inducing peptide tyrosine tyrosine (PYY) gut hormone was increased in prebiotic versus control fathers. Increased serum PYY persisted in female prebiotic adult offspring. Hepatic triglycerides were decreased in prebiotic fathers with a similar trend (p = 0.07) seen in female offspring. Gut microbial composition showed significantly reduced alpha diversity in prebiotic fathers at 9 and 12 weeks of age (p < 0.001), as well as concurrent differences in beta diversity (p < 0.001), characterized by differences in Bifidobacteriaceae, Lactobacillaceae and Erysipelotrichaceae, and particularly Bifidobacterium animalis. Female prebiotic offspring had higher alpha diversity at 3 and 9 weeks of age (p < 0.002) and differences in beta diversity at 15 weeks of age (p = 0.04). Increases in Bacteroidetes in female offspring and Christensenellaceae in male offspring were seen at nine weeks of age. CONCLUSIONS Although paternal prebiotic intake before conception improves metabolic and microbiota outcomes in fathers, effects on offspring were limited with increased serum satiety hormone levels and changes to only select gut bacteria.
Collapse
Affiliation(s)
- Faye Chleilat
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Alana Schick
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Raylene A. Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
363
|
Abstract
PURPOSE OF REVIEW Type 2 diabetes mellitus is widespread throughout the world and is a powerful risk factor for the development of atherosclerotic cardiovascular disease (ASCVD). This manuscript explored the mechanisms underlying dyslipidemia in type 2 diabetes as well as currently available treatment options and guideline recommendations. RECENT FINDINGS Type 2 diabetes is associated with a characteristic pattern of dyslipidemia, often termed diabetic dyslipidemia. Patients with type 2 diabetes often present with low HDL levels, elevated levels of small dense LDL particles, and elevated triglyceride levels. LDL lowering is the cornerstone of managing diabetic dyslipidemia, and statins are the mainstay of therapy. The cholesterol absorption inhibitor ezetimibe and PCSK9 inhibitors have also been shown to lower risk in patients with diabetes. Recently, the eicosapentaenoic (EPA) only n-3 fatty acid, icosapent ethyl, has also shown benefit for cardiovascular risk reduction in patients with diabetes. To date, no agents targeting HDL increase have shown cardiovascular benefit in patients on background statin therapy. Diabetic dyslipidemia is significant cardiovascular disease risk factor, and LDL-lowering therapy with statins, PCSK9 inhibitors, and ezetimibe continues to be mainstay therapy to reduce cardiovascular risk. Future studies targeting low HDL and high triglycerides levels associated with type 2 diabetes could provide additional novel therapies to manage diabetic dyslipidemia.
Collapse
|
364
|
Vilar-Gomez E, Gawrieh S, Liang T, McIntyre AD, Hegele RA, Chalasani N. Interrogation of selected genes influencing serum LDL-Cholesterol levels in patients with well characterized NAFLD. J Clin Lipidol 2021; 15:275-291. [PMID: 33454241 PMCID: PMC8187295 DOI: 10.1016/j.jacl.2020.12.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/07/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The clinical significance of rare mutations in LDL metabolism genes on nonalcoholic fatty liver disease (NAFLD) severity is not well understood. OBJECTIVE To examine the significance of mutations in LDL metabolism genes including apolipoprotein B (APOB), proprotein convertase subtilisin kexin 9 (PCSK9) and LDL receptor (LDLR) in patients with NAFLD. METHODS Patients with biopsy-confirmed NAFLD from the NASH Clinical Research Network studies were stratified into 3 groups of LDL-C (≤50 mg/dL, 130-150 mg/dL, ≥ 190 mg/dL) and then 120 (40 per group) were randomly selected from the strata. We examined the presence of mutations on LDL genes and analyzed its association with selected NAFLD-related features. Multivariable analyses were adjusted for age, race, gender and use of statins. RESULTS Among 40 patients with LDL-C ≤ 50 mg/dL, 7 (18%) patients had heterozygous variants in APOB and 2 had heterozygous variants in PCSK9 (5%). We also found heterozygous mutations in 3 (8%) patients with LDL-C ≥ 190 mg/dL; 2 and 1 located in LDLR and APOE genes, respectively. Compared to wild-type controls with LDL-C ≤ 50, APOB carriers displayed higher levels of alanine aminotransferase (85.86 ± 35.14 U/L vs 45.61 ± 20.84 U/L, Adj. P = 0.002) and steatosis >66% (57% vs 24%, Adj. P = 0.050). These associations remained statistically significant after excluding statin users. Other histological features of NAFLD severity were not different between wild-type controls and APOB mutation carriers. CONCLUSION Mutations in the APOB gene are common among NAFLD patients with very low LDL-C and may be associated with increased aminotransferase levels and steatosis severity.
Collapse
Affiliation(s)
- Eduardo Vilar-Gomez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Samer Gawrieh
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tiebing Liang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Adam D McIntyre
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Naga Chalasani
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
365
|
Heyde I, Begemann K, Oster H. Contributions of white and brown adipose tissues to the circadian regulation of energy metabolism. Endocrinology 2021; 162:6102571. [PMID: 33453099 PMCID: PMC7864004 DOI: 10.1210/endocr/bqab009] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/17/2022]
Abstract
The term energy metabolism comprises the entirety of chemical processes associated with uptake, conversion, storage, and breakdown of nutrients. All these must be tightly regulated in time and space to ensure metabolic homeostasis in an environment characterized by cycles such as the succession of day and night. Most organisms evolved endogenous circadian clocks to achieve this goal. In mammals, a ubiquitous network of cellular clocks is coordinated by a pacemaker residing in the hypothalamic suprachiasmatic nucleus. Adipocytes harbor their own circadian clocks, and large aspects of adipose physiology are regulated in a circadian manner through transcriptional regulation of clock-controlled genes. White adipose tissue (WAT) stores energy in the form of triglycerides at times of high energy levels that then serve as fuel in times of need. It also functions as an endocrine organ, releasing factors in a circadian manner to regulate food intake and energy turnover in other tissues. Brown adipose tissue (BAT) produces heat through nonshivering thermogenesis, a process also controlled by the circadian clock. We here review how WAT and BAT contribute to the circadian regulation of energy metabolism. We describe how adipose rhythms are regulated by the interplay of systemic signals and local clocks and summarize how adipose-originating circadian factors feed-back on metabolic homeostasis. The role of adipose tissue in the circadian control of metabolism becomes increasingly clear as circadian disruption leads to alterations in adipose tissue regulation, promoting obesity and its sequelae. Stabilizing adipose tissue rhythms, in turn, may help to combat disrupted energy homeostasis and obesity.
Collapse
Affiliation(s)
- Isabel Heyde
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
- Correspondence: Henrik Oster, PhD, Institute of Neurobiology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
366
|
Kurtz R, Anderman MF, Shepard BD. GPCRs get fatty: the role of G protein-coupled receptor signaling in the development and progression of nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2021; 320:G304-G318. [PMID: 33205999 PMCID: PMC8202238 DOI: 10.1152/ajpgi.00275.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by the abnormal deposition of lipids within the liver not due to alcohol consumption, is a growing epidemic affecting over 30% of the United States population. Both simple fatty liver and its more severe counterpart, nonalcoholic steatohepatitis, represent one of the most common forms of liver disease. Recently, several G protein-coupled receptors have emerged as targets for therapeutic intervention for these disorders. These include those with known hepatic function as well as those involved in global metabolic regulation. In this review, we highlight these emerging therapeutic targets, focusing on several common themes including their activation by microbial metabolites, stimulatory effect on insulin and incretin secretion, and contribution to glucose tolerance. The overlap in ligands, localization, and downstream effects of activation indicate the interdependent nature of these receptors and highlight the importance of this signaling family in the development and prevention of NAFLD.
Collapse
Affiliation(s)
- Ryan Kurtz
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Meghan F. Anderman
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Blythe D. Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia
| |
Collapse
|
367
|
Esteve-Luque V, Padró-Miquel A, Fanlo-Maresma M, Corbella E, Corbella X, Pintó X, Candás-Estébanez B. Implication between Genetic Variants from APOA5 and ZPR1 and NAFLD Severity in Patients with Hypertriglyceridemia. Nutrients 2021; 13:nu13020552. [PMID: 33567543 PMCID: PMC7914661 DOI: 10.3390/nu13020552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/28/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Lipid metabolism disorders, especially hypertriglyceridemia (HTG), are risk factors for non-alcoholic fatty liver disease (NAFLD). However, the association between genetic factors related to HTG and the risk of NAFLD has been scarcely studied. Methods: A total of 185 subjects with moderate HTG were prospectively included. We investigated the association between genetic factors’ (five allelic variants with polygenic hypertriglyceridemia) clinical and biochemical biomarkers with NAFLD severity. The five allelic variants’ related clinical and biochemical data of HTG were studied in all the subjects. NAFLD was assessed by abdominal ultrasound and patients were divided into two groups, one with no or mild NAFLD and another with moderate/severe NAFLD. Results: Patients with moderate/severe NAFLD had higher weight and waist values and a higher prevalence of insulin resistance than patients with no or mild NAFLD. Moderate/severe NAFLD was independently associated with APOA5 rs3134406 and ZPR1 rs964184 variants, and also showed a significant inverse relationship with lipoprotein(a) [Lp(a)] concentrations. Conclusions: APOA5 rs3135506 and ZPR1 rs964184 variants and lipoprotein(a) are associated with moderate/severe NAFLD. This association was independent of body weight, insulin resistance, and other factors related to NAFLD.
Collapse
Affiliation(s)
- Virginia Esteve-Luque
- Cardiovascular Risk Unit, Internal Medicine Department, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (V.E.-L.); (M.F.-M.); (E.C.); (X.C.)
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- Medicine and Translational Research, University of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Ariadna Padró-Miquel
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- Clinical Laboratory, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Fanlo-Maresma
- Cardiovascular Risk Unit, Internal Medicine Department, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (V.E.-L.); (M.F.-M.); (E.C.); (X.C.)
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- Medicine and Translational Research, University of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Emili Corbella
- Cardiovascular Risk Unit, Internal Medicine Department, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (V.E.-L.); (M.F.-M.); (E.C.); (X.C.)
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Xavier Corbella
- Cardiovascular Risk Unit, Internal Medicine Department, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (V.E.-L.); (M.F.-M.); (E.C.); (X.C.)
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- School of Medicine, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | - Xavier Pintó
- Cardiovascular Risk Unit, Internal Medicine Department, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (V.E.-L.); (M.F.-M.); (E.C.); (X.C.)
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- School of Medicine, Universitat de Barcelona, 08907 Barcelona, Spain
- Correspondence:
| | - Beatriz Candás-Estébanez
- Bellvitge Biomedical Research Institute (IDIBELL), 08908 L’Hospitalet de Llobregat, Spain; (A.P.-M.); (B.C.-E.)
- Clinical Laboratory, Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain
- CIBEROBN Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Clinical Biochemistry, SCIAS-Hospital de Barcelona, 08034 Barcelona, Spain
| |
Collapse
|
368
|
Buzatto AZ, Malkawi A, Sabi EM, Mujamammi AH, Li L, Abdel Rahman AM. Tissue Lipidomic Alterations Induced by Prolonged Dexamethasone Treatment. J Proteome Res 2021; 20:1558-1570. [PMID: 33557525 DOI: 10.1021/acs.jproteome.0c00759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dexamethasone is a synthetic glucocorticoid medication vastly used to treat abnormal immune responses and inflammation. Although the medication is well-established in the medical community, the prolonged treatment with high dosages of dexamethasone may lead to severe adverse effects through mechanisms that are not yet well-known. Lipids are a large class of hydrophobic molecules involved in energy storage, signaling, modulation of gene expression, and membranes. Hence, untargeted lipidomics may help unravel the biochemical alterations following prolonged treatment with high dosages of dexamethasone. We performed comprehensive lipidomic analyses of brain, heart, kidney, liver, and muscle samples obtained from rats that were treated with intramuscular injections of dexamethasone for 14 weeks compared to healthy controls. The employed methodology and statistical analysis showed that phosphatidic acids, glycerophospholipids, plasmalogens, and fatty acids are deeply affected by prolonged use of the medication. Brain tissue was only mildly affected, but skeletal muscle showed a strong accumulation of lipids that may be correlated with alterations in the energy metabolism, myopathy, and oxidative processes. This work provides new insights into the mechanisms of action and adverse effects for one of the most commonly prescribed class of drugs in the world.
Collapse
Affiliation(s)
| | - Abeer Malkawi
- Department of Chemistry, University of Quebec at Montreal, Montreal, QC H3C3P8, Canada
| | - Essa M Sabi
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh 11211, Saudi Arabia
| | - Ahmed H Mujamammi
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh 11211, Saudi Arabia
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Anas M Abdel Rahman
- Metabolomics Section, Clinical Genomics, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia.,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada
| |
Collapse
|
369
|
Deng Z, Gao S, An Y, Huang Y, Liu H, Zhu W, Lu W, He M, Xie W, Yu D, Li Y. Effects of earthworm extract on the lipid profile and fatty liver induced by a high-fat diet in guinea pigs. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:292. [PMID: 33708919 PMCID: PMC7944315 DOI: 10.21037/atm-20-5362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Non-alcoholic fatty liver disease (NAFLD), characterized by the accumulation of excess fat in the liver in people who consume little or no alcohol, is becoming increasingly common around the world, especially in developed countries. Extracts from earthworms have been used as alternative therapies for a variety of diseases but not in NAFLD. Therefore, the aim of this study was to investigate the effect of earthworm extract (EE) on diet-induced fatty liver disease in guinea pigs. Methods EE was extracted, and the effect of EE on the lipid levels and liver damage in guinea pigs fed a high-fat diet (HFD) was assessed. Thirty male guinea pigs at 3 weeks of age were allocated equally to five groups, namely, chow diet, HFD, and HFD with different dosages (0.3, 1.4 and 6.8 µg per kg bodyweight per day) of EE for 4 weeks, and their body weight was monitored throughout the experiment. Liver tissues were examined for gross morphology and histology. Serum levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), alanine transaminase (ALT) and aspartate aminotransferase (AST) were determined using an autoanalyser. Results HFD induced NAFLD in guinea pigs. HFD-fed guinea pigs that received EE treatment showed milder increases in the serum levels of TC, TG and LDL-C, as well as in the body weight growth rate, compared to the HFD group without EE supplementation. EE intervention reduced the number of lipid-containing hepatocytes, hepatocellular ballooning and sinusoidal distortion in the liver in HFD-fed animals. ALT in serum was significantly elevated by HFD. No statistically significant difference in ALT levels was found between the chow diet group and the HFD group with EE treatment. Conclusions This study demonstrates that the administration of EE suppressed the induction of serum TC, TG and LDL-C in response to HFD. EE also reduced liver damage in HFD-fed guinea pigs. These findings suggest that EE has alleviating effects on dyslipidaemia and liver damage associated with NAFLD.
Collapse
Affiliation(s)
- Zhenhan Deng
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,School of Medicine, Shenzhen University, Shenzhen, China
| | - Shanshan Gao
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yunfei An
- Department of Animal Science, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yong Huang
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Haifeng Liu
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Weimin Zhu
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wei Lu
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Miao He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Wenqing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Dengjie Yu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
370
|
He D, Yan L, Hu Y, Wu Q, Wu M, Choi JI, Tong H. Optimization of Porphyran Extraction from Pyropia yezoensis by Response Surface Methodology and Its Lipid-Lowering Effects. Mar Drugs 2021; 19:53. [PMID: 33498781 PMCID: PMC7911723 DOI: 10.3390/md19020053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/28/2022] Open
Abstract
Macroalgae polysaccharides are phytochemicals that are beneficial to human health. In this study, response surface methodology was applied to optimize the extraction procedure of Pyropia yezoensis porphyran (PYP). The optimum extraction parameters were: 100 °C (temperature), 120 min (time), and 29.32 mL/g (liquid-solid ratio), and the maximum yield of PYP was 22.15 ± 0.55%. The physicochemical characteristics of PPYP, purified from PYP, were analyzed, along with its lipid-lowering effect, using HepG2 cells and Drosophila melanogaster larvae. PPYP was a β-type sulfated hetero-rhamno-galactan-pyranose with a molecular weight of 151.6 kDa and a rhamnose-to-galactose molar ratio of 1:5.3. The results demonstrated that PPYP significantly reduced the triglyceride content in palmitic acid (PA)-induced HepG2 cells and high-sucrose-fed D. melanogaster larvae by regulating the expression of lipid metabolism-related genes, reducing lipogenesis and increasing fatty acid β-oxidation. To summarize, PPYP can lower lipid levels in HepG2 cells and larval fat body (the functional homolog tissue of the human liver), suggesting that PPYP may be administered as a potential marine lipid-lowering drug.
Collapse
Affiliation(s)
- Dan He
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (D.H.); (L.Y.); (Y.H.); (Q.W.)
- Department of Biotechnology and Bioengineering, Chonnam National University, Gwangju 61186, Korea
| | - Liping Yan
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (D.H.); (L.Y.); (Y.H.); (Q.W.)
| | - Yingxia Hu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (D.H.); (L.Y.); (Y.H.); (Q.W.)
| | - Qifang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (D.H.); (L.Y.); (Y.H.); (Q.W.)
| | - Mingjiang Wu
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (D.H.); (L.Y.); (Y.H.); (Q.W.)
| | - Jong-il Choi
- Department of Biotechnology and Bioengineering, Chonnam National University, Gwangju 61186, Korea
| | - Haibin Tong
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China; (D.H.); (L.Y.); (Y.H.); (Q.W.)
| |
Collapse
|
371
|
Tian YD, Chung MH, Quan QL, Lee DH, Kim EJ, Chung JH. UV-Induced Reduction of ACVR1C Decreases SREBP1 and ACC Expression by the Suppression of SMAD2 Phosphorylation in Normal Human Epidermal Keratinocytes. Int J Mol Sci 2021; 22:ijms22031101. [PMID: 33499275 PMCID: PMC7865598 DOI: 10.3390/ijms22031101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 01/14/2023] Open
Abstract
Activin A receptor type 1C (ACVR1C), a type I transforming growth factor-β (TGF-β) receptor, has been implicated in sensitive skin and psoriasis and is involved in the regulation of metabolic homeostasis as well as cell proliferation and differentiation. In this study, we identified a novel role of ACVR1C in the ultraviolet (UV)-irradiation-induced reduction of epidermal lipogenesis in human skin. UV irradiation decreased ACVR1C expression and epidermal triglyceride (TG) synthesis in human skin in vivo and in primary normal human epidermal keratinocytes (NHEK) in vitro. Lipogenic genes, including genes encoding acetyl-CoA carboxylase (ACC) and sterol regulatory element binding protein-1 (SREBP1), were significantly downregulated in UV-irradiated NHEK. ACVR1C knockdown by shRNA resulted in greater decreases in SREBP1 and ACC in response to UV irradiation. Conversely, the overexpression of ACVR1C attenuated the UV-induced decreases in SREBP1 and ACC. Further mechanistic study revealed that SMAD2 phosphorylation mediated the ACVR1C-induced lipogenic gene modulation. Taken together, a decrease in ACVR1C may cause UV-induced reductions in SREBP1 and ACC as well as epidermal TG synthesis via the suppression of SMAD2 phosphorylation. ACVR1C may be a target for preventing or treating UV-induced disruptions in lipid metabolism and associated skin disorders.
Collapse
Affiliation(s)
- Yu-Dan Tian
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (Q.-L.Q.); (D.H.L.)
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea;
- Department of Dermatology, Seoul National University Hospital, Seoul 03080, Korea
| | - Min Hwa Chung
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea;
| | - Qing-Ling Quan
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (Q.-L.Q.); (D.H.L.)
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea;
- Department of Dermatology, Seoul National University Hospital, Seoul 03080, Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (Q.-L.Q.); (D.H.L.)
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea;
- Department of Dermatology, Seoul National University Hospital, Seoul 03080, Korea
| | - Eun Ju Kim
- Department of Dermatology, Seoul National University Hospital, Seoul 03080, Korea
- Correspondence: (E.J.K.); (J.H.C.)
| | - Jin Ho Chung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea;
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Korea; (Q.-L.Q.); (D.H.L.)
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, Korea;
- Department of Dermatology, Seoul National University Hospital, Seoul 03080, Korea
- Institute on Aging, Seoul National University, Seoul 03080, Korea
- Correspondence: (E.J.K.); (J.H.C.)
| |
Collapse
|
372
|
Systemic Overexpression of GDF5 in Adipocytes but Not Hepatocytes Alleviates High-Fat Diet-Induced Nonalcoholic Fatty Liver in Mice. Can J Gastroenterol Hepatol 2021; 2021:8894685. [PMID: 33542911 PMCID: PMC7843185 DOI: 10.1155/2021/8894685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Our recent study demonstrated that growth differentiation factor 5 (GDF5) could promote white adipose tissue thermogenesis and alleviate high-fat diet- (HFD-) induced obesity in fatty acid-binding protein 4- (Fabp4-) GDF5 transgenic mice (TG). Here, we further investigated the effects of systemic overexpression of the GDF5 gene in adipocytes HFD-induced nonalcoholic fatty liver disease (NAFLD). METHODS Fabp4-GDF5 TG mice were administered an HFD feeding. NAFLD-related indicators associated with lipid metabolism and inflammation were measured. A GDF5 lentiviral vector was constructed, and the LO2 NAFLD cell model was induced by FFA solution (oleic acid and palmitic acid). The alterations in liver function, liver lipid metabolism, and related inflammatory indicators were analyzed. RESULTS The liver weight was significantly reduced in the TG group, which was in accordance with the significantly downregulated expression of TNFα, MCP1, Aim2, and SREBP-1c and significantly upregulated expression of CPT-1α and ACOX2 in TG mouse livers. Compared to that of cells in the FAA-free control group, LO2 cells with in situ overexpression of GDF5 developed lipid droplets after FFA treatment; the levels of triglycerides, alanine aminotransferase (ALT), and aspartate aminotransferase (AST) were significantly increased in both the GDF5 lentivirus and control lentivirus groups compared with those of the FAA-free group. Additionally, the levels of FAS, SREBP-1, CPT-1α, and inflammation-associated genes, such as ASC and NLRC4, were unaltered despite GDF5 treatment. CONCLUSION Systemic overexpression of GDF5 in adipose tissue in vivo significantly reduced HFD-induced NAFLD liver damage in mice. The overexpression of GDF5 in hepatocytes failed to improve lipid accumulation and inflammation-related reactions induced by mixed fatty acids, suggesting that the protective effect of GDF5 in NAFLD was mainly due to the reduction in adipose tissue and improvements in metabolism. Hence, our study suggests that the management of NAFLD should be targeted to reduce the overall amount of body fat and improve metabolic status before the progression to nonalcoholic steatohepatitis occurs.
Collapse
|
373
|
Kalra S, Unnikrishnan AG, Baruah MP, Sahay R, Bantwal G. Metabolic and Energy Imbalance in Dysglycemia-Based Chronic Disease. Diabetes Metab Syndr Obes 2021; 14:165-184. [PMID: 33488105 PMCID: PMC7816219 DOI: 10.2147/dmso.s286888] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/02/2020] [Indexed: 12/16/2022] Open
Abstract
Metabolic flexibility is the ability to efficiently adapt metabolism based on nutrient availability and requirement that is essential to maintain homeostasis in times of either caloric excess or restriction and during the energy-demanding state. This regulation is orchestrated in multiple organ systems by the alliance of numerous metabolic pathways under the master control of the insulin-glucagon-sympathetic neuro-endocrine axis. This, in turn, regulates key metabolic enzymes and transcription factors, many of which interact closely with and culminate in the mitochondrial energy generation machinery. Metabolic flexibility is compromised due to the continuous mismatch between availability and intake of calorie-dense foods and reduced metabolic demand due to sedentary lifestyle and age-related metabolic slowdown. The resultant nutrient overload leads to mitochondrial trafficking of substrates manifesting as mitochondrial dysfunction characterized by ineffective substrate switching and incomplete substrate utilization. At the systemic level, the manifestation of metabolic inflexibility comprises reduced skeletal muscle glucose disposal rate, impaired suppression of hepatic gluconeogenesis and adipose tissue lipolysis manifesting as insulin resistance. This is compounded by impaired β-cell function and progressively reduced β-cell mass. A consequence of insulin resistance is the upregulation of the mitogen-activated protein kinase pathway leading to a pro-hypertensive, atherogenic, and thrombogenic environment. This is further aggravated by oxidative stress, advanced glycation end products, and inflammation, which potentiates the risk of micro- and macro-vascular complications. This review aims to elucidate underlying mechanisms mediating the onset of metabolic inflexibility operating at the main target organs and to understand the progression of metabolic diseases. This could potentially translate into a pharmacological tool that can manage multiple interlinked conditions of dysglycemia, hypertension, and dyslipidemia by restoring metabolic flexibility. We discuss the breadth and depth of metabolic flexibility and its impact on health and disease.
Collapse
Affiliation(s)
- Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, India
- Department of Endocrinology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | | | - Manash P Baruah
- Department of Endocrinology, Excel Hospitals, Guwahati, India
| | - Rakesh Sahay
- Department of Endocrinology, Osmania Medical College, Hyderabad, Telangana, India
| | - Ganapathi Bantwal
- Department of Endocrinology, St. John’s Medical College and Hospital, Bangalore, Karnataka, India
| |
Collapse
|
374
|
The potential ameliorative impacts of cerium oxide nanoparticles against fipronil-induced hepatic steatosis. Sci Rep 2021; 11:1310. [PMID: 33446707 PMCID: PMC7809457 DOI: 10.1038/s41598-020-79479-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Fipronil (FIP) is a phenylpyrazole insecticide that is commonly used in agricultural and veterinary fields for controlling a wide range of insects, but it is a strong environmentally toxic substance. Exposure to FIP has been reported to increase the hepatic fat accumulation through altered lipid metabolism, which ultimately can contribute to nonalcoholic fatty liver disease (NAFLD) development. The present study aimed to examine the function of cerium oxide nanoparticles (CeNPs) in protecting against hepatotoxicity and lipogenesis induced by FIP. Twenty-eight male albino rats were classified into four groups: FIP (5 mg/kg/day per os), CTR, CeNPs (35 mg/kg/day p.o.), and FIP + CeNPs (5 (FIP) + 35 (CeNPs) mg/kg/day p.o.) for 28 consecutive days. Serum lipid profiles, hepatic antioxidant parameters and pathology, and mRNA expression of adipocytokines were assessed. The results revealed that FIP increased cholesterol, height-density lipoprotein, triacylglyceride, low-density lipoprotein (LDL-c), and very-low-density lipoprotein (VLDL-c) concentrations. It also increased nitric oxide (NO) and malondialdehyde (MDA) hepatic levels and reduced glutathione peroxidase (GPx) and superoxide dismutase (SOD) enzyme activities. Additionally, FIP up-regulated the fatty acid-binding protein (FABP), acetyl Co-A carboxylase (ACC1), and peroxisome proliferator-activated receptor-alpha (PPAR-α). Immunohistochemically, a strong proliferation of cell nuclear antigen (PCNA), ionized calcium-binding adapter molecule 1 (Iba-1), cyclooxygenase-2 (COX-2) reactions in the endothelial cells of the hepatic sinusoids, and increased expression of caspase3 were observed following FIP intoxication. FIP also caused histological changes in hepatic tissue. The CeNPs counteracted the hepatotoxic effect of FIP exposure. So, this study recorded an ameliorative effect of CeNPs against FIP-induced hepatotoxicity.
Collapse
|
375
|
Kim JH, Matsubara T, Lee J, Fenollar-Ferrer C, Han K, Kim D, Jia S, Chang CJ, Yang H, Nagano T, Krausz KW, Yim SH, Gonzalez FJ. Lysosomal SLC46A3 modulates hepatic cytosolic copper homeostasis. Nat Commun 2021; 12:290. [PMID: 33436590 PMCID: PMC7804329 DOI: 10.1038/s41467-020-20461-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 12/02/2020] [Indexed: 01/05/2023] Open
Abstract
The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes hepatic toxicity associated with prominent lipid accumulation in humans. Here, the authors report that the lysosomal copper transporter SLC46A3 is induced by TCDD and underlies the hepatic lipid accumulation in mice, potentially via effects on mitochondrial function. SLC46A3 was localized to the lysosome where it modulated intracellular copper levels. Forced expression of hepatic SLC46A3 resulted in decreased mitochondrial membrane potential and abnormal mitochondria morphology consistent with lower copper levels. SLC46A3 expression increased hepatic lipid accumulation similar to the known effects of TCDD exposure in mice and humans. The TCDD-induced hepatic triglyceride accumulation was significantly decreased in Slc46a3-/- mice and was more pronounced when these mice were fed a high-fat diet, as compared to wild-type mice. These data are consistent with a model where lysosomal SLC46A3 induction by TCDD leads to cytosolic copper deficiency resulting in mitochondrial dysfunction leading to lower lipid catabolism, thus linking copper status to mitochondrial function, lipid metabolism and TCDD-induced liver toxicity.
Collapse
Affiliation(s)
- Jung-Hwan Kim
- Department of Pharmacology, School of Medicine, Institute of Health Sciences, Department of Convergence Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea.
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Tsutomu Matsubara
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Anatomy and Regenerative Biology, Osaka City University Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Jaekwon Lee
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Cristina Fenollar-Ferrer
- Laboratory of Molecular & Cellular Neurobiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kyungreem Han
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Donghwan Kim
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shang Jia
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Christopher J Chang
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Heejung Yang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Tomokazu Nagano
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Sumitomo Dainippon Pharma Co. Ltd., Osaka, Japan
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sun-Hee Yim
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Environmental Toxicology, Texas Tech University, Lubbock, TX, 41163, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
376
|
Jauregibeitia I, Portune K, Gaztambide S, Rica I, Tueros I, Velasco O, Grau G, Martín A, Castaño L, Larocca AV, Di Nolfo F, Ferreri C, Arranz S. Molecular Differences Based on Erythrocyte Fatty Acid Profile to Personalize Dietary Strategies between Adults and Children with Obesity. Metabolites 2021; 11:43. [PMID: 33435565 PMCID: PMC7827034 DOI: 10.3390/metabo11010043] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/30/2022] Open
Abstract
As the obesity epidemic continues to grow inexorably worldwide, the need to develop effective strategies to prevent and control obesity seems crucial. The use of molecular tools can be useful to characterize different obesity phenotypes to provide more precise nutritional recommendations. This study aimed to determine the fatty acid (FA) profile of red blood cell (RBC) membranes, together with the evaluation of their dietary intake and biochemical parameters, of children and adults with obesity. An observational study was carried out on 196 children (113 with normal weight and 83 with obesity) and 91 adults (30 with normal weight and 61 with obesity). Mature RBC membrane phospholipids were analyzed for FA composition by gas chromatography-mass spectrometry (GC-MS). Dietary habits were evaluated using validated food frequency questionnaires (FFQ). Children with obesity presented higher levels of ω-6 polyunsaturated FAs (mainly linoleic acid, p = 0.01) and lower values of ω-3 FAs (mainly DHA, p < 0.001) compared with adults. Regarding blood biochemical parameters, children with obesity presented lower levels of glucose, LDL cholesterol, and alanine aminotransferase compared with adults with obesity. These lipidomic differences could be considered to provide specific nutritional recommendations for different age groups, based on an adequate fat intake.
Collapse
Affiliation(s)
- Iker Jauregibeitia
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio–Bizkaia, Spain; (I.J.); (K.P.); (I.T.)
| | - Kevin Portune
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio–Bizkaia, Spain; (I.J.); (K.P.); (I.T.)
| | - Sonia Gaztambide
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM/CIBERER, UPV/EHU, Endo–ERN, 48903 Barakaldo, Spain; (S.G.); (I.R.); (O.V.); (G.G.); (A.M.); (L.C.)
| | - Itxaso Rica
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM/CIBERER, UPV/EHU, Endo–ERN, 48903 Barakaldo, Spain; (S.G.); (I.R.); (O.V.); (G.G.); (A.M.); (L.C.)
| | - Itziar Tueros
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio–Bizkaia, Spain; (I.J.); (K.P.); (I.T.)
| | - Olaia Velasco
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM/CIBERER, UPV/EHU, Endo–ERN, 48903 Barakaldo, Spain; (S.G.); (I.R.); (O.V.); (G.G.); (A.M.); (L.C.)
| | - Gema Grau
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM/CIBERER, UPV/EHU, Endo–ERN, 48903 Barakaldo, Spain; (S.G.); (I.R.); (O.V.); (G.G.); (A.M.); (L.C.)
| | - Alicia Martín
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM/CIBERER, UPV/EHU, Endo–ERN, 48903 Barakaldo, Spain; (S.G.); (I.R.); (O.V.); (G.G.); (A.M.); (L.C.)
| | - Luis Castaño
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, CIBERDEM/CIBERER, UPV/EHU, Endo–ERN, 48903 Barakaldo, Spain; (S.G.); (I.R.); (O.V.); (G.G.); (A.M.); (L.C.)
| | - Anna Vita Larocca
- Lipidomic Laboratory, Lipinutragen srl, Via di Corticella 181/4, 40128 Bologna, Italy; (A.V.L.); (F.D.N.)
| | - Federica Di Nolfo
- Lipidomic Laboratory, Lipinutragen srl, Via di Corticella 181/4, 40128 Bologna, Italy; (A.V.L.); (F.D.N.)
| | - Carla Ferreri
- Consiglio Nazionale delle Ricerche, ISOF, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Sara Arranz
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio–Bizkaia, Spain; (I.J.); (K.P.); (I.T.)
| |
Collapse
|
377
|
Arak H, Karimi Torshizi MA. Comparative consequences of two sources of aflatoxins in ducklings experimental aflatoxicosis. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2021; 12:305-311. [PMID: 34815841 PMCID: PMC8576148 DOI: 10.30466/vrf.2020.104547.2488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 01/08/2020] [Indexed: 11/19/2022]
Abstract
The purpose of this study was to investigate the effect of two aflatoxins (AFs) sources for experimental induction of aflatoxicosis in ducklings. Dietary supplementation of aflatoxin-contaminated grounded rice grains previously was cultured by Aspergillus parasiticus and dietary supplementation of aflatoxin methanolic extract of contaminated rice grains. A total number of 18 four-day-old ducklings were treated with varying sources of AFs. Treatments included: A: Control (basal diet without AFs), B: Contaminated feed with 0.20 mg kg-1 AFs (ground rice grains), C: Contaminated feed with 0.20 mg kg-1 AFs (methanol extract of contaminated rice grains). Results showed a significant decrease in serum levels of total protein, albumin, glucose, triglyceride, cholesterol, HDL, LDL and creatinine. The serum's hepatic enzymes levels were not affected in AFs-treated groups but lactate dehydrogenase (LDH) concentration increased by the two AFs sources. The relative weight of the liver and spleen was significantly increased in AFs-fed groups. Histopathological liver examination showed the vacuolar degeneration with small and large lipid droplets in hepatocytes in the AFs- contaminated groups. Dietary AFs resulted in a significant decrease of villus height, villus width and villus surface area of the small intestine compared to the control group. In conclusion, the results showed that the induction of experimental aflatoxicosis via two investigated AFs sources had slight differences concerning the studied parameters. It seems the group consumed ground rice grains indicated slightly fewer aflatoxicosis symptoms than the methanolic extract.
Collapse
Affiliation(s)
| | - Mohammad Amir Karimi Torshizi
- Correspondence Mohammad Amir Karimi Torshizi. PhD, Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran. E-mail:
| |
Collapse
|
378
|
Liao Q, Zhou Y, Xia L, Cao D. Lipid Metabolism and Immune Checkpoints. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:191-211. [PMID: 33740251 DOI: 10.1007/978-981-33-6785-2_12] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Immune checkpoints are essential for the regulation of immune cell functions. Although the abrogation of immunosurveillance of tumor cells is known, the regulators of immune checkpoints are not clear. Lipid metabolism is one of the important metabolic activities in organisms. In lipid metabolism, a large number of metabolites produced can regulate the gene expression and activation of immune checkpoints through various pathways. In addition, increasing evidence has shown that lipid metabolism leads to transient generation or accumulation of toxic lipids that result in endoplasmic reticulum (ER) stress and then regulate the transcriptional and posttranscriptional modifications of immune checkpoints, including transcription, protein folding, phosphorylation, palmitoylation, etc. More importantly, the lipid metabolism can also affect exosome transportation of checkpoints and the degradation of checkpoints by affecting ubiquitination and lysosomal trafficking. In this chapter, we mainly empathize on the roles of lipid metabolism in the regulation of immune checkpoints, such as gene expression, activation, and degradation.
Collapse
Affiliation(s)
- Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Deliang Cao
- Department of Medical Microbiology, Immunology and Cell Biology, Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
379
|
Agarwal R. Aging Liver and Interpretation of Liver Tests. GERIATRIC GASTROENTEROLOGY 2021:1329-1352. [DOI: 10.1007/978-3-030-30192-7_49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
380
|
The Regulation of Fat Metabolism During Aerobic Exercise. Biomolecules 2020; 10:biom10121699. [PMID: 33371437 PMCID: PMC7767423 DOI: 10.3390/biom10121699] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
Since the lipid profile is altered by physical activity, the study of lipid metabolism is a remarkable element in understanding if and how physical activity affects the health of both professional athletes and sedentary subjects. Although not fully defined, it has become clear that resistance exercise uses fat as an energy source. The fatty acid oxidation rate is the result of the following processes: (a) triglycerides lipolysis, most abundant in fat adipocytes and intramuscular triacylglycerol (IMTG) stores, (b) fatty acid transport from blood plasma to muscle sarcoplasm, (c) availability and hydrolysis rate of intramuscular triglycerides, and (d) transport of fatty acids through the mitochondrial membrane. In this review, we report some studies concerning the relationship between exercise and the aforementioned processes also in light of hormonal controls and molecular regulations within fat and skeletal muscle cells.
Collapse
|
381
|
Schiano E, Annunziata G, Ciampaglia R, Iannuzzo F, Maisto M, Tenore GC, Novellino E. Bioactive Compounds for the Management of Hypertriglyceridemia: Evidence From Clinical Trials and Putative Action Targets. Front Nutr 2020; 7:586178. [PMID: 33330588 PMCID: PMC7734325 DOI: 10.3389/fnut.2020.586178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/26/2020] [Indexed: 01/22/2023] Open
Abstract
Hypertriglyceridemia refers to the presence of elevated concentrations of triglycerides (TG) in the bloodstream (TG >200 mg/dL). This lipid alteration is known to be associated with an increased risk of atherosclerosis, contributing overall to the onset of atherosclerotic cardiovascular disease (CVD). Guidelines for the management of hypertriglyceridemia are based on both lifestyle intervention and pharmacological treatment, but poor adherence, medication-related costs and side effects can limit the success of these interventions. For this reason, the search for natural alternative approaches to reduce plasma TG levels currently represents a hot research field. This review article summarizes the most relevant clinical trials reporting the TG-reducing effect of different food-derived bioactive compounds. Furthermore, based on the evidence obtained from in vitro studies, we provide a description and classification of putative targets of action through which several bioactive compounds can exert a TG-lowering effect. Future research may lead to investigations of the efficacy of novel nutraceutical formulations consisting in a combination of bioactive compounds which contribute to the management of plasma TG levels through different action targets.
Collapse
Affiliation(s)
| | | | | | - Fortuna Iannuzzo
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Maria Maisto
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
382
|
Zou Y, Sheng G, Yu M, Xie G. The association between triglycerides and ectopic fat obesity: An inverted U-shaped curve. PLoS One 2020; 15:e0243068. [PMID: 33253312 PMCID: PMC7703893 DOI: 10.1371/journal.pone.0243068] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/14/2020] [Indexed: 02/07/2023] Open
Abstract
Background Ectopic fat obesity and triglycerides are risk factors for diabetes and multiple cardiovascular diseases. However, there have been limited studies on the association between triglycerides and ectopic fat obesity. The purpose of this study was to explore the association between triglycerides and ectopic fat obesity. Methods and results In this cross-sectional study, we retrospectively analyzed 15464 adult participants recruited by Murakami Memorial Hospital (8430 men and 7034 women, average age of 43.71 ± 8.90). All patients were divided into two groups according to the threshold used to diagnose hypertriglyceridemia. The logistic regression model was used to analyze the association between triglycerides and the risk of ectopic fat obesity, and the generalized additive model was used to identify the nonlinear association. In this study population, the prevalence of ectopic fat obesity was 17.73%. After adjusting other covariables, triglycerides were positively correlated with the risk of ectopic fat obesity (OR: 1.54, 95% CI:1.41–1.69, P<0.0001). Through smooth curve fitting, we found that there was an inverted U-shaped curve association between triglycerides and ectopic fat obesity. This association remained unchanged even if the adjusted covariables were removed from the model, and the inflection point of the curve was 3.98. When triglyceride levels were ≤3.98, triglycerides were positively correlated with the risk of ectopic fat obesity (OR:1.784, 95% CI:1.611–1.975, P<0.0001). When triglyceride levels were >3.98 (right side of the inflection point), there was a negative correlation (OR:0.519, 95% CI:0.333–0.810, P = 0.0039). Conclusions Our research showed that there is a significant association between triglycerides and ectopic fat obesity. This relation is not a simple linear relationship but instead an inverted U-shaped curve association.
Collapse
Affiliation(s)
- Yang Zou
- Medical Department of Graduate School, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi Province, China
| | - Guotai Sheng
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi Province, China
| | - Meng Yu
- Medical Department of Graduate School, Nanchang University, Nanchang, Jiangxi Province, China
| | - Guobo Xie
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, Jiangxi Province, China
- * E-mail:
| |
Collapse
|
383
|
Wang X, Song T, Sun Y, Men L, Gu Y, Zhang S, Chen X. Proteomic Analysis Reveals the Effect of Trichostatin A and Bone Marrow-Derived Dendritic Cells on the Fatty Acid Metabolism of NIH3T3 Cells under Oxygen-Glucose Deprivation Conditions. J Proteome Res 2020; 20:960-971. [PMID: 33226813 DOI: 10.1021/acs.jproteome.0c00713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fibroblasts mediate acute wound healing and long-term tissue remodeling with scarring after tissue injury. Following myocardial infarction (MI), necrotized cardiomyocytes become replaced by secreted extracellular matrix proteins produced by fibroblasts. Dendritic cells (DCs) can migrate from the bone marrow to the infarct areas and infarct border areas to mediate collagen accumulation after MI. Trichostatin A (TSA) is known to regulate apoptosis and proliferation in fibroblasts and affect the functions of DCs under oxygen-glucose deprivation (OGD) conditions. In this study, we used label-free quantitative proteomics to investigate the effects of TSA and bone marrow-derived dendritic cells (BMDCs) on NIH3T3 fibroblasts under OGD conditions. The results showed that the fatty acid degradation pathway was significantly upregulated in NIH3T3 cells under OGD conditions and that the fatty acid synthesis pathway was significantly downregulated in NIH3T3 cells treated with conditioned media (CM) from BMDCs treated with TSA under OGD conditions [BMDCs-CM(TSA)]. In addition, BMDCs-CM(TSA) significantly decreased the levels of triglycerides and free fatty acids and mediated fatty acid metabolism-related proteins in NIH3T3 cells under OGD conditions. In summary, this proteomics analysis showed that TSA and BMDCs affect fatty acid metabolism in NIH3T3 cells under OGD conditions.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Tongtong Song
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Yunpeng Sun
- Cardiac Surgery Department, The First Hospital of Jilin University, Changchun 130000, China
| | - Lihui Men
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Yiwen Gu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Siwei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130000, China
| |
Collapse
|
384
|
Bionaz M, Vargas-Bello-Pérez E, Busato S. Advances in fatty acids nutrition in dairy cows: from gut to cells and effects on performance. J Anim Sci Biotechnol 2020; 11:110. [PMID: 33292523 PMCID: PMC7667790 DOI: 10.1186/s40104-020-00512-8] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
High producing dairy cows generally receive in the diet up to 5-6% of fat. This is a relatively low amount of fat in the diet compared to diets in monogastrics; however, dietary fat is important for dairy cows as demonstrated by the benefits of supplementing cows with various fatty acids (FA). Several FA are highly bioactive, especially by affecting the transcriptome; thus, they have nutrigenomic effects. In the present review, we provide an up-to-date understanding of the utilization of FA by dairy cows including the main processes affecting FA in the rumen, molecular aspects of the absorption of FA by the gut, synthesis, secretion, and utilization of chylomicrons; uptake and metabolism of FA by peripheral tissues, with a main emphasis on the liver, and main transcription factors regulated by FA. Most of the advances in FA utilization by rumen microorganisms and intestinal absorption of FA in dairy cows were made before the end of the last century with little information generated afterwards. However, large advances on the molecular aspects of intestinal absorption and cellular uptake of FA were made on monogastric species in the last 20 years. We provide a model of FA utilization in dairy cows by using information generated in monogastrics and enriching it with data produced in dairy cows. We also reviewed the latest studies on the effects of dietary FA on milk yield, milk fatty acid composition, reproduction, and health in dairy cows. The reviewed data revealed a complex picture with the FA being active in each step of the way, starting from influencing rumen microbiota, regulating intestinal absorption, and affecting cellular uptake and utilization by peripheral tissues, making prediction on in vivo nutrigenomic effects of FA challenging.
Collapse
Affiliation(s)
- Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, 97331, USA.
| | - Einar Vargas-Bello-Pérez
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 3, DK-1870, Frederiksberg C, Denmark
| | - Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
385
|
Exploring the Role of a Novel Peptide from Allomyrina dichotoma Larvae in Ameliorating Lipid Metabolism in Obesity. Int J Mol Sci 2020; 21:ijms21228537. [PMID: 33198343 PMCID: PMC7698306 DOI: 10.3390/ijms21228537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to identify an anti-obesity peptide from Allomyrina dichotoma and investigate the lipid metabolic mechanism. Enzymatically hydrolyzed A. dichotoma larvae were further separated using tangential flow filtration and consecutive chromatographic processes. Finally, an anti-obesity peptide that showed the highest inhibitory effect on lipid accumulation was obtained, and the sequence was Glu-Ile-Ala-Gln-Asp-Phe-Lys-Thr-Asp-Leu (EIA10). EIA10 decreased lipid aggregation in vitro and significantly reduced the accumulation of body weight gain, liver weight, and adipose tissue weight in high-fat-fed mice. Compared with the control group, the levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL), insulin, and homeostasis model assessment of insulin resistance (HOMA-IR) in the high-fat diet (HFD) group increased significantly, and the content of high-density lipoprotein cholesterol (HDL) in the serum decreased significantly. On the contrary, the levels of TC, TG, and insulin in the EIA10 group decreased significantly, and the HDL content increased significantly compared with the HFD group. Additionally, EIA10 dramatically decreased mRNA and protein levels of transcription factors involved in lipid adipogenesis. Taken together, our results suggest that EIA10 could be a promising agent for the treatment and prevention of obesity.
Collapse
|
386
|
Krupenko NI, Sharma J, Pediaditakis P, Helke KL, Hall MS, Du X, Sumner S, Krupenko SA. Aldh1l2 knockout mouse metabolomics links the loss of the mitochondrial folate enzyme to deregulation of a lipid metabolism observed in rare human disorder. Hum Genomics 2020; 14:41. [PMID: 33168096 PMCID: PMC7654619 DOI: 10.1186/s40246-020-00291-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 10/14/2020] [Indexed: 12/29/2022] Open
Abstract
Background Mitochondrial folate enzyme ALDH1L2 (aldehyde dehydrogenase 1 family member L2) converts 10-formyltetrahydrofolate to tetrahydrofolate and CO2 simultaneously producing NADPH. We have recently reported that the lack of the enzyme due to compound heterozygous mutations was associated with neuro-ichthyotic syndrome in a male patient. Here, we address the role of ALDH1L2 in cellular metabolism and highlight the mechanism by which the enzyme regulates lipid oxidation. Methods We generated Aldh1l2 knockout (KO) mouse model, characterized its phenotype, tissue histology, and levels of reduced folate pools and applied untargeted metabolomics to determine metabolic changes in the liver, pancreas, and plasma caused by the enzyme loss. We have also used NanoString Mouse Inflammation V2 Code Set to analyze inflammatory gene expression and evaluate the role of ALDH1L2 in the regulation of inflammatory pathways. Results Both male and female Aldh1l2 KO mice were viable and did not show an apparent phenotype. However, H&E and Oil Red O staining revealed the accumulation of lipid vesicles localized between the central veins and portal triads in the liver of Aldh1l2-/- male mice indicating abnormal lipid metabolism. The metabolomic analysis showed vastly changed metabotypes in the liver and plasma in these mice suggesting channeling of fatty acids away from β-oxidation. Specifically, drastically increased plasma acylcarnitine and acylglycine conjugates were indicative of impaired β-oxidation in the liver. Our metabolomics data further showed that mechanistically, the regulation of lipid metabolism by ALDH1L2 is linked to coenzyme A biosynthesis through the following steps. ALDH1L2 enables sufficient NADPH production in mitochondria to maintain high levels of glutathione, which in turn is required to support high levels of cysteine, the coenzyme A precursor. As the final outcome, the deregulation of lipid metabolism due to ALDH1L2 loss led to decreased ATP levels in mitochondria. Conclusions The ALDH1L2 function is important for CoA-dependent pathways including β-oxidation, TCA cycle, and bile acid biosynthesis. The role of ALDH1L2 in the lipid metabolism explains why the loss of this enzyme is associated with neuro-cutaneous diseases. On a broader scale, our study links folate metabolism to the regulation of lipid homeostasis and the energy balance in the cell. Supplementary Information The online version contains supplementary material available at 10.1186/s40246-020-00291-3.
Collapse
Affiliation(s)
- Natalia I Krupenko
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA
| | - Jaspreet Sharma
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Peter Pediaditakis
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Kristi L Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Madeline S Hall
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Xiuxia Du
- Department of Bioinformatics & Genomics, UNC Charlotte, Charlotte, NC, USA
| | - Susan Sumner
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA
| | - Sergey A Krupenko
- Nutrition Research Institute, University of North Carolina, Chapel Hill, NC, USA. .,Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
387
|
Fang S, Li P, Zhu C, Han X, Bao P, Guo W. Research progress of ulinastatin in the treatment of liver diseases. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2720-2726. [PMID: 33284867 PMCID: PMC7716140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/22/2020] [Indexed: 06/12/2023]
Abstract
Ulinastatin (UTI) is a trypsin inhibitor observed in urine. UTI can treat some diseases by inhibiting the broad-spectrum hydrolysis activity of various enzymes and other pharmacological effects. UTI can widely treat pancreatitis, systemic multiple organ dysfunction syndrome, circulatory failure, and toxic shock clinically. The liver is a major metabolic organ of the human body. Various biological metabolic reactions require the liver's participation. When various physical and chemical factors drive the body, it will damage the liver to varying degrees. As a clinically effective drug, UTI is also known to treat some liver diseases. This article mainly describes UTI's research progress in treating septic liver injury, hepatitis, liver fibrosis, autoimmune liver disease with liver failure, and liver ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Shangping Fang
- School of Anaesthesia, Wannan Medical CollegeWuhu, Anhui, China
| | - Pengfei Li
- School of Anaesthesia, Wannan Medical CollegeWuhu, Anhui, China
| | - Chenxu Zhu
- School of Anaesthesia, Wannan Medical CollegeWuhu, Anhui, China
| | - Xiaoxiao Han
- School of Anaesthesia, Wannan Medical CollegeWuhu, Anhui, China
| | - Pengju Bao
- School of Anaesthesia, Wannan Medical CollegeWuhu, Anhui, China
| | - Wenjun Guo
- Department of Anesthesiology, Yi Jishan Hospital Affiliated to Wannan Medical CollegeWuhu, Anhui, China
| |
Collapse
|
388
|
Lei Y, Hoogerland JA, Bloks VW, Bos T, Bleeker A, Wolters H, Wolters JC, Hijmans BS, van Dijk TH, Thomas R, van Weeghel M, Mithieux G, Houtkooper RH, de Bruin A, Rajas F, Kuipers F, Oosterveer MH. Hepatic Carbohydrate Response Element Binding Protein Activation Limits Nonalcoholic Fatty Liver Disease Development in a Mouse Model for Glycogen Storage Disease Type 1a. Hepatology 2020; 72:1638-1653. [PMID: 32083759 PMCID: PMC7702155 DOI: 10.1002/hep.31198] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND AIMS Glycogen storage disease (GSD) type 1a is an inborn error of metabolism caused by defective glucose-6-phosphatase catalytic subunit (G6PC) activity. Patients with GSD 1a exhibit severe hepatomegaly due to glycogen and triglyceride (TG) accumulation in the liver. We have shown that the activity of carbohydrate response element binding protein (ChREBP), a key regulator of glycolysis and de novo lipogenesis, is increased in GSD 1a. In the current study, we assessed the contribution of ChREBP to nonalcoholic fatty liver disease (NAFLD) development in a mouse model for hepatic GSD 1a. APPROACH AND RESULTS Liver-specific G6pc-knockout (L-G6pc-/- ) mice were treated with adeno-associated viruses (AAVs) 2 or 8 directed against short hairpin ChREBP to normalize hepatic ChREBP activity to levels observed in wild-type mice receiving AAV8-scrambled short hairpin RNA (shSCR). Hepatic ChREBP knockdown markedly increased liver weight and hepatocyte size in L-G6pc-/- mice. This was associated with hepatic accumulation of G6P, glycogen, and lipids, whereas the expression of glycolytic and lipogenic genes was reduced. Enzyme activities, flux measurements, hepatic metabolite analysis and very low density lipoprotein (VLDL)-TG secretion assays revealed that hepatic ChREBP knockdown reduced downstream glycolysis and de novo lipogenesis but also strongly suppressed hepatic VLDL lipidation, hence promoting the storage of "old fat." Interestingly, enhanced VLDL-TG secretion in shSCR-treated L-G6pc-/- mice associated with a ChREBP-dependent induction of the VLDL lipidation proteins microsomal TG transfer protein and transmembrane 6 superfamily member 2 (TM6SF2), the latter being confirmed by ChIP-qPCR. CONCLUSIONS Attenuation of hepatic ChREBP induction in GSD 1a liver aggravates hepatomegaly because of further accumulation of glycogen and lipids as a result of reduced glycolysis and suppressed VLDL-TG secretion. TM6SF2, critical for VLDL formation, was identified as a ChREBP target in mouse liver. Altogether, our data show that enhanced ChREBP activity limits NAFLD development in GSD 1a by balancing hepatic TG production and secretion.
Collapse
Affiliation(s)
- Yu Lei
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Joanne A. Hoogerland
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Vincent W. Bloks
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Trijnie Bos
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Aycha Bleeker
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Henk Wolters
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Justina C. Wolters
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Brenda S. Hijmans
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Theo H. van Dijk
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Rachel Thomas
- Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic DiseasesAmsterdam Gastroenterology and MetabolismAmsterdam Cardiovascular SciencesAmsterdamthe Netherlands
- Core Facility of MetabolomicsAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Gilles Mithieux
- National Institute of Health and Medical Research, U1213LyonFrance
- University of LyonLyonFrance
- University of Lyon 1VilleurbanneFrance
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic DiseasesAmsterdam Gastroenterology and MetabolismAmsterdam Cardiovascular SciencesAmsterdamthe Netherlands
| | - Alain de Bruin
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Fabienne Rajas
- National Institute of Health and Medical Research, U1213LyonFrance
- University of LyonLyonFrance
- University of Lyon 1VilleurbanneFrance
| | - Folkert Kuipers
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Maaike H. Oosterveer
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| |
Collapse
|
389
|
Giglio RV, Carruba G, Cicero AF, Banach M, Patti AM, Nikolic D, Cocciadiferro L, Zarcone M, Montalto G, Stoian AP, Banerjee Y, Rizvi AA, Toth PP, Rizzo M. Pasta Supplemented with Opuntia ficus-indica Extract Improves Metabolic Parameters and Reduces Atherogenic Small Dense Low-Density Lipoproteins in Patients with Risk Factors for the Metabolic Syndrome: A Four-Week Intervention Study. Metabolites 2020; 10:metabo10110428. [PMID: 33114614 PMCID: PMC7694062 DOI: 10.3390/metabo10110428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
Food supplementation with Opuntia ficus-indica (OFI) has been associated with a significant reduction in total cholesterol, body fat, hyperglycemia and blood pressure. Since OFI may also have antioxidant and anti-atherogenic properties, we hypothesized that its supplementation might reduce atherogenic lipoproteins, including small, dense low-density lipoproteins (sdLDL). Forty-nine patients (13 men and 36 women, mean age: 56 ± 5 years) with one or two criteria for the metabolic syndrome weekly consumed 500 g of pasta supplemented with 3% OFI extract (30% of insoluble polysaccharides with high antioxidant power) for 1 month. The full LDL subclass profile was assessed by gel electrophoresis (Lipoprint, Quantimetrix, Redondo Beach, CA, USA). After 1 month of pasta supplementation, waist circumference (p = 0.0297), plasma glucose (p < 0.0001), triglycerides (p = 0.0137), plasma creatinine (p = 0.0244), urea and aspartate transaminase (p < 0.0001 for each) significantly decreased. A percentage increase in larger, less atherogenic LDL-1 (p = 0.0002), with a concomitant reduction in smaller, denser LDL-2 (p < 0.0001) and LDL-3 (p = 0.0004), were found. LDL-4 and-5 decreased, although not significantly. This is the first intervention study suggesting that pasta enriched with an OFI extract may have beneficial effects on some metabolic parameters and the LDL particle sizes, reducing atherogenic sdLDL. Future studies will help to establish if these findings impact cardiovascular outcomes.
Collapse
Affiliation(s)
- Rosaria Vincenza Giglio
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (A.M.P.); (G.M.); (M.R.)
| | - Giuseppe Carruba
- Division of Research and Internationalization, ARNAS-Civico Di Cristina e Benfratelli, 90127 Palermo, Italy; (G.C.); (L.C.); (M.Z.)
| | - Arrigo F.G. Cicero
- Medical and Surgical Sciences Department, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy;
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, 90-419 Lodz, Poland;
- Polish Mother’s Memorial Hospital Research Institute (PMMHRI) in Lodz, 93-338 Lodz, Poland
| | - Angelo Maria Patti
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (A.M.P.); (G.M.); (M.R.)
| | - Dragana Nikolic
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (A.M.P.); (G.M.); (M.R.)
- Correspondence: ; Tel.: +39-091-655-4703
| | - Letizia Cocciadiferro
- Division of Research and Internationalization, ARNAS-Civico Di Cristina e Benfratelli, 90127 Palermo, Italy; (G.C.); (L.C.); (M.Z.)
| | - Maurizio Zarcone
- Division of Research and Internationalization, ARNAS-Civico Di Cristina e Benfratelli, 90127 Palermo, Italy; (G.C.); (L.C.); (M.Z.)
| | - Giuseppe Montalto
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (A.M.P.); (G.M.); (M.R.)
| | - Anca Pantea Stoian
- Department of Diabetes, Nutrition and Metabolic Diseases, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Yajnavalka Banerjee
- Department of Biochemistry, Mohammed Bin Rashid University of Medicine and Health Sciences, 505055 Dubai, UAE;
| | - Ali A. Rizvi
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, SC 29203, USA;
- Division of Endocrinology, Metabolism, and Lipids, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Peter P. Toth
- CGH Medical Center, Sterling, IL 61081, USA;
- School of Medicine, University of Illinois, Peoria, IL 60612, USA
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Manfredi Rizzo
- Department of Health Promotion Sciences Maternal and Infantile Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (R.V.G.); (A.M.P.); (G.M.); (M.R.)
- Division of Endocrinology, Diabetes and Metabolism, University of South Carolina School of Medicine, Columbia, SC 29203, USA;
| |
Collapse
|
390
|
Boyce GR, Shoeb M, Kodali V, Meighan TG, Roach KA, McKinney W, Stone S, Powell MJ, Roberts JR, Zeidler-Erdely PC, Erdely A, Antonini JM. Welding fume inhalation exposure and high-fat diet change lipid homeostasis in rat liver. Toxicol Rep 2020; 7:1350-1355. [PMID: 33102138 PMCID: PMC7569188 DOI: 10.1016/j.toxrep.2020.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 01/21/2023] Open
Abstract
It is estimated that greater than 1 million workers are exposed to welding fume (WF) by inhalation daily. The potentially toxic metals found in WF are known to cause multiple adverse pulmonary and systemic effects, including cardiovascular disease, and these metals have also been shown to translocate to the liver. This occupational exposure combined with a high fat (HF) Western diet, which has been shown to cause hyperlipidemia and non-alcoholic fatty liver disease (NAFLD), has the potential to cause significant mixed exposure metabolic changes in the liver. The goal of this study was to use matrix assisted laser desorption ionization imaging mass spectrometry (MALDI-IMS) to analyze the spatial distribution and abundance changes of lipid species in Sprague Dawley rat liver maintained on a HF diet combined with WF inhalation. The results of the MALDI-IMS analysis revealed unique hepatic lipid profiles for each treatment group. The HF diet group had significantly increased abundance of triglycerides and phosphatidylinositol lipids, as well as decreased lysophosphatidic lipids and cardiolipin. Ceramide-1-phosphate was found at higher abundance in the regular (REG) diet WF-exposed group which has been shown to regulate the eicosanoid pathway involved in pro-inflammatory response. The results of this study showed that the combined effects of WF inhalation and a HF diet significantly altered the hepatic lipidome. Additionally, pulmonary exposure to WF alone increased lipid markers of inflammation.
Collapse
Affiliation(s)
- Greg R. Boyce
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
- Corresponding author.
| | - Mohammad Shoeb
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Vamsi Kodali
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Terence G. Meighan
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Katherine A. Roach
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Walter McKinney
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Samuel Stone
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | | | - Jenny R. Roberts
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | | | - Aaron Erdely
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - James M. Antonini
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| |
Collapse
|
391
|
Wang T, Zhao Y, You Z, Li X, Xiong M, Li H, Yan N. Endoplasmic Reticulum Stress Affects Cholesterol Homeostasis by Inhibiting LXRα Expression in Hepatocytes and Macrophages. Nutrients 2020; 12:nu12103088. [PMID: 33050595 PMCID: PMC7601278 DOI: 10.3390/nu12103088] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis (AS) is the most common cardiovascular disease, and reverse cholesterol transport (RCT) plays an important role in maintaining cholesterol homeostasis. Both endoplasmic reticulum (ER) stress and LXRα can affect the metabolism of cholesterol. However, whether ER stress can modulate cholesterol metabolism by LXRα in hepatocytes and macrophages remains unclear. Therefore, in this study, we aimed to explore the relationship between ER stress induced by tunicamycin and LXRα in hepatocytes and macrophages and clarify their possible mechanisms and roles in AS. C57BL/6 mice and Huh-7 and THP-1 cells were treated with tunicamycin and LXR-623 (an agonist of LXRα) alone or in combination. Tunicamycin-induced ER stress caused liver injury; promoted the accumulation of cholesterol and triglycerides; inhibited the expression of LXRα, ABCA1 and ABCG1 in the livers of mice, thus reducing serum high-density lipoprotein (HDL)-C, low-density lipoprotein (LDL)-C, total cholesterol and triglyceride levels; however, LXR-623 could attenuate ER stress and reverse these changes. We also obtained the same results in Huh-7 and THP-1 cells. ER stress induced by tunicamycin could clearly be reversed by activating LXRα because it promoted cholesterol efflux by enhancing the expression of ABCA1 and ABCG1 in hepatocytes and macrophages, contributing to attenuation of the development of AS.
Collapse
|
392
|
Li H, Li J, Qu Z, Qian H, Zhang J, Wang H, Xu X, Liu S. Intrauterine exposure to low-dose DBP in the mice induces obesity in offspring via suppression of UCP1 mediated ER stress. Sci Rep 2020; 10:16360. [PMID: 33004990 PMCID: PMC7529907 DOI: 10.1038/s41598-020-73477-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
Dibutyl phthalate (DBP) is recognized as an environmental endocrine disruptor that has been detected in fetal and postnatal samples. Recent evidence found that in utero DBP exposure was associated with an increase of adipose tissue weight and serum lipids in offspring, but the precise mechanism is unknown. Here we aimed to study the effects of in utero DBP exposure on obesity in offspring and examine possible mechanisms. SPF C57BL/6J pregnant mice were gavaged with either DBP (5 mg /kg/day) or corn oil, from gestational day 12 until postnatal day 7. After the offspring were weaned, the mice were fed a standard diet for 21 weeks, and in the last 2 weeks 20 mice were selected for TUDCA treatment. Intrauterine exposure to low-dose DBP promoted obesity in offspring, with evidence of glucose and lipid metabolic disorders and a decreased metabolic rate. Compared to controls, the DBP exposed mice had lower expression of UCP1 and significantly higher expression of Bip and Chop, known markers of endoplasmic reticulum (ER) stress. However, TUDCA treatment of DBP exposed mice returned these parameters nearly to the levels of the controls, with increased expression of UCP1, lower expression of Bip and Chop and ameliorated obesity. Intrauterine exposure of mice to low-dose DBP appears to promote obesity in offspring by inhibiting UCP1 via ER stress, a process that was largely reversed by treatment with TUDCA.
Collapse
Affiliation(s)
- Huan Li
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Jianqiao Li
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Zhenting Qu
- Jilin Combine Traditional Chinese and Western Hospital, Jilin, 132012, China
| | - Honghao Qian
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Jing Zhang
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Hongyan Wang
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Xiaolei Xu
- School of Public Health, Beihua University, Jilin, 132013, China
| | - Shengyuan Liu
- Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, 518054, China.
| |
Collapse
|
393
|
Liou CJ, Wu SJ, Shen SC, Chen LC, Chen YL, Huang WC. Phloretin ameliorates hepatic steatosis through regulation of lipogenesis and Sirt1/AMPK signaling in obese mice. Cell Biosci 2020; 10:114. [PMID: 33014333 PMCID: PMC7526428 DOI: 10.1186/s13578-020-00477-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022] Open
Abstract
Background Phloretin is isolated from apple trees and could increase lipolysis in 3T3-L1 adipocytes. Previous studies have found that phloretin could prevent obesity in mice. In this study, we investigated whether phloretin ameliorates non-alcoholic fatty liver disease (NAFLD) in high-fat diet (HFD)-induced obese mice, and evaluated the regulation of lipid metabolism in hepatocytes. Methods HepG2 cells were treated with 0.5 mM oleic acid to induce lipid accumulation, and then treated with phloretin to evaluate the molecular mechanism of lipogenesis. In another experiment, male C57BL/6 mice were fed normal diet or HFD (60% fat, w/w) for 16 weeks. After the fourth week, mice were treated with or without phloretin by intraperitoneal injection for 12 weeks. Results Phloretin significantly reduced excessive lipid accumulation and decreased sterol regulatory element-binding protein 1c, blocking the expression of fatty acid synthase in oleic acid-induced HepG2 cells. Phloretin increased Sirt1, and phosphorylation of AMP activated protein kinase to suppress acetyl-CoA carboxylase expression, reducing fatty acid synthesis in hepatocytes. Phloretin also reduced body weight and fat weight compared to untreated HFD-fed mice. Phloretin also reduced liver weight and liver lipid accumulation and improved hepatocyte steatosis in obese mice. In liver tissue from obese mice, phloretin suppressed transcription factors of lipogenesis and fatty acid synthase, and increased lipolysis and fatty acid β-oxidation. Furthermore, phloretin regulated serum leptin, adiponectin, triglyceride, low-density lipoprotein, and free fatty acid levels in obese mice. Conclusions These findings suggest that phloretin improves hepatic steatosis by regulating lipogenesis and the Sirt-1/AMPK pathway in the liver.
Collapse
Affiliation(s)
- Chian-Jiun Liou
- Department of Nursing, Division of Basic Medical Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33303 Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan City, 33303 Taiwan
| | - Shu-Ju Wu
- Department of Nutrition and Health Sciences, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33303 Taiwan.,Aesthetic Medical Center, Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan, 33303 Taiwan
| | - Szu-Chuan Shen
- Graduate Program of Nutrition Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei City, 11676 Taiwan
| | - Li-Chen Chen
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan City, 33303 Taiwan
| | - Ya-Ling Chen
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei City, 11031 Taiwan
| | - Wen-Chung Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Guishan Dist., Taoyuan City, 33303 Taiwan.,Graduate Institute of Health Industry Technology, Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, No.261, Wenhua 1st Rd., Guishan Dist., Taoyuan City, 33303 Taiwan
| |
Collapse
|
394
|
Lee HC, Shin SJ, Huang JK, Lin MY, Lin YH, Ke LY, Jiang HJ, Tsai WC, Chao MF, Lin YH. The role of postprandial very-low-density lipoprotein in the development of atrial remodeling in metabolic syndrome. Lipids Health Dis 2020; 19:210. [PMID: 32962696 PMCID: PMC7507670 DOI: 10.1186/s12944-020-01386-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/10/2020] [Indexed: 11/30/2022] Open
Abstract
Background Negatively charged very-low-density lipoprotein (VLDL-χ) in metabolic syndrome (MetS) patients exerts cytotoxic effects on endothelial cells and atrial myocytes. Atrial cardiomyopathy, manifested by atrial remodeling with a dilated diameter, contributes to atrial fibrillation pathogenesis and predicts atrial fibrillation development. The correlation of VLDL-χ with atrial remodeling is unknown. This study investigated the association between VLDL-χ and remodeling of left atrium. Methods Consecutively, 87 MetS and 80 non-MetS individuals between 23 and 74 years old (50.6% men) without overt cardiovascular diseases were included in the prospective cohort study. Blood samples were collected while fasting and postprandially (at 0.5, 1, 2, and 4 h after a unified meal). VLDL was isolated by ultracentrifugation; the percentile concentration of VLDL-χ (%) was determined by ultra-performance liquid chromatography. The correlations of left atrium diameter (LAD) with variables including VLDL-χ, LDL-C, HDL-C, triglycerides, glucose, and blood pressure, were analyzed by multiple linear regression models. A hierarchical linear model was conducted to test the independencies of each variable’s correlation with LAD. Results The mean LAD was 3.4 ± 0.5 cm in non-MetS subjects and 3.9 ± 0.5 cm in MetS patients (P < 0.01). None of the fasting lipid profiles were associated with LAD. VLDL-χ, BMI, waist circumference, hip circumference, and blood pressure were positively correlated with LAD (all P < 0.05) after adjustment for age and sex. Significant interactions between VLDL-χ and blood pressure, waist circumference, and hip circumference were observed. When adjusted for obesity- and blood pressure-related variables, 2-h postprandial VLDL-χ (mean 1.30 ± 0.61%) showed a positive correlation with LAD in MetS patients. Each 1% VLDL-χ increase was estimated to increase LAD by 0.23 cm. Conclusions Postprandial VLDL-χ is associated with atrial remodeling particularly in the MetS group. VLDL-χ is a novel biomarker and may be a therapeutic target for atrial cardiomyopathy in MetS patients. Trial registration ISRCTN 69295295. Retrospectively registered 9 June 2020.
Collapse
Affiliation(s)
- Hsiang-Chun Lee
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan. .,Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Institute/Center of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Shyi-Jang Shin
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jih-Kai Huang
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Yen Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Hsun Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Liang-Yin Ke
- Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Lipid Science and Aging Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - He-Jiun Jiang
- Department of Metabolism, Affiliated Hospital of Kaohsiung Medical University, Kaohsiung, Taiwan.,College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Min-Fang Chao
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsiung Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
395
|
Hepatic miR-192-3p reactivation alleviates steatosis by targeting glucocorticoid receptor. JHEP Rep 2020; 2:100179. [PMID: 33134908 PMCID: PMC7588854 DOI: 10.1016/j.jhepr.2020.100179] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/28/2020] [Accepted: 08/18/2020] [Indexed: 01/08/2023] Open
Abstract
Background & Aims The paradox of hepatic insulin resistance describes the inability for liver to respond to bioenergetics hormones in suppressing gluconeogenesis whilst maintaining lipid synthesis. Here, we report the deficiency of miR-192-3p in the livers of mice with diabetes and its role in alleviating hepatic steatosis. Methods As conventional pre-microRNA (miRNA) stem-loop overexpression only boosts guiding strand (i.e. miR-192-5p) expression, we adopted an artificial AAV(DJ)-directed, RNA Pol III promoter-driven miRNA hairpin construct for star-strand-specific overexpression in the liver. Liver steatosis and insulin resistance markers were evaluated in primary hepatocytes, mice with diabetes, and mice with excessive carbohydrate consumption. Results Functional loss of miR-192-3p in liver exacerbated hepatic micro-vesicular steatosis and insulin resistance in either mice with diabetes or wild-type mice with excessive fructose consumption. Liver-specific overexpression of miR-192-3p effectively halted hepatic steatosis and ameliorated insulin resistance in these mice models. Likewise, hepatocytes overexpressing miR-192-3p exhibited improved lipid accumulation, accompanied with decreases in lipogenesis and lipid-accumulation-related transcripts. Mechanistically, glucocorticoid receptor (GCR, also known as nuclear receptor subfamily 3, group C, member 1 [NR3C1]) was demonstrated to be negatively regulated by miR-192-3p. The effect of miR-192-3p on mitigating micro-vesicular steatosis was ablated by the reactivation of NR3C1. Conclusions The star strand miR-192-3p was an undermined glycerolipid regulator involved in controlling fat accumulation and insulin sensitivity in liver through blockade of hepatic GCR signalling; this miRNA may serve as a potential therapeutic option for the common co-mobility of diabetic mellitus and fatty liver disease. Lay summary The potential regulatory activity of star strand microRNA (miRNA) species has been substantially underestimated. In this study, we investigate the role and mechanism of an overlooked star strand miRNA (miR-192-3p) in regulating hepatic steatosis and insulin signalling in the livers of mice with diabetes and mice under excessive carbohydrate consumption. Liver-specific knockdown of miR-192-3p recapitulated functional loss of the miRNA as in mice with diabetes. This knockdown was characterised by pronounced hepatic micro-vesicular steatosis coupled to insulin resistance. In vivo overexpression of miR-192-3p alleviated hepatic steatosis in mice with diabetes and wild-type mice with excessive fructose consumption. Glucocorticoid receptor (also known as NR3C1) was discovered as the immediate target of miR-192-3p in regulating hepatic lipid turnover and storage.
Collapse
Key Words
- 3′-UTR, 3′-untranslated region
- AAV, adeno-associated virus
- CPT, carnitine palmitoyl transferase
- DEG, differentially expressed gene
- DEX, dexamethasone
- DM, diabetes mellitus
- DNL, de novo lipogenesis
- Diabetes mellitus
- FA, fatty acid
- FAO, fatty acid oxidation
- FASN, fatty acid synthase
- GCR, glucocorticoid receptor
- Glucocorticoid receptor
- HFD, high-fat diet
- HFrD, high-fructose drink
- HOMA-IR, homeostatic model assessment of insulin resistance
- Hepatic steatosis
- High carbohydrate consumption
- MicroRNA
- NAFLD, non-alcoholic fatty liver disease
- NR3C1, nuclear receptor subfamily 3, group C, member 1
- NT, non-targeting
- OA, oleic acid
- OGTT, oral glucose tolerance test
- SCD1, stearoyl-CoA desaturase-1
- T2DM, type 2 diabetes mellitus
- TAG, triacylglyceride/triglyceride
- Transcription repressor
- VAT, visceral adipose tissue
- miRNA, microRNA
- shRNA, short hairpin RNA
Collapse
|
396
|
Jiao T, Yao X, Zhao Y, Zhou Y, Gao Y, Fan S, Chen P, Li X, Jiang Y, Yang X, Gonzalez FJ, Huang M, Bi H. Dexamethasone-Induced Liver Enlargement Is Related to PXR/YAP Activation and Lipid Accumulation but Not Hepatocyte Proliferation. Drug Metab Dispos 2020; 48:830-839. [PMID: 32561593 PMCID: PMC7497622 DOI: 10.1124/dmd.120.000061] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Dexamethasone (Dex), a widely prescribed anti-inflammatory drug, was reported to induce liver enlargement (hepatomegaly) in clinical practice and in animal models. However, the underlying mechanisms are not elucidated. Dex is a known activator of pregnane X receptor (PXR). Yes-associated protein (YAP) has been implicated in chemically induced liver enlargement. Here, the roles of PXR and YAP pathways were investigated in Dex-induced hepatomegaly. Upregulation of PXR downstream proteins, including cytochrome P450 (CYP) 3A11, 2B10, and organic anion transporter polypeptide 2 (OATP2), indicated PXR signaling was activated after high dose of Dex (50 mg/kg, i.p.), and Dex at 100 μM activated PXR in the dual-luciferase reporter gene assay. Dex also increased the expression of total YAP, nuclear YAP, and YAP downstream proteins, including connective tissue growth factor and cysteine-rich angiogenic inducer 61, indicating activation of the YAP pathway. Furthermore, nuclear translocation of YAP was promoted by activation of PXR. However, hepatocyte proliferation was inhibited with significant decrease in the expression of proliferation-related proteins cyclin D1 and proliferating cell nuclear antigen as well as other regulatory factors, such as forkhead box protein M1, c-MYC, and epidermal growth factor receptor. The inhibitory effect of Dex on hepatocyte proliferation was likely due to its anti-inflammation effect of suppression of inflammation factors. β-catenin staining revealed enlarged hepatocytes, which were mostly attributable to the accumulation of lipids, such as triglycerides. In summary, high-dose Dex increased liver size accompanied by enlarged hepatocytes, and this was due to the activation of PXR/YAP and their effects on lipid accumulation but not hepatocyte proliferation. These findings provide new insights for understanding the mechanism of Dex-induced hepatomegaly. SIGNIFICANCE STATEMENT: This study identified the roles of pregnane X receptor (PXR) and yes-associated protein (YAP) pathways in dexamethasone (Dex)-induced hepatomegaly. Dex induced PXR/YAP activation, enlarged hepatocytes, and promoted liver enlargement with lipid accumulation, such as triglycerides. However, hepatocyte proliferation was inhibited by the anti-inflammatory effect of Dex. These findings provide new insights for understanding the mechanism of Dex-induced hepatomegaly.
Collapse
Affiliation(s)
- Tingying Jiao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Xinpeng Yao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yingyuan Zhao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yanying Zhou
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yue Gao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Shicheng Fan
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Panpan Chen
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Xuan Li
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Yiming Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Xiao Yang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Frank J Gonzalez
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (T.J., X.P.Y., Yi.Z., Ya.Z., Y.G., S.F., P.C., X.L., Y.J., X.Y., M.H., H.B.) and Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland (F.J.G.)
| |
Collapse
|
397
|
Hu J, Wang H, Li X, Liu Y, Mi Y, Kong H, Xi D, Yan W, Luo X, Ning Q, Wang X. Fibrinogen-like protein 2 aggravates nonalcoholic steatohepatitis via interaction with TLR4, eliciting inflammation in macrophages and inducing hepatic lipid metabolism disorder. Theranostics 2020; 10:9702-9720. [PMID: 32863955 PMCID: PMC7449923 DOI: 10.7150/thno.44297] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 07/24/2020] [Indexed: 12/17/2022] Open
Abstract
Rationale: The functions of fibrinogen-like protein 2 (fgl2) have been studied in many inflammatory and neoplastic diseases, but the role of fgl2 in nonalcoholic fatty liver disease has not yet been elucidated. In this study, we sought to investigate the role of fgl2 in the pathogenesis of nonalcoholic steatohepatitis (NASH). Methods: Hepatic fgl2 expression was tested in patients with nonalcoholic fatty liver (NAFL) or NASH and controls. Wild-type and fgl2-/- C57BL/6 mice were subjected to a methionine/choline-deficient (MCD) diet or a high-fat diet (HFD) to establish NASH models. Bone marrow-derived macrophages (BMDMs) stimulated with LPS or free fatty acids were used for the in vitro study. Results: In both humans and mice with NASH, macrophage accumulation was concomitant with significantly increased fgl2 expression in the liver. Fgl2 deficiency attenuated liver steatosis and inflammation in diet-induced murine models of NASH. In both liver tissues and BMDMs from NASH mice, fgl2 deficiency resulted in reduced levels of proinflammatory cytokines and reactive oxygen species (ROS) compared with levels in wild-type controls. Activation of NF-κB, p38-MAPK and NLRP3 inflammasomes was also suppressed upon fgl2 disruption. Moreover, lipogenic genes (Fasn and SREBP-2) were downregulated while lipolytic genes (PPAR and CPT1A) were upregulated in the livers of fgl2-/- NASH mice. Primary hepatocytes incubated with the medium collected from fgl2-/- BMDMs showed less fat deposition than those incubated with WT BMDMs. Furthermore, we discovered that fgl2 combined with TLR4 mediates the activation of the Myd88-dependent signaling pathway, which may contribute to inflammation and lipid metabolism disorders. Conclusions: These data suggest that fgl2 aggravates the progression of NASH through activation of NF-κB, p38-MAPK and NLRP3 inflammasomes in macrophages, which consequently induces overproduction of proinflammatory cytokines and lipid metabolism disorders. An interaction of fgl2 and TLR4 may in part contribute to the activation of inflammatory signaling pathways in macrophages.
Collapse
Affiliation(s)
- Junjian Hu
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongwu Wang
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xitang Li
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yonggang Liu
- Tianjin Second People's Hospital and Tianjin Institute of Hepatology, Tianjin, China
| | - Yuqiang Mi
- Tianjin Second People's Hospital and Tianjin Institute of Hepatology, Tianjin, China
| | - Hongyan Kong
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dong Xi
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weiming Yan
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoping Luo
- Department and institute of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Ning
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojing Wang
- Department and institute of infectious diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
398
|
Kalyesubula M, Mopuri R, Rosov A, Alon T, Edery N, Moallem U, Dvir H. Hyperglycemia-stimulating diet induces liver steatosis in sheep. Sci Rep 2020; 10:12189. [PMID: 32699301 PMCID: PMC7376193 DOI: 10.1038/s41598-020-68909-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/02/2020] [Indexed: 01/15/2023] Open
Abstract
Hepatic steatosis is strongly associated with chronic liver disease and systemic metabolic disorder. Adipose lipolysis is a recognized principal source of intrahepatic fat in various metabolic disorders, including non-alcoholic fatty liver disease. We hypothesized that, in the premorbid state, hepatic de novo lipogenesis (DNL) driven by excess carbohydrates abundance might play a more significant role. We employed a novel nutritional model in sheep of two distinct carbohydrates abundances. During 4 months of the dietary treatment, lambs were monitored for metabolic and terminal liver parameters. Lambs grown on the high-calorie (HC) diet were consistently more hyperglycemic and hyperinsulinemic than lambs grown on the lower-calorie (LC) diet (P < 0.0001). As a result, the HC lambs developed systemic- (HOMA-IR of 7.3 vs. 3.1; P < 0.0001), and adipose- (ADIPO-IR of 342.7 vs. 74.4; P < 0.0001) insulin resistance, significant adiposity (P < 0.0001), and higher plasma triglycerides (P < 0.05). Circulating leukocytes in the HC lambs had higher mRNA expression levels of the proinflammatory markers CCL2 (P < 0.01) and TNF-alpha (P < 0.04), and IL1B trended higher (P < 0.1). Remarkably, lambs on the HC diet developed substantial liver steatosis (mean fat content of 8.1 vs. 5.3% in the LC group; P < 0.0001) with a higher histological steatosis score (2.1 vs. 0.4; P < 0.0002). Hepatic steatosis was most-strongly associated with blood glucose and insulin levels but negatively correlated with circulating fatty acids-indicating a more significant contribution from hepatic DNL than from adipose lipolysis. Sheep may prove an attractive large-animal model of fatty liver and metabolic comorbidities resulting from excess carbohydrate-based energy early in life.
Collapse
Affiliation(s)
- Mugagga Kalyesubula
- Institute of Animal Science, Volcani Center - ARO, Rishon LeZion, Israel
- Department of Animal Science, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ramgopal Mopuri
- Institute of Animal Science, Volcani Center - ARO, Rishon LeZion, Israel
| | - Alexander Rosov
- Institute of Animal Science, Volcani Center - ARO, Rishon LeZion, Israel
| | - Tamir Alon
- Institute of Animal Science, Volcani Center - ARO, Rishon LeZion, Israel
- Department of Animal Science, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Nir Edery
- Pathology Laboratory, Kimron Veterinary Institute, Veterinary Services, Rishon LeZion, Israel
| | - Uzi Moallem
- Institute of Animal Science, Volcani Center - ARO, Rishon LeZion, Israel
| | - Hay Dvir
- Institute of Animal Science, Volcani Center - ARO, Rishon LeZion, Israel.
| |
Collapse
|
399
|
Muscella A, Stefàno E, Marsigliante S. The effects of exercise training on lipid metabolism and coronary heart disease. Am J Physiol Heart Circ Physiol 2020; 319:H76-H88. [PMID: 32442027 DOI: 10.1152/ajpheart.00708.2019] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Blood lipoproteins are formed by various amounts of cholesterol (C), triglycerides (TGs), phospholipids, and apolipoproteins (Apos). ApoA1 is the major structural protein of high-density lipoprotein (HDL), accounting for ~70% of HDL protein, and mediates many of the antiatherogenic functions of HDL. Conversely, ApoB is the predominant low-density lipoprotein (LDL) Apo and is an indicator of circulating LDL, associated with higher coronary heart disease (CHD) risk. Thus, the ratio of ApoB to ApoA1 (ApoB/ApoA1) is used as a surrogate marker of the risk of CHD related to lipoproteins. Elevated or abnormal levels of lipids and/or lipoproteins in the blood are a significant CHD risk factor, and several studies support the idea that aerobic exercise decreases CHD risk by partially lowering serum TG and LDL-cholesterol (LDL-C) levels and increasing HDL-C levels. Exercise also exerts an effect on HDL-C maturation and composition and on reverse C transport from peripheral cells to the liver to favor its catabolism and excretion. This process prevents atherosclerosis, and several studies showed that exercise training increases heart lipid metabolism and protects against cardiovascular disease. In these and other ways, it more and more appears that regular exercise, nutrition, and strategies to modulate lipid profile should be viewed as an integrated whole. The purpose of this review is to assess the effects of endurance training on the nontraditional lipid biomarkers, including ApoB, ApoA1, and ApoB/ApoA1, in CHD risk.
Collapse
Affiliation(s)
- Antonella Muscella
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali, Università del Salento, Lecce, Italy
| | - Erika Stefàno
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali, Università del Salento, Lecce, Italy
| | - Santo Marsigliante
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali, Università del Salento, Lecce, Italy
| |
Collapse
|
400
|
Narankiewicz D, Ruiz-Nava J, Buonaiuto V, Ruiz-Moreno MI, López-Carmona MD, Pérez-Belmonte LM, Gómez-Huelgas R, Bernal-López MR. Utility of Liver Function Tests and Fatty Liver Index to Categorize Metabolic Phenotypes in a Mediterranean Population. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17103518. [PMID: 32443453 PMCID: PMC7277926 DOI: 10.3390/ijerph17103518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/21/2022]
Abstract
The aim of this study was to analyze the utility of liver function tests (LFT) and fatty liver index (FLI), a surrogate marker of non-alcoholic fatty liver disease, in the categorization of metabolic phenotypes in a Mediterranean population. A cross-sectional study was performed on a random representative sample of 2233 adults assigned to a health center in Málaga, Spain. The metabolic phenotypes were determined based on body mass index (BMI) categorization and the presence or absence of two or more cardiometabolic abnormalities (high blood pressure, low high-density lipoprotein (HDL) cholesterol, hypertriglyceridemia, pre-diabetes) or type 2 diabetes. No difference was observed between metabolically healthy and metabolically abnormal phenotypes on LFT. The mean FLI of the population was 41.1 ± 28.6. FLI was significantly higher (p < 0.001) in the metabolically abnormal phenotypes in all BMI categories. The proportion of individuals with pathological FLI (≥60) was significantly higher in the metabolically abnormal overweight and obese phenotypes (p < 0.001). On a multivariate model adjusted for sex, age, and waist circumference, a significant correlation was found between pathological FLI and metabolically abnormal phenotypes in the overweight and obese BMI categories. Area under the curve (AUC) of FLI as a biomarker was 0.76, 0.74, and 0.72 for the metabolically abnormal normal-weight, overweight, and obese groups, respectively. Liver biochemistry is poorly correlated with metabolic phenotypes. Conversely, a good correlation between FLI, as a marker of non-alcoholic fatty liver disease (NAFLD), and metabolically abnormal phenotypes in all BMI ranges was found. Our study suggests that FLI may be a useful marker for characterizing metabolically abnormal phenotypes in individuals who are overweight or obese.
Collapse
Affiliation(s)
- Dariusz Narankiewicz
- Preventive Medicine Department, Virgen de la Victoria University Hospital, 29010 Malaga, Spain;
| | - Josefina Ruiz-Nava
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
| | - Veronica Buonaiuto
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
| | - María Isabel Ruiz-Moreno
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
| | - María Dolores López-Carmona
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
| | - Luis Miguel Pérez-Belmonte
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
| | - Ricardo Gómez-Huelgas
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
- Ciber Fisiopatología de la Obesidad y Nutrición. Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (R.G.-H.); (M.R.B.-L.); Tel.: +34-951-291-169 (R.G.-H.); 34-951-290-346 (M.R.B.-L.); Fax: +34-951-290-006 (R.G.-H.); +34-951-290-302 (M.R.B.-L.)
| | - María Rosa Bernal-López
- Internal Medicine Department, Regional University Hospital of Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Malaga, Spain; (J.R.-N.); (V.B.); (M.I.R.-M.); (M.D.L.-C.); (L.M.P.-B.)
- Ciber Fisiopatología de la Obesidad y Nutrición. Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (R.G.-H.); (M.R.B.-L.); Tel.: +34-951-291-169 (R.G.-H.); 34-951-290-346 (M.R.B.-L.); Fax: +34-951-290-006 (R.G.-H.); +34-951-290-302 (M.R.B.-L.)
| |
Collapse
|