1
|
Lin Y, Ye H, Chen Y, Zhang R, Chen Y, Ou W. Integrative Analyses of Genes of Pediatric Non-alcoholic Fatty Liver Disease Associated with Energy Metabolism. Dig Dis Sci 2024:10.1007/s10620-024-08702-4. [PMID: 39496907 DOI: 10.1007/s10620-024-08702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Pediatric non-alcoholic fatty liver disease (NAFLD) is a chronic steatosis of the liver associated with energy metabolism in children and adolescents, failure to intervene promptly can elevate the risk of developing hepatocellular carcinoma. Therefore, this study aimed to understand the underlying mechanism of pediatric NAFLD and investigate potential biomarkers and therapeutic targets. METHODS We investigated genes using the GSE185051 data set related to energy metabolism from the GeneCards database, constructed protein-protein interaction network, identified hub genes and established networks representing interactions between these hub genes and miRNA, RNA-binding proteins, transcription factors, and drugs. Subsequently, we performed Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis, Gene Set Enrichment Analysis (GSEA), and immune infiltration analysis. RESULTS Our analysis identified 9 hub genes through the PPI network. The target molecules were identified through the interaction network between hub genes and miRNAs, RNA-binding proteins, transcription factors, and drugs. GO analysis revealed that hub genes were associated with oxidative stress responses and other pathways. KEGG analysis highlighted their involvement in pathways such as insulin resistance, among others. GSEA revealed that hub genes were highly enriched in pathways related to Omega-9 fatty acid synthesis, among others. Immune infiltration analysis suggested that mast cells and T follicular helper cells play significant roles in the pathogenesis of NAFLD. CONCLUSION We identified the hub genes in pediatric NAFLD closely related to energy metabolism. These findings offer the potential for identifying potential novel diagnostic biomarkers, and establishing therapeutic targets for pediatric NAFLD.
Collapse
Affiliation(s)
- Yijun Lin
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Hong Ye
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China.
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China.
| | - Yan Chen
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Rui Zhang
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Yuyun Chen
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Weijie Ou
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| |
Collapse
|
2
|
Sahin C, Melanson JR, Le Billan F, Magomedova L, Ferreira TAM, Oliveira AS, Pollock-Tahari E, Saikali MF, Cash SB, Woo M, Romeiro LAS, Cummins CL. A novel fatty acid mimetic with pan-PPAR partial agonist activity inhibits diet-induced obesity and metabolic dysfunction-associated steatotic liver disease. Mol Metab 2024; 85:101958. [PMID: 38763495 PMCID: PMC11170206 DOI: 10.1016/j.molmet.2024.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVE The prevalence of metabolic diseases is increasing globally at an alarming rate; thus, it is essential that effective, accessible, low-cost therapeutics are developed. Peroxisome proliferator-activated receptors (PPARs) are transcription factors that tightly regulate glucose homeostasis and lipid metabolism and are important drug targets for the treatment of type 2 diabetes and dyslipidemia. We previously identified LDT409, a fatty acid-like compound derived from cashew nut shell liquid, as a novel pan-active PPARα/γ/δ compound. Herein, we aimed to assess the efficacy of LDT409 in vivo and investigate the molecular mechanisms governing the actions of the fatty acid mimetic LDT409 in diet-induced obese mice. METHODS C57Bl/6 mice (6-11-month-old) were fed a chow or high fat diet (HFD) for 4 weeks; mice thereafter received once daily intraperitoneal injections of vehicle, 10 mg/kg Rosiglitazone, 40 mg/kg WY14643, or 40 mg/kg LDT409 for 18 days while continuing the HFD. During treatments, body weight, food intake, glucose and insulin tolerance, energy expenditure, and intestinal lipid absorption were measured. On day 18 of treatment, tissues and plasma were collected for histological, molecular, and biochemical analysis. RESULTS We found that treatment with LDT409 was effective at reversing HFD-induced obesity and associated metabolic abnormalities in mice. LDT409 lowered food intake and hyperlipidemia, while improving insulin tolerance. Despite being a substrate of both PPARα and PPARγ, LDT409 was crucial for promoting hepatic fatty acid oxidation and reducing hepatic steatosis in HFD-fed mice. We also highlighted a role for LDT409 in white and brown adipocytes in vitro and in vivo where it decreased fat accumulation, increased lipolysis, induced browning of WAT, and upregulated thermogenic gene Ucp1. Remarkably, LDT409 reversed HFD-induced weight gain back to chow-fed control levels. We determined that the LDT409-induced weight-loss was associated with a combination of increased energy expenditure (detectable before weight loss was apparent), decreased food intake, increased systemic fat utilization, and increased fecal lipid excretion in HFD-fed mice. CONCLUSIONS Collectively, LDT409 represents a fatty acid mimetic that generates a uniquely favorable metabolic response for the treatment of multiple abnormalities including obesity, dyslipidemia, metabolic dysfunction-associated steatotic liver disease, and diabetes. LDT409 is derived from a highly abundant natural product-based starting material and its development could be pursued as a therapeutic solution to the global metabolic health crisis.
Collapse
Affiliation(s)
- Cigdem Sahin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Jenna-Rose Melanson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Florian Le Billan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Lilia Magomedova
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Thais A M Ferreira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Andressa S Oliveira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Evan Pollock-Tahari
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Michael F Saikali
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Sarah B Cash
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada; Banting and Best Diabetes Centre, Toronto, ON, M5G 2C4, Canada
| | - Luiz A S Romeiro
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; Banting and Best Diabetes Centre, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
3
|
Boesch M, Lindhorst A, Feio-Azevedo R, Brescia P, Silvestri A, Lannoo M, Deleus E, Jaekers J, Topal H, Topal B, Ostyn T, Wallays M, Smets L, Van Melkebeke L, Härtlova A, Roskams T, Bedossa P, Verbeek J, Govaere O, Francque S, Sifrim A, Voet T, Rescigno M, Gericke M, Korf H, van der Merwe S. Adipose tissue macrophage dysfunction is associated with a breach of vascular integrity in NASH. J Hepatol 2024; 80:397-408. [PMID: 37977244 DOI: 10.1016/j.jhep.2023.10.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/03/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND & AIMS In non-alcoholic fatty liver disease (NAFLD), monocytes infiltrate visceral adipose tissue promoting local and hepatic inflammation. However, it remains unclear what drives inflammation and how the immune landscape in adipose tissue differs across the NAFLD severity spectrum. We aimed to assess adipose tissue macrophage (ATM) heterogeneity in a NAFLD cohort. METHODS Visceral adipose tissue macrophages from lean and obese patients, stratified by NAFLD phenotypes, underwent single-cell RNA sequencing. Adipose tissue vascular integrity and breaching was assessed on a protein level via immunohistochemistry and immunofluorescence to determine targets of interest. RESULTS We discovered multiple ATM populations, including resident vasculature-associated macrophages (ResVAMs) and distinct metabolically active macrophages (MMacs). Using trajectory analysis, we show that ResVAMs and MMacs are replenished by a common transitional macrophage (TransMac) subtype and that, during NASH, MMacs are not effectively replenished by TransMac precursors. We postulate an accessory role for MMacs and ResVAMs in protecting the adipose tissue vascular barrier, since they both interact with endothelial cells and localize around the vasculature. However, across the NAFLD severity spectrum, alterations occur in these subsets that parallel an adipose tissue vasculature breach characterized by albumin extravasation into the perivascular tissue. CONCLUSIONS NAFLD-related macrophage dysfunction coincides with a loss of adipose tissue vascular integrity, providing a plausible mechanism by which tissue inflammation is perpetuated in adipose tissue and downstream in the liver. IMPACT AND IMPLICATIONS Our study describes for the first time the myeloid cell landscape in human visceral adipose tissue at single-cell level within a cohort of well-characterized patients with non-alcoholic fatty liver disease. We report unique non-alcoholic steatohepatitis-specific transcriptional changes within metabolically active macrophages (MMacs) and resident vasculature-associated macrophages (ResVAMs) and we demonstrate their spatial location surrounding the vasculature. These dysfunctional transcriptional macrophage states coincided with the loss of adipose tissue vascular integrity, providing a plausible mechanism by which tissue inflammation is perpetuated in adipose tissue and downstream in the liver. Our study provides a theoretical basis for new therapeutic strategies to be directed towards reinstating the endogenous metabolic, homeostatic and cytoprotective functions of ResVAMs and MMacs, including their role in protecting vascular integrity.
Collapse
Affiliation(s)
- Markus Boesch
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | | | - Rita Feio-Azevedo
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Paola Brescia
- IRCCS Humanitas Research Hospital, Manzoni 56, 20089 Rozzano, Milan, Italy
| | | | | | - Ellen Deleus
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Joris Jaekers
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Halit Topal
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Baki Topal
- Department of Abdominal Surgery, UZ Leuven, Leuven, Belgium
| | - Tessa Ostyn
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, 3000 Leuven, Belgium
| | - Marie Wallays
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Lena Smets
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | - Lukas Van Melkebeke
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Anetta Härtlova
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Tania Roskams
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, 3000 Leuven, Belgium
| | - Pierre Bedossa
- Department of Pathology, Physiology and Imaging, Beaujon Hospital Paris Diderot University, Paris, France
| | - Jef Verbeek
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium
| | - Olivier Govaere
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, 3000 Leuven, Belgium
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium; Translational Research in Inflammation and Immunology (TWI2N), Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Alejandro Sifrim
- KU Leuven Institute for Single Cell Omics (LISCO), 3000 Leuven, Belgium; Laboratory of Multi-omic Integrative Bioinformatics, Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Thierry Voet
- KU Leuven Institute for Single Cell Omics (LISCO), 3000 Leuven, Belgium; Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20072 Pieve Emanuele, Milan, Italy
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Hannelie Korf
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium.
| | - Schalk van der Merwe
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, UZ Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Colella F, Ramachandran P. Adipose tissue macrophage dysfunction in human MASLD - Cause or consequence? J Hepatol 2024; 80:390-393. [PMID: 38122832 DOI: 10.1016/j.jhep.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Affiliation(s)
- Fabio Colella
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Prakash Ramachandran
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
5
|
Lu B, Wu C, Zhang J, Zhang J, Zhang J. Oral Ionic Liquid for Transdermal Delivery and Obesity Treatment. ACS Biomater Sci Eng 2023. [PMID: 37115006 DOI: 10.1021/acsbiomaterials.3c00118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Obesity is currently a prerequisite for more than 70% of adults, including chronic obesity and long-term obesity. With the increase of diabetes patients in the world, it is urgent to develop effective oral drugs to replace insulin. However, the gastrointestinal tract is a main obstacle to oral drug preparations. Here, a highly effective oral drug was developed, mainly formulated as an ionic liquid (IL) prepared by l-(-)-carnitine and geranic acid. Density functional theory (DFT) calculations showed that l-(-)-carnitine and geranic acid can exist stably through hydrogen bonding. IL can significantly enhance the transdermal transport of drugs. In vitro study of intestinal permeability showed that particles formed by IL can prevent the absorption of intestinal fat. Compared with the control group, oral administration of IL (10 mL kg-1) significantly reduced blood glucose, white adipose tissue in the liver and epididymis, and the expression of SREBP-1c and ACC in IL. Therefore, these results and high-throughput sequencing analysis showed that IL can effectively reduce the intestinal absorption of adipose tissue to reduce blood glucose. IL has good biocompatibility and stability. Therefore, IL has a certain application value in the field of oral drug-delivery carriers, which provides an effective means for the treatment of diabetes and is a potential tool to solve the epidemic of obesity.
Collapse
Affiliation(s)
- Beibei Lu
- Department of Dermatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen 518020, Guangdong, China
- Department of Shenzhen People's Hospital Geriatrics Center, Shenzhen 518020, Guangdong, China
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
| | - Chengyu Wu
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
| | - Jichuan Zhang
- Department of Chemistry, University of Idaho, Moscow, Idaho 83844-2343, United States
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen 518020, Guangdong, China
- Department of Shenzhen People's Hospital Geriatrics Center, Shenzhen 518020, Guangdong, China
| | - Jiaheng Zhang
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
| |
Collapse
|
6
|
Terra M, García-Arévalo M, Avelino T, Degaki K, Malospirito C, de Carvalho M, Torres F, Saito Â, Figueira A. AM-879, a PPARy non-agonist and Ser273 phosphorylation blocker, promotes insulin sensitivity without adverse effects in mice. Metabol Open 2023; 17:100221. [PMID: 36588655 PMCID: PMC9800205 DOI: 10.1016/j.metop.2022.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Obesity is one of the main risk factors for type 2 diabetes, and peroxisome proliferator-activated receptor γ (PPARγ) is considered a promising pathway on insulin sensitivity and adipose tissue metabolism. The search for molecules acting as insulin sensitizers have increased, especially for molecules that block PPARγ-Ser273 phosphorylation, without reaching full agonism. We evaluated the in vivo effects of AM-879, a PPARγ non-agonist, and found that AM-879 exerts different effects in mice depending on the dose. At lower doses, this ligand decreased BAT, increased leptin and Crh expression. However, at a higher dose, it promoted improvement on insulin sensitivity, ameliorates expression of metabolism-related genes, decreased the expression of genes related to liver toxicity, maintaining body weight and adipocyte size. These results present a new lead molecule to ameliorates insulin resistance and confirm AM-879 as a PPARγ non-agonist which blocks Ser273 phosphorylation as a good strategy to modulate insulin sensitivity without developing the adverse effects promoted by PPARγ full agonists.
Collapse
Affiliation(s)
- M.F. Terra
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
- Post Graduate Program in Functional and Molecular Biology, Institute of Biology, State University of Campinas (Unicamp), Campinas, Brazil
| | - M. García-Arévalo
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - T.M. Avelino
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
- Post Graduate Program in Pharmacological Science, State University of Campinas (Unicamp), Campinas, Brazil
| | - K.Y. Degaki
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - C.C. Malospirito
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
- Post Graduate Program in Functional and Molecular Biology, Institute of Biology, State University of Campinas (Unicamp), Campinas, Brazil
| | - M. de Carvalho
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - F.R. Torres
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - Â. Saito
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| | - A.C.M. Figueira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
| |
Collapse
|
7
|
Gart E, van Duyvenvoorde W, Snabel JM, de Ruiter C, Attema J, Caspers MPM, Lek S, van Heuven BJ, Speksnijder AGCL, Giera M, Menke A, Salic K, Bence KK, Tesz GJ, Keijer J, Kleemann R, Morrison MC. Translational characterization of the temporal dynamics of metabolic dysfunctions in liver, adipose tissue and the gut during diet-induced NASH development in Ldlr-/-.Leiden mice. Heliyon 2023; 9:e13985. [PMID: 36915476 PMCID: PMC10006542 DOI: 10.1016/j.heliyon.2023.e13985] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Background NAFLD progression, from steatosis to inflammation and fibrosis, results from an interplay of intra- and extrahepatic mechanisms. Disease drivers likely include signals from white adipose tissue (WAT) and gut. However, the temporal dynamics of disease development remain poorly understood. Methods High-fat-diet (HFD)-fed Ldlr-/-.Leiden mice were compared to chow-fed controls. At t = 0, 8, 16, 28 and 38w mice were euthanized, and liver, WAT depots and gut were analyzed biochemically, histologically and by lipidomics and transcriptomics together with circulating factors to investigate the sequence of pathogenic events and organ cross-talk during NAFLD development. Results HFD-induced obesity was associated with an increase in visceral fat, plasma lipids and hyperinsulinemia at t = 8w, along with increased liver steatosis and circulating liver damage biomarkers. In parallel, upstream regulator analysis predicted that lipid catabolism regulators were deactivated and lipid synthesis regulators were activated. Subsequently, hepatocyte hypertrophy, oxidative stress and hepatic inflammation developed. Hepatic collagen accumulated from t = 16 w and became pronounced at t = 28-38 w. Epididymal WAT was maximally hypertrophic from t = 8 w, which coincided with inflammation development. Mesenteric and subcutaneous WAT hypertrophy developed slower and did not appear to reach a maximum, with minimal inflammation. In gut, HFD significantly increased permeability, induced a shift in microbiota composition from t = 8 w and changed circulating gut-derived metabolites. Conclusion HFD-fed Ldlr-/-.Leiden mice develop obesity, dyslipidemia and insulin resistance, essentially as observed in obese NAFLD patients, underlining their translational value. We demonstrate that marked epididymal-WAT inflammation, and gut permeability and dysbiosis precede the development of NAFLD stressing the importance of a multiple-organ approach in the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, the Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Jessica M Snabel
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Christa de Ruiter
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Joline Attema
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Martien P M Caspers
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - Serene Lek
- Clinnovate Health UK Ltd, Glasgow, United Kingdom
| | | | | | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Aswin Menke
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Kanita Salic
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Kendra K Bence
- Pfizer Worldwide Research, Development & Medical, Internal Medicine Research Unit, Cambridge, MA, USA
| | - Gregory J Tesz
- Pfizer Worldwide Research, Development & Medical, Internal Medicine Research Unit, Cambridge, MA, USA
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, the Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 CK Leiden, the Netherlands
| |
Collapse
|
8
|
Vreeken D, Seidel F, de La Roij G, Vening W, den Hengst WA, Verschuren L, Özsezen S, Kessels RPC, Duering M, Mutsaerts HJMM, Kleemann R, Wiesmann M, Hazebroek EJ, Kiliaan AJ. Impact of White Adipose Tissue on Brain Structure, Perfusion, and Cognitive Function in Patients With Severe Obesity: The BARICO Study. Neurology 2023; 100:e703-e718. [PMID: 36332987 PMCID: PMC9969926 DOI: 10.1212/wnl.0000000000201538] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND AND OBJECTIVE While underlying pathophysiology linking obesity to brain health is not completely understood, white adipose tissue (WAT) is considered a key player. In obesity, WAT becomes dysregulated, showing hyperplasia, hypertrophy, and eventually inflammation. This disbalance leads to dysregulated secretion of adipokines influencing both (cardio)vascular and brain health. Within this study, we investigated the association between omental WAT (oWAT) and subcutaneous WAT (scWAT) with brain structure and perfusion and cognition in adults with severe obesity. METHODS Within the cross-sectional BARICO study, brain structure and perfusion and cognitive function were measured before bariatric surgery (BS) using MRI and cognitive assessments. During BS, oWAT and scWAT depots were collected and analyzed by histopathology. The number and diameter of adipocytes were quantified together with the amount of crown-like structures (CLS) as an indication of inflammation. Blood samples were collected to analyze adipokines and inflammatory markers. Neuroimaging outcomes included brain volumes, cortical thickness, white matter (WM) integrity, WM hyperintensities, cerebral blood flow using arterial spin labeling (ASL), and the ASL spatial coefficient of variation (sCoV), reflecting cerebrovascular health. RESULTS Seventy-one patients were included (mean age 45.1 ± 5.8 years; 83.1% women; mean body mass index 40.8 ± 3.8 kg/m2). scWAT showed more CLS (z = -2.72, p < 0.01, r = -0.24) and hypertrophy compared with oWAT (F(1,64) = 3.99, p < 0.05, η2 = 0.06). Adiponectin levels were inversely associated with the average diameter of scWAT (β = -0.31, 95% CI -0.54 to -0.08) and oWAT (β = -0.33, 95% CI -0.55 to -0.09). Furthermore, the adipocyte diameter in oWAT was positively associated with the sCoV in the parietal cortex (β = 0.33, 95% CI 0.10-0.60), and the number of adipocytes (per mm2) was positively associated with sCoV in the nucleus accumbens (NAcc) (β = 0.34, 95% CI 0.09-0.61). Cognitive function did not correlate with any WAT parameter or plasma marker. These associations were highly influenced by age and sex. sCoV in the NAcc was positively associated with fasting plasma glucose (β = 0.35, 95% CI 0.10-0.56). DISCUSSION scWAT and oWAT are different in morphology and in their relationship with plasma markers and cerebrovascular health. Although scWAT showed more CLS and hypertrophy, scWAT was not associated with brain readouts. This study showed, however, important relationships between oWAT morphology and cerebrovascular health in obesity. TRIAL REGISTRATION INFORMATION Trial Registration Number NTR7288 (trialregister.nl/trial/7090).
Collapse
Affiliation(s)
- Debby Vreeken
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Florine Seidel
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Guido de La Roij
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Wouter Vening
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Willem A den Hengst
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Lars Verschuren
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Serdar Özsezen
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Roy P C Kessels
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Marco Duering
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Henk J M M Mutsaerts
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Robert Kleemann
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Maximilian Wiesmann
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Eric J Hazebroek
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands
| | - Amanda J Kiliaan
- From the Department of Medical Imaging (D.V., F.S., G.L.R., M.W., A.J.K.), Anatomy, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Bariatric Surgery (D.V., W.V., W.A.H., E.J.H.), Vitalys, Part of Rijnstate Hospital, Arnhem, The Netherlands; Donders Institute for Brain (D.V., F.S., R.P.C.K., M.W., A.J.K.), Cognition, and Behavior and Radboudumc Alzheimer Center, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Metabolic Health Research (F.S., R.K.), Netherlands Organisation for Applied Scientific Research (TNO), Leiden; Department of Microbiology and Systems Biology (L.V., S.Ö.), Netherlands Organisation for Applied Scientific Research (TNO), Zeist; Vincent van Gogh Institute for Psychiatry (R.P.C.K.), Venray, The Netherlands; Department of Medical Psychology and Radboudumc Alzheimer Center (R.P.C.K.), Radboud University Medical Center, Nijmegen, The Netherlands; Medical Image Analysis Center (MIAC) and Qbig (M.D.), and Department of Biomedical Engineering, University of Basel, Switzerland; Department of Radiology and Nuclear Medicine (H.J.M.M.M.), Amsterdam UMC, Amsterdam Neuroscience, The Netherlands; and Division of Human Nutrition and Health (E.J.H.), Wageningen University, The Netherlands.
| |
Collapse
|
9
|
Tang G, Ma C, Li L, Zhang S, Li F, Wu J, Yin Y, Zhu Q, Liang Y, Wang R, Huang H, Zhao TJ, Yang H, Li P, Chen FJ. PITPNC1 promotes the thermogenesis of brown adipose tissue under acute cold exposure. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2287-2300. [PMID: 36166181 DOI: 10.1007/s11427-022-2157-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/29/2022] [Indexed: 06/16/2023]
Abstract
Brown adipose tissue (BAT) plays an essential role in non-shivering thermogenesis. The phosphatidylinositol transfer protein, cytoplasmic 1 (PITPNC1) is identified as a lipid transporter that reciprocally transfers phospholipids between intracellular membrane structures. However, the physiological significance of PITPNC1 and its regulatory mechanism remain unclear. Here, we demonstrate that PITPNC1 is a key player in thermogenesis of BAT. While Pitpnc1-/- mice do not differ with wildtype mice in body weight and insulin sensitivity on either chow or high-fat diet, they develop hypothermia when subjected to acute cold exposure at 4°C. The Pitpnc1-/- brown adipocytes exhibit defective β-oxidation and abnormal thermogenesis-related metabolism pathways in mitochondria. The deficiency of lipid mobilization in Pitpnc1-/- brown adipocytes might be the result of excessive accumulation of phosphatidylcholine and a reduction of phosphatidic acid. Our findings have uncovered significant roles of PITPNC1 in mitochondrial phospholipid homeostasis and BAT thermogenesis.
Collapse
Affiliation(s)
- Guoqing Tang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Chengxin Ma
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Liangkui Li
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shaoyan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Fengsheng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Jin Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yesheng Yin
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Qing Zhu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yan Liang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - He Huang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
- Shanghai Qi Zhi Institute, Shanghai, 200030, China
| | - Hongyuan Yang
- School of Biotechnology and Biomolecular Sciences, the University of New South Wales, Sydney, NSW, 2052, Australia
| | - Peng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Shanghai Qi Zhi Institute, Shanghai, 200030, China
| | - Feng-Jung Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Institutes of Biomedical Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
- Shanghai Qi Zhi Institute, Shanghai, 200030, China.
| |
Collapse
|
10
|
Liang H, Qileng A, Shen H, Zhou Y, Liu W, Lei H, Liu Y. Handheld Platform for Sensitive Rosiglitazone Detection: Immunosensor Based on a Time-Based Readout Device. Anal Chem 2022; 94:4294-4302. [PMID: 35107977 DOI: 10.1021/acs.analchem.1c04957] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The detection of rosiglitazone (RSG) in food is of great importance since the excessive intake of RSG could cause adverse effects on the human body. Although liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry are the preliminary methods for the detection of hazardous materials in food, they are not suitable for point-of-care or on-site detection. Herein, a time-based readout (TBR) device with an application software (APP) controlled by a smart phone was developed for the sensitive and selective immunoassay of RSG. The homemade TBR device was based on a two-electrode system, where the immune molecule-modified glassy carbon electrode was used as the bioanode, and Prussian blue-modified FTO was used as the cathode. By using Au-modified octahedral Cu2O with high catalytic activity as mimetic peroxidase, an insulating layer was generated on the cathode by catalyzing 4-chloro-1-naphthol (4-CN) into benzo-4-chlorohexadienone (B4Q). The time to reach a fixed potential varied indirectly with the concentrations of RSG and was recognized by the APP, while the electrochromic property on the cathode was also correspondingly changed. Under optimum conditions, both the square root of the time and the chroma value of the electrochromism exhibited linear responses for the detection of RSG ranging from 5 × 10-10 to 5 × 10-7 g/L, while the limits of detection were 8.2 × 10-11 and 1.3 × 10-10 g/L, respectively. With easy operation and portability, this TBR device showed a promising application for point-of-care monitoring of hazardous materials in food or the environment.
Collapse
Affiliation(s)
- Hongzhi Liang
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Aori Qileng
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.,The Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Haoran Shen
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Yaowei Zhou
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Weipeng Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Hongtao Lei
- The Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yingju Liu
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.,The Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
11
|
Sharma S, Shen T, Chitranshi N, Gupta V, Basavarajappa D, Mirzaei M, You Y, Krezel W, Graham SL, Gupta V. Retinoid X Receptor: Cellular and Biochemical Roles of Nuclear Receptor with a Focus on Neuropathological Involvement. Mol Neurobiol 2022; 59:2027-2050. [PMID: 35015251 PMCID: PMC9015987 DOI: 10.1007/s12035-021-02709-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Retinoid X receptors (RXRs) present a subgroup of the nuclear receptor superfamily with particularly high evolutionary conservation of ligand binding domain. The receptor exists in α, β, and γ isotypes that form homo-/heterodimeric complexes with other permissive and non-permissive receptors. While research has identified the biochemical roles of several nuclear receptor family members, the roles of RXRs in various neurological disorders remain relatively under-investigated. RXR acts as ligand-regulated transcription factor, modulating the expression of genes that plays a critical role in mediating several developmental, metabolic, and biochemical processes. Cumulative evidence indicates that abnormal RXR signalling affects neuronal stress and neuroinflammatory networks in several neuropathological conditions. Protective effects of targeting RXRs through pharmacological ligands have been established in various cell and animal models of neuronal injury including Alzheimer disease, Parkinson disease, glaucoma, multiple sclerosis, and stroke. This review summarises the existing knowledge about the roles of RXR, its interacting partners, and ligands in CNS disorders. Future research will determine the importance of structural and functional heterogeneity amongst various RXR isotypes as well as elucidate functional links between RXR homo- or heterodimers and specific physiological conditions to increase drug targeting efficiency in pathological conditions.
Collapse
Affiliation(s)
- Samridhi Sharma
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| | - Ting Shen
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Melbourne, VIC, Australia
| | - Devaraj Basavarajappa
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Yuyi You
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.,Save Sight Institute, University of Sydney, Sydney, NSW, Australia
| | - Wojciech Krezel
- Institut de Génétique Et de Biologie Moléculaire Et Cellulaire, INSERM U1258, CNRS UMR 7104, Unistra, 67404, Illkirch-Graffenstaden, France
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.,Save Sight Institute, University of Sydney, Sydney, NSW, Australia
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
12
|
Krill Oil Treatment Increases Distinct PUFAs and Oxylipins in Adipose Tissue and Liver and Attenuates Obesity-Associated Inflammation via Direct and Indirect Mechanisms. Nutrients 2021; 13:nu13082836. [PMID: 34444996 PMCID: PMC8401900 DOI: 10.3390/nu13082836] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/31/2022] Open
Abstract
The development of obesity is characterized by the metabolic overload of tissues and subsequent organ inflammation. The health effects of krill oil (KrO) on obesity-associated inflammation remain largely elusive, because long-term treatments with KrO have not been performed to date. Therefore, we examined the putative health effects of 28 weeks of 3% (w/w) KrO supplementation to an obesogenic diet (HFD) with fat derived mostly from lard. The HFD with KrO was compared to an HFD control group to evaluate the effects on fatty acid composition and associated inflammation in epididymal white adipose tissue (eWAT) and the liver during obesity development. KrO treatment increased the concentrations of EPA and DHA and associated oxylipins, including 18-HEPE, RvE2 and 14-HDHA in eWAT and the liver. Simultaneously, KrO decreased arachidonic acid concentrations and arachidonic-acid-derived oxylipins (e.g., HETEs, PGD2, PGE2, PGF2α, TXB2). In eWAT, KrO activated regulators of adipogenesis (e.g., PPARγ, CEBPα, KLF15, STAT5A), induced a shift towards smaller adipocytes and increased the total adipocyte numbers indicative for hyperplasia. KrO reduced crown-like structures in eWAT, and suppressed HFD-stimulated inflammatory pathways including TNFα and CCL2/MCP-1 signaling. The observed eWAT changes were accompanied by reduced plasma leptin and increased plasma adiponectin levels over time, and improved insulin resistance (HOMA-IR). In the liver, KrO suppressed inflammatory signaling pathways, including those controlled by IL-1β and M-CSF, without affecting liver histology. Furthermore, KrO deactivated hepatic REL-A/p65-NF-κB signaling, consistent with increased PPARα protein expression and a trend towards an increase in IkBα. In conclusion, long-term KrO treatment increased several anti-inflammatory PUFAs and oxylipins in WAT and the liver. These changes were accompanied by beneficial effects on general metabolism and inflammatory tone at the tissue level. The stimulation of adipogenesis by KrO allows for safe fat storage and may, together with more direct PPAR-mediated anti-inflammatory mechanisms, attenuate inflammation.
Collapse
|
13
|
Doblado L, Lueck C, Rey C, Samhan-Arias AK, Prieto I, Stacchiotti A, Monsalve M. Mitophagy in Human Diseases. Int J Mol Sci 2021; 22:ijms22083903. [PMID: 33167334 PMCID: PMC8069949 DOI: 10.3390/ijms22083903] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Mitophagy is a selective autophagic process, essential for cellular homeostasis, that eliminates dysfunctional mitochondria. Activated by inner membrane depolarization, it plays an important role during development and is fundamental in highly differentiated post-mitotic cells that are highly dependent on aerobic metabolism, such as neurons, muscle cells, and hepatocytes. Both defective and excessive mitophagy have been proposed to contribute to age-related neurodegenerative diseases, such as Parkinson’s and Alzheimer’s diseases, metabolic diseases, vascular complications of diabetes, myocardial injury, muscle dystrophy, and liver disease, among others. Pharmacological or dietary interventions that restore mitophagy homeostasis and facilitate the elimination of irreversibly damaged mitochondria, thus, could serve as potential therapies in several chronic diseases. However, despite extraordinary advances in this field, mainly derived from in vitro and preclinical animal models, human applications based on the regulation of mitochondrial quality in patients have not yet been approved. In this review, we summarize the key selective mitochondrial autophagy pathways and their role in prevalent chronic human diseases and highlight the potential use of specific interventions.
Collapse
Affiliation(s)
- Laura Doblado
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| | - Claudia Lueck
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| | - Claudia Rey
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
| | - Alejandro K. Samhan-Arias
- Department of Biochemistry, Universidad Autónoma de Madrid e Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain;
| | - Ignacio Prieto
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Isaac Peral 42, 28015 Madrid, Spain;
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, Universita’ Degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
- Correspondence: (A.S.); (M.M.)
| | - Maria Monsalve
- Instituto de Investigaciones Biomédicas “Alberto Sols” (CSIC-UAM), Arturo Duperier 4, 28029 Madrid, Spain; (L.D.); (C.L.); (C.R.)
- Correspondence: (A.S.); (M.M.)
| |
Collapse
|
14
|
López-Reyes A, Martinez-Armenta C, Espinosa-Velázquez R, Vázquez-Cárdenas P, Cruz-Ramos M, Palacios-Gonzalez B, Gomez-Quiroz LE, Martínez-Nava GA. NLRP3 Inflammasome: The Stormy Link Between Obesity and COVID-19. Front Immunol 2020; 11:570251. [PMID: 33193349 PMCID: PMC7662564 DOI: 10.3389/fimmu.2020.570251] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022] Open
Abstract
Several countries around the world have faced an important obesity challenge for the past four decades as the result of an obesogenic environment. This disease has a multifactorial origin and it is associated with multiple comorbidities including type 2 diabetes, hypertension, osteoarthritis, metabolic syndrome, cancer, and dyslipidemia. With regard to dyslipidemia, hypertriglyceridemia is a well-known activator of the NLRP3 inflammasome, triggering adipokines and cytokines secretion which in addition induce a systemic inflammatory state that provides an adequate scenario for infections, particularly those mediated by viruses such as HIV, H1N1 influenza, and SARS-CoV-2. The SARS-CoV-2 infection causes the coronavirus disease 2019 (COVID-19) and it is responsible for the pandemic that we are currently living. COVID-19 causes an aggressive immune response known as cytokine release syndrome or cytokine storm that causes multiorgan failure and in most cases leads to death. In the present work, we aimed to review the molecular mechanisms by which obesity-associated systemic inflammation could cause a more severe clinical presentation of COVID-19. The SARS-CoV-2 infection could potentiate or accelerate the pre-existing systemic inflammatory state of individuals with obesity, via the NLRP3 inflammasome activation and the release of pro-inflammatory cytokines from cells trough Gasdermin-pores commonly found in cell death by pyroptosis.
Collapse
Affiliation(s)
- Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, México
- Facultad de Ciencias de la Salud, Universidad Anáhuac, Ciudad de México, México
| | - Carlos Martinez-Armenta
- Postgrado en Biología Experimental, Dirección de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, México
| | | | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, México
| | - Marlid Cruz-Ramos
- Cátedras de Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Cancerología, Ciudad de México, México
| | - Berenice Palacios-Gonzalez
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | - Luis Enrique Gomez-Quiroz
- Laboratorio de Fisiología Celular, Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, México
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Líquido Sinovial, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, México
| |
Collapse
|
15
|
Metabolically Healthy Obesity-Heterogeneity in Definitions and Unconventional Factors. Metabolites 2020; 10:metabo10020048. [PMID: 32012784 PMCID: PMC7074352 DOI: 10.3390/metabo10020048] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023] Open
Abstract
The concept of heterogeneity among obese individuals in their risk for developing metabolic dysfunction and associated complications has been recognized for decades. At the origin of the heterogeneity idea is the acknowledgement that individuals with central obesity are more prone to developing type 2 diabetes and cardiovascular disease than those with peripheral obesity. There have been attempts to categorize subjects according to their metabolic health and degree of obesity giving rise to different obese and non-obese phenotypes that include metabolically unhealthy normal-weight (MUHNW), metabolically healthy obese (MHO), and metabolically unhealthy obese (MUO). Individuals belonging to the MHO phenotype are obese according to their body mass index although exhibiting fewer or none metabolic anomalies such as type 2 diabetes, dyslipidemia, hypertension, and/or unfavorable inflammatory and fribinolytic profiles. However, some authors claim that MHO is only transient in nature. Additionally, the phenotype categorization is controversial as it lacks standardized definitions possibly blurring the distinction between obesity phenotypes and confounding the associations with health outcomes. To add to the discussion, the factors underlying the origin or protection from metabolic deterioration and cardiometabolic risk for these subclasses are being intensely investigated and several hypotheses have been put forward. In the present review, we compare the different definitions of obesity phenotypes and present several possible factors underlying them (adipose tissue distribution and cellularity, contaminant accumulation on the adipose tissue, dysbiosis and metabolic endotoxemia imposing on to the endocannabinoid tone and inflammasome, and nutrient intake and dietary patterns) having inflammatory activation at the center.
Collapse
|
16
|
Ritchie M, Hanouneh IA, Noureddin M, Rolph T, Alkhouri N. Fibroblast growth factor (FGF)-21 based therapies: A magic bullet for nonalcoholic fatty liver disease (NAFLD)? Expert Opin Investig Drugs 2020; 29:197-204. [PMID: 31948295 DOI: 10.1080/13543784.2020.1718104] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Introduction: Fibroblast growth factor (FGF) 21 is a member of the FGF19 sub-family of signaling molecules. They have been found to act at the localized paracrine/autocrine and systemic endocrine levels because of their extracellular matrix and co-receptor protein binding characteristics. While the molecule circulates systemically, it has specificity conferred by a co-factor binding protein β-Klotho which is preferentially expressed in hepatic and adipose tissues. This protein, in conjunction with the FGF receptor (FGFR), propagates the downstream effects of the growth factor signaling cascade, which has been linked to fat and glucose metabolism. FGF21 has been recognized as a possible pathway for the treatment of nonalcoholic fatty liver disease (NAFLD). Targeting of the FGF21/FGFR/β-Klotho pathway may halt or reverse hepatic fat infiltration, inflammation, and fibrosis.Areas covered: This article summarizes preclinical and clinical data on the efficacy and safety of two FGF21 agonist therapies in development.Expert opinion: Preclinical and clinical data justify further investigation of FGF21 agonist therapies for the treatment of NAFLD. However, issues including injection site reactions and possible effects on bone homeostasis mean that safety must be evaluated carefully.
Collapse
Affiliation(s)
- Michael Ritchie
- Department of Internal Medicine, Abbott Northwestern Hospital and Minnesota Gastroenterology, Minneapolis, MN, USA
| | - Ibrahim A Hanouneh
- Department of Internal Medicine, Abbott Northwestern Hospital and Minnesota Gastroenterology, Minneapolis, MN, USA.,Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Mazen Noureddin
- Department of Gastroenterology and Hepatology, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Timothy Rolph
- Department of Research and Development, Akero Therapeutics, San Francisco, CA, USA
| | - Naim Alkhouri
- Department of Hepatology, Texas Liver Institute, University of Texas Health San Antonio (UTHSA), San Antonio, TX, USA
| |
Collapse
|
17
|
Sibuyi NRS, Moabelo KL, Meyer M, Onani MO, Dube A, Madiehe AM. Nanotechnology advances towards development of targeted-treatment for obesity. J Nanobiotechnology 2019; 17:122. [PMID: 31842876 PMCID: PMC6913004 DOI: 10.1186/s12951-019-0554-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Obesity through its association with type 2 diabetes (T2D), cancer and cardiovascular diseases (CVDs), poses a serious health threat, as these diseases contribute to high mortality rates. Pharmacotherapy alone or in combination with either lifestyle modification or surgery, is reliable in maintaining a healthy body weight, and preventing progression to obesity-induced diseases. However, the anti-obesity drugs are limited by non-specificity and unsustainable weight loss effects. As such, novel and improved approaches for treatment of obesity are urgently needed. Nanotechnology-based therapies are investigated as an alternative strategy that can treat obesity and be able to overcome the drawbacks associated with conventional therapies. The review presents three nanotechnology-based anti-obesity strategies that target the white adipose tissues (WATs) and its vasculature for the reversal of obesity. These include inhibition of angiogenesis in the WATs, transformation of WATs to brown adipose tissues (BATs), and photothermal lipolysis of WATs. Compared to conventional therapy, the targeted-nanosystems have high tolerability, reduced side effects, and enhanced efficacy. These effects are reproducible using various nanocarriers (liposomes, polymeric and gold nanoparticles), thus providing a proof of concept that targeted nanotherapy can be a feasible strategy that can combat obesity and prevent its comorbidities.
Collapse
Affiliation(s)
- Nicole Remaliah Samantha Sibuyi
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, (DST/Mintek NIC), Biolabels Node, Department of Biotechnology, University of the Western Cape (UWC), Bellville, 7535, South Africa
| | - Koena Leah Moabelo
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, (DST/Mintek NIC), Biolabels Node, Department of Biotechnology, University of the Western Cape (UWC), Bellville, 7535, South Africa
- Nanobiotechnology Research Group, Department of Biotechnology, UWC, Bellville, 7535, South Africa
| | - Mervin Meyer
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, (DST/Mintek NIC), Biolabels Node, Department of Biotechnology, University of the Western Cape (UWC), Bellville, 7535, South Africa
| | - Martin Opiyo Onani
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, (DST/Mintek NIC), Biolabels Node, Department of Biotechnology, University of the Western Cape (UWC), Bellville, 7535, South Africa
- Organometallics and Nanomaterials, Department of Chemistry, UWC, Bellville, 7535, South Africa
| | - Admire Dube
- Infectious Disease Nanomedicine Research Group, School of Pharmacy, UWC, Bellville, 7535, South Africa
| | - Abram Madimabe Madiehe
- Department of Science and Technology/Mintek Nanotechnology Innovation Centre, (DST/Mintek NIC), Biolabels Node, Department of Biotechnology, University of the Western Cape (UWC), Bellville, 7535, South Africa.
- Nanobiotechnology Research Group, Department of Biotechnology, UWC, Bellville, 7535, South Africa.
| |
Collapse
|
18
|
Cheng CF, Ku HC, Cheng JJ, Chao SW, Li HF, Lai PF, Chang CC, Don MJ, Chen HH, Lin H. Adipocyte browning and resistance to obesity in mice is induced by expression of ATF3. Commun Biol 2019; 2:389. [PMID: 31667363 PMCID: PMC6813364 DOI: 10.1038/s42003-019-0624-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 09/13/2019] [Indexed: 02/07/2023] Open
Abstract
Billions of people have obesity-related metabolic syndromes such as diabetes and hyperlipidemia. Promoting the browning of white adipose tissue has been suggested as a potential strategy, but a drug still needs to be identified. Here, genetic deletion of activating transcription factor 3 (ATF3-/- ) in mice under a high-fat diet (HFD) resulted in obesity and insulin resistance, which was abrogated by virus-mediated ATF3 restoration. ST32da, a synthetic ATF3 inducer isolated from Salvia miltiorrhiza, promoted ATF3 expression to downregulate adipokine genes and induce adipocyte browning by suppressing the carbohydrate-responsive element-binding protein-stearoyl-CoA desaturase-1 axis. Furthermore, ST32da increased white adipose tissue browning and reduced lipogenesis in HFD-induced obese mice. The anti-obesity efficacy of oral ST32da administration was similar to that of the clinical drug orlistat. Our study identified the ATF3 inducer ST32da as a promising therapeutic drug for treating diet-induced obesity and related metabolic disorders.
Collapse
MESH Headings
- 3T3-L1 Cells
- Activating Transcription Factor 3/deficiency
- Activating Transcription Factor 3/genetics
- Activating Transcription Factor 3/metabolism
- Adipocytes, Brown/metabolism
- Adipocytes, Brown/pathology
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- Anti-Obesity Agents/pharmacology
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Body Temperature Regulation/physiology
- Diet, High-Fat/adverse effects
- Disease Models, Animal
- Humans
- Insulin Resistance
- Lipogenesis/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Obesity/genetics
- Obesity/metabolism
- Obesity/prevention & control
- Orlistat/pharmacology
- Plant Extracts/pharmacology
- Plants, Medicinal/chemistry
- Salvia miltiorrhiza/chemistry
Collapse
Affiliation(s)
- Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Department of Pediatrics, Tzu Chi University, Hualien, Taiwan
| | - Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, Taiwan
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jing-Jy Cheng
- Ph.D. Program in Clinical Drug Discovery from Botanical Herbs, Taipei Medical, University, Taipei, Taiwan
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Shi-Wei Chao
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Fen Li
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Pei-Fang Lai
- Department of Emergency Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Che-Chang Chang
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jaw Don
- National Research Institute of Chinese Medicine, Taipei, Taiwan
| | - Hsi-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Heng Lin
- Ph.D. Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
19
|
Sex-Specific Differences in Fat Storage, Development of Non-Alcoholic Fatty Liver Disease and Brain Structure in Juvenile HFD-Induced Obese Ldlr-/-.Leiden Mice. Nutrients 2019; 11:nu11081861. [PMID: 31405127 PMCID: PMC6723313 DOI: 10.3390/nu11081861] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Sex-specific differences play a role in metabolism, fat storage in adipose tissue, and brain structure. At juvenile age, brain function is susceptible to the effects of obesity; little is known about sex-specific differences in juvenile obesity. Therefore, this study examined sex-specific differences in adipose tissue and liver of high-fat diet (HFD)-induced obese mice, and putative alterations between male and female mice in brain structure in relation to behavioral changes during the development of juvenile obesity. METHODS In six-week-old male and female Ldlr-/-.Leiden mice (n = 48), the impact of 18 weeks of HFD-feeding was examined. Fat distribution, liver pathology and brain structure and function were analyzed imunohisto- and biochemically, in cognitive tasks and with MRI. RESULTS HFD-fed female mice were characterized by an increased perigonadal fat mass, pronounced macrovesicular hepatic steatosis and liver inflammation. Male mice on HFD displayed an increased mesenteric fat mass, pronounced adipose tissue inflammation and microvesicular hepatic steatosis. Only male HFD-fed mice showed decreased cerebral blood flow and reduced white matter integrity. CONCLUSIONS At young age, male mice are more susceptible to the detrimental effects of HFD than female mice. This study emphasizes the importance of sex-specific differences in obesity, liver pathology, and brain function.
Collapse
|
20
|
Cox B, Tsamou M, Vrijens K, Neven KY, Winckelmans E, de Kok TM, Plusquin M, Nawrot TS. A Co-expression Analysis of the Placental Transcriptome in Association With Maternal Pre-pregnancy BMI and Newborn Birth Weight. Front Genet 2019; 10:354. [PMID: 31110514 PMCID: PMC6501552 DOI: 10.3389/fgene.2019.00354] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/02/2019] [Indexed: 12/15/2022] Open
Abstract
Maternal body mass index (BMI) before pregnancy is known to affect both fetal growth and later-life health of the newborn, yet the implicated molecular mechanisms remain largely unknown. As the master regulator of the fetal environment, the placenta is a valuable resource for the investigation of processes involved in the developmental programming of metabolic health. We conducted a genome-wide placental transcriptome study aiming at the identification of functional pathways representing the molecular link between maternal BMI and fetal growth. We used RNA microarray (Agilent 8 × 60 K), medical records, and questionnaire data from 183 mother-newborn pairs from the ENVIRONAGE birth cohort study (Flanders, Belgium). Using a weighted gene co-expression network analysis, we identified 17 correlated gene modules. Three of these modules were associated with both maternal pre-pregnancy BMI and newborn birth weight. A gene cluster enriched for genes involved in immune response and myeloid cell differentiation was positively associated with maternal BMI and negatively with low birth weight. Two other gene modules, upregulated in association with maternal BMI as well as birth weight, were involved in processes related to organ and tissue development, with blood vessel morphogenesis and extracellular matrix structure as top Gene Ontology terms. In line with this, erythrocyte-, angiogenesis-, and extracellular matrix-related genes were among the identified hub genes. The association between maternal BMI and newborn weight was significantly mediated by gene expression for 5 of the hub genes (FZD4, COL15A1, GPR124, COL6A1, and COL1A1). As some of the identified hub genes have been linked to obesity in adults, our observation in placental tissue suggests that biological processes may be affected from prenatal life onwards, thereby identifying new molecular processes linking maternal BMI and fetal metabolic programming.
Collapse
Affiliation(s)
- Bianca Cox
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Maria Tsamou
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Karen Vrijens
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Kristof Y Neven
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Ellen Winckelmans
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Theo M de Kok
- Department of Toxicogenomics, Maastricht University, Maastricht, Netherlands
| | - Michelle Plusquin
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Tim S Nawrot
- Center for Environmental Sciences, Hasselt University, Hasselt, Belgium.,Department of Public Health, Environment and Health Unit, Leuven University (KU Leuven), Leuven, Belgium
| |
Collapse
|
21
|
Morrison MC, Verschuren L, Salic K, Verheij J, Menke A, Wielinga PY, Iruarrizaga‐Lejarreta M, Gole L, Yu W, Turner S, Caspers MP, Martínez‐Arranz I, Pieterman E, Stoop R, van Koppen A, van den Hoek AM, Mato JM, Hanemaaijer R, Alonso C, Kleemann R. Obeticholic Acid Modulates Serum Metabolites and Gene Signatures Characteristic of Human NASH and Attenuates Inflammation and Fibrosis Progression in Ldlr-/-.Leiden Mice. Hepatol Commun 2018; 2:1513-1532. [PMID: 30556039 PMCID: PMC6287481 DOI: 10.1002/hep4.1270] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023] Open
Abstract
Concerns have been raised about whether preclinical models sufficiently mimic molecular disease processes observed in nonalcoholic steatohepatitis (NASH) patients, bringing into question their translational value in studies of therapeutic interventions in the process of NASH/fibrosis. We investigated the representation of molecular disease patterns characteristic for human NASH in high-fat diet (HFD)-fed Ldlr-/-.Leiden mice and studied the effects of obeticholic acid (OCA) on these disease profiles. Multiplatform serum metabolomic profiles and genome-wide liver transcriptome from HFD-fed Ldlr-/-.Leiden mice were compared with those of NASH patients. Mice were profiled at the stage of mild (24 weeks HFD) and severe (34 weeks HFD) fibrosis, and after OCA intervention (24-34 weeks; 10 mg/kg/day). Effects of OCA were analyzed histologically, biochemically, by immunohistochemistry, using deuterated water technology (de novo collagen formation), and by its effect on the human-based transcriptomics and metabolomics signatures. The transcriptomics and metabolomics profile of Ldlr-/-.Leiden mice largely reflected the molecular signature of NASH patients. OCA modulated the expression of these molecular profiles and quenched specific proinflammatory-profibrotic pathways. OCA attenuated specific facets of cellular inflammation in liver (F4/80-positive cells) and reduced crown-like structures in adipose tissue. OCA reduced de novo collagen formation and attenuated further progression of liver fibrosis, but did not reduce fibrosis below the level before intervention. Conclusion: HFD-fed Ldlr-/-.Leiden mice recapitulate molecular transcriptomic and metabolomic profiles of NASH patients, and these signatures are modulated by OCA. Intervention with OCA in developing fibrosis reduces collagen deposition and de novo synthesis but does not resolve already manifest fibrosis in the period studied. These data show that human molecular signatures can be used to evaluate the translational character of preclinical models for NASH.
Collapse
Affiliation(s)
- Martine C. Morrison
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems BiologyNetherlands Organisation for Applied Scientific ResearchZeistThe Netherlands
| | - Kanita Salic
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Joanne Verheij
- Department of PathologyAmsterdam Medical CenterAmsterdamThe Netherlands
| | - Aswin Menke
- Department of PathologyTriskelion B.V.ZeistThe Netherlands
| | - Peter Y. Wielinga
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | | | - Laurent Gole
- Computational BioImage Analysis Unit, Agency of Science, Technology and Research (A*STAR), Institute of Molecular and Cell BiologySingapore
| | - Wei‐Miao Yu
- Computational BioImage Analysis Unit, Agency of Science, Technology and Research (A*STAR), Institute of Molecular and Cell BiologySingapore
| | | | - Martien P.M. Caspers
- Department of Microbiology and Systems BiologyNetherlands Organisation for Applied Scientific ResearchZeistThe Netherlands
| | | | - Elsbet Pieterman
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Reinout Stoop
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Arianne van Koppen
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | - Anita M. van den Hoek
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | | | - Roeland Hanemaaijer
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| | | | - Robert Kleemann
- Department of Metabolic Health ResearchNetherlands Organization for Applied Scientific ResearchLeidenThe Netherlands
| |
Collapse
|
22
|
Wang J, He L, Huwatibieke B, Liu L, Lan H, Zhao J, Li Y, Zhang W. Ghrelin Stimulates Endothelial Cells Angiogenesis through Extracellular Regulated Protein Kinases (ERK) Signaling Pathway. Int J Mol Sci 2018; 19:ijms19092530. [PMID: 30149681 PMCID: PMC6164813 DOI: 10.3390/ijms19092530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 02/07/2023] Open
Abstract
Adipose tissue is hyper-vascularized. Vessels in adipose tissue not only supply nutrients and oxygen to nourish adipocytes, but also provide cytokines that regulate mass and function of adipose tissue. Understanding the fundamental mechanisms how vessels modulate adipocyte functions would provide new therapeutic options for treatment of metabolic disease and obesity. In recent years, researches about ghrelin are focused on glucose and lipid metabolism, but its effect on vascular function remains uncharacterized. In the present study, ghrelin receptor gene deletion mice (Ghsr-/- mice) were used to study ghrelin-regulated vascular metabolism in white adipose tissue. Ghsr-/- mice demonstrated lower food intake, lower body weight, and resistance to high-fat diet-induced obesity. The number of vessels in white adipose tissue was decreased in Ghsr-/- mice when compared with wild type mice fed with high-fat diet. To further define ghrelin effects in vitro, we used endothelial progenitor cells from wild type and Ghsr-/- mice as well as human umbilical vein endothelial cells in our experiments. We found that ghrelin stimulated endothelial cells angiogenesis and migration through the MEK-ERK signaling pathway. [d-Lys3]-GHRP-6 and PD98059 could reverse the effects of ghrelin on endothelial cells. Our study indicates that ghrelin activates its receptor on endothelial cells to promote angiogenesis and migration via a mechanism involving the extracellular regulated protein kinases (ERK) signaling pathway.
Collapse
Affiliation(s)
- Jun Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Lin He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing 100191, China.
| | - Bahetiyaer Huwatibieke
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Lingchao Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - He Lan
- Department of Clinical Laboratory, Capital Medical University, Beijing 100053, China.
| | - Jing Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Yin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China.
| |
Collapse
|
23
|
Mao L, Lei J, Schoemaker MH, Ma B, Zhong Y, Lambers TT, Van Tol EAF, Zhou Y, Nie T, Wu D. Long-chain polyunsaturated fatty acids and extensively hydrolyzed casein-induced browning in a Ucp-1 reporter mouse model of obesity. Food Funct 2018; 9:2362-2373. [PMID: 29589625 DOI: 10.1039/c7fo01835e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Browning in adipose tissues, which can be affected by diet, may mitigate the detrimental effects of adiposity and improve longer-term metabolic health. Here, browning-inducing effects of long-chain polyunsaturated fatty acids, e.g., arachidonic acid (ARA)/docosahexaenoic acid (DHA) and extensively hydrolyzed casein (eHC) were investigated in uncoupling protein 1 (Ucp-1) reporter mice. To address the overall functionality, their potential role in supporting a healthy metabolic profile under obesogenic dietary challenges later in life was evaluated. At weaning Ucp1+/LUC reporter mice were fed a control low fat diet (LFD) with or without ARA + DHA, eHC or eHC + ARA + DHA for 8 weeks until week 12 after which interventions continued for another 12 weeks under a high-fat diet (HFD) challenge. Serology (metabolic responses and inflammation) and in vivo and ex vivo luciferase activity were determined; in the meantime browning-related proteins UCP-1 and the genes peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), PR domain containing 16 (PRDM16) and Ucp-1 were examined. ARA + DHA, eHC or their combination reduced body weight gain and adipose tissue weight compared to the HFD mice. The interventions induced Ucp-1 expression in adipose tissues prior to and during the HFD exposure. Ucp-1 induction was accompanied by higher PGC1a and PRDM16 expression. Glucose tolerance and insulin sensitivity were improved coinciding with lower serum cholesterol, triglycerides, free fatty acids, insulin, leptin, resistin, fibroblast growth factor 21, alanine aminotransferase, aspartate aminotransferase and higher adiponectin than the HFD group. HFD-associated increased systemic (IL-1β and TNF-α) and adipose tissue inflammation (F4/80, IL-1β, TNF-α, IL-6) was reduced. Studies in a Ucp-1 reporter mouse model revealed that early intervention with ARA/DHA and eHC improves metabolic flexibility and attenuates obesity during HFD challenge later in life. Increased browning is suggested as, at least, part of the underlying mechanism.
Collapse
Affiliation(s)
- Liufeng Mao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Morrison MC, Kleemann R, van Koppen A, Hanemaaijer R, Verschuren L. Key Inflammatory Processes in Human NASH Are Reflected in Ldlr -/-.Leiden Mice: A Translational Gene Profiling Study. Front Physiol 2018. [PMID: 29527177 PMCID: PMC5829089 DOI: 10.3389/fphys.2018.00132] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Introduction: It is generally accepted that metabolic inflammation in the liver is an important driver of disease progression in NASH and associated matrix remodeling/fibrosis. However, the exact molecular inflammatory mechanisms are poorly defined in human studies. Investigation of key pathogenic mechanisms requires the use of pre-clinical models, for instance for time-resolved studies. Such models must reflect molecular disease processes of importance in patients. Herein we characterized inflammation in NASH patients on the molecular level by transcriptomics and investigated whether key human disease pathways can be recapitulated experimentally in Ldlr−/−.Leiden mice, an established pre-clinical model of NASH. Methods: Human molecular inflammatory processes were defined using a publicly available NASH gene expression profiling dataset (GSE48452) allowing the comparison of biopsy-confirmed NASH patients with normal controls. Gene profiling data from high-fat diet (HFD)-fed Ldlr−/−.Leiden mice (GSE109345) were used for assessment of the translational value of these mice. Results: In human NASH livers, we observed regulation of 65 canonical pathways of which the majority was involved in inflammation (32%), lipid metabolism (16%), and extracellular matrix/remodeling (12%). A similar distribution of pathways across these categories, inflammation (36%), lipid metabolism (24%) and extracellular matrix/remodeling (8%) was observed in HFD-fed Ldlr−/−.Leiden mice. Detailed evaluation of these pathways revealed that a substantial proportion (11 out of 13) of human NASH inflammatory pathways was recapitulated in Ldlr−/−.Leiden mice. Furthermore, the activation state of identified master regulators of inflammation (i.e., specific transcription factors, cytokines, and growth factors) in human NASH was largely reflected in Ldlr−/−.Leiden mice, further substantiating its translational value. Conclusion: Human NASH is characterized by upregulation of specific inflammatory processes (e.g., “Fcγ Receptor-mediated Phagocytosis in Macrophages and Monocytes,” “PI3K signaling in B Lymphocytes”) and master regulators (e.g., TNF, CSF2, TGFB1). The majority of these processes and regulators are modulated in the same direction in Ldlr−/−.Leiden mice fed HFD with a human-like macronutrient composition, thus demonstrating that specific experimental conditions recapitulate human disease on the molecular level of disease pathways and upstream/master regulators.
Collapse
Affiliation(s)
- Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Arianne van Koppen
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Roeland Hanemaaijer
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| | - Lars Verschuren
- Department of Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, Netherlands
| |
Collapse
|
25
|
Miyazawa M, Subbaramaiah K, Bhardwaj P, Zhou XK, Wang H, Falcone DJ, Giri DD, Dannenberg AJ. Pioglitazone Inhibits Periprostatic White Adipose Tissue Inflammation in Obese Mice. Cancer Prev Res (Phila) 2017; 11:215-226. [PMID: 29222347 DOI: 10.1158/1940-6207.capr-17-0296] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 12/11/2022]
Abstract
Obesity is associated with an increased incidence of high-grade prostate cancer and poor prognosis for prostate cancer patients. Recently, we showed that obesity-related periprostatic white adipose tissue (WAT) inflammation, characterized by crown-like structures (CLS) consisting of dead or dying adipocytes surrounded by macrophages, was associated with high-grade prostate cancer. It is possible, therefore, that agents that suppress periprostatic WAT inflammation will alter the development or progression of prostate cancer. Pioglitazone, a ligand of PPARγ, is used to treat diabetes and possesses anti-inflammatory properties. Here, our main objectives were to determine whether pioglitazone inhibited obesity-related periprostatic WAT inflammation in mice and then to elucidate the underlying mechanism. Treatment with pioglitazone reduced the density of CLS in periprostatic fat and suppressed levels of TNFα, TGFβ, and the chemokine monocyte chemoattractant protein-1 (MCP-1). Importantly, the ability of pioglitazone to suppress periprostatic WAT inflammation was abrogated in MCP-1 knockout mice. Pioglitazone caused dose-dependent induction of both adiponectin, an anti-inflammatory adipokine, and its receptor AdipoR2 in cultured 3T3-L1 cells and in periprostatic WAT of obese mice. Pioglitazone blocked TNFα-mediated induction of MCP-1 in 3T3-L1 cells, an effect that was attenuated when either adiponectin or AdipoR2 were silenced. Taken together, pioglitazone-mediated induction of adiponectin suppressed the elevation in MCP-1 levels, thereby attenuating obesity-related periprostatic WAT inflammation. These findings strengthen the rationale for future efforts to determine whether targeting the PPARγ-adiponectin-MCP-1 axis will decrease periprostatic adipose inflammation and thereby reduce the risk of high-grade prostate cancer or improve outcomes for men with prostate cancer. Cancer Prev Res; 11(4); 215-26. ©2017 AACR.
Collapse
Affiliation(s)
- Miki Miyazawa
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Kotha Subbaramaiah
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Priya Bhardwaj
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Hanhan Wang
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York
| | - Domenick J Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Dilip D Giri
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
26
|
Berryhill GE, Lemay DG, Trott JF, Aimo L, Lock AL, Hovey RC. The Transcriptome of Estrogen-Independent Mammary Growth in Female Mice Reveals That Not All Mammary Glands Are Created Equally. Endocrinology 2017; 158:3126-3139. [PMID: 28938404 PMCID: PMC5659702 DOI: 10.1210/en.2017-00395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 07/21/2017] [Indexed: 01/22/2023]
Abstract
Allometric growth of ducts in the mammary glands (MGs) is widely held to be estrogen dependent. We previously discovered that the dietary fatty acid trans-10, cis-12 conjugated linoleic acid (CLA) stimulates estrogen-independent allometric growth and terminal end bud formation in ovariectomized mice. Given the similar phenotype induced by estrogen and CLA, we investigated the shared and/or divergent mechanisms underlying these changes. We confirmed MG growth induced by CLA is temporally distinct from that elicited by estrogen. We then used RNA sequencing to compare the transcriptome of the MG during similar proliferative and morphological states. Both estrogen and CLA affected the genes involved in proliferation. The transcriptome for estrogen-treated mice included canonical estrogen-induced genes, including Pgr, Areg, and Foxa1. In contrast, their expression was unchanged by CLA. However, CLA, but not estrogen, altered expression of a unique set of inflammation-associated genes, consistent with stromal changes. This CLA-altered signature included increased expression of epidermal growth factor receptor (EGFR) pathway components, consistent with the demonstration that CLA-induced MG growth is EGFR dependent. Our findings highlight a unique role for diet-induced inflammation that underlies estrogen-independent MG development.
Collapse
Affiliation(s)
- Grace E. Berryhill
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| | - Danielle G. Lemay
- UC Davis Genome Center, University of California Davis, Davis, California 95616-8521
- US Department of Agriculture, Agricultural Research Services, Western Human Nutrition Research Center, Davis, California 95616
| | - Josephine F. Trott
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| | - Lucila Aimo
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| | - Adam L. Lock
- Department of Animal Science, Michigan State University, East Lansing, Michigan 48824-1225
| | - Russell C. Hovey
- Department of Animal Science, University of California Davis, Davis, California 95616-8521
| |
Collapse
|
27
|
Butyrate Reduces HFD-Induced Adipocyte Hypertrophy and Metabolic Risk Factors in Obese LDLr-/-.Leiden Mice. Nutrients 2017; 9:nu9070714. [PMID: 28686216 PMCID: PMC5537829 DOI: 10.3390/nu9070714] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 12/18/2022] Open
Abstract
Adipose tissue (AT) has a modulating role in obesity-induced metabolic complications like type 2 diabetes mellitus (T2DM) via the production of so-called adipokines such as leptin, adiponectin, and resistin. The adipokines are believed to influence other tissues and to affect insulin resistance, liver function, and to increase the risk of T2DM. In this study, we examined the impact of intervention with the short-chain fatty acid butyrate following a high-fat diet (HFD) on AT function and other metabolic risk factors associated with obesity and T2DM in mice during mid- and late life. In both mid- and late adulthood, butyrate reduced HFD-induced adipocyte hypertrophy and elevations in leptin levels, which were associated with body weight, and cholesterol and triglyceride levels. HFD feeding stimulated macrophage accumulation primarily in epididymal AT in both mid- and late life adult mice, which correlated with liver inflammation in late adulthood. In late-adult mice, butyrate diminished increased insulin levels, which were related to adipocyte size and macrophage content in epididymal AT. These results suggest that dietary butyrate supplementation is able to counteract HFD-induced detrimental changes in AT function and metabolic outcomes in late life. These changes underlie the obesity-induced elevated risk of T2DM, and therefore it is suggested that butyrate has potential to attenuate risk factors associated with obesity and T2DM.
Collapse
|
28
|
Schoemaker MH, Kleemann R, Morrison MC, Verheij J, Salic K, van Tol EAF, Kooistra T, Wielinga PY. A casein hydrolysate based formulation attenuates obesity and associated non-alcoholic fatty liver disease and atherosclerosis in LDLr-/-.Leiden mice. PLoS One 2017; 12:e0180648. [PMID: 28678821 PMCID: PMC5498059 DOI: 10.1371/journal.pone.0180648] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/19/2017] [Indexed: 11/29/2022] Open
Abstract
Background Obesity frequently associates with the development of non-alcoholic fatty liver disease (NAFLD) and atherosclerosis. Chronic inflammation in white adipose tissue (WAT) seems to be an important driver of these manifestations. Objective This study investigated a combination of an extensively hydrolyzed casein (eHC), docosahexaenoic acid (DHA), arachidonic acid (ARA), and Lactobacillus Rhamnosus GG (LGG) (together referred to as nutritional ingredients, NI) on the development of obesity, metabolic risk factors, WAT inflammation, NAFLD and atherosclerosis in high-fat diet-fed LDLr-/-.Leiden mice, a model that mimics disease development in humans. Methods LDLr-/-.Leiden male mice (n = 15/group) received a high-fat diet (HFD, 45 Kcal%) for 21 weeks with or without the NI (23.7% eHC, 0.083% DHA, 0.166% ARA; all w/w and 1x109 CFU LGG gavage 3 times/week). HFD and HFD+NI diets were isocaloric. A low fat diet (LFD, 10 Kcal%) was used for reference. Body weight, food intake and metabolic risk factors were assessed over time. At week 21, tissues were analyzed for WAT inflammation (crown-like structures), NAFLD and atherosclerosis. Effects of the individual NI components were explored in a follow-up experiment (n = 7/group). Results When compared to HFD control, treatment with the NI strongly reduced body weight to levels of the LFD group, and significantly lowered (P<0.01) plasma insulin, cholesterol, triglycerides, leptin and serum amyloid A (P<0.01). NI also reduced WAT mass and inflammation. Strikingly, NI treatment significantly reduced macrovesicular steatosis, lobular inflammation and liver collagen (P<0.05), and attenuated atherosclerosis development (P<0.01). Of the individual components, the effects of eHC were most pronounced but could not explain the entire effects of the NI formulation. Conclusions A combination of eHC, ARA, DHA and LGG attenuates obesity and associated cardiometabolic diseases (NAFLD, atherosclerosis) in LDLr-/-.Leiden mice. The observed reduction of inflammation in adipose tissue and in the liver provides a rationale for these comprehensive health effects.
Collapse
Affiliation(s)
- Marieke H. Schoemaker
- Mead Johnson Pediatric Nutrition Institute, Global R&D, Nijmegen, the Netherlands
- * E-mail:
| | - Robert Kleemann
- TNO, the Netherlands Organization for Scientific Research, Metabolic Health Research, Leiden, the Netherlands
| | - Martine C. Morrison
- TNO, the Netherlands Organization for Scientific Research, Metabolic Health Research, Leiden, the Netherlands
| | - Joanne Verheij
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Kanita Salic
- TNO, the Netherlands Organization for Scientific Research, Metabolic Health Research, Leiden, the Netherlands
| | - Eric A. F. van Tol
- Mead Johnson Pediatric Nutrition Institute, Global R&D, Nijmegen, the Netherlands
| | - Teake Kooistra
- TNO, the Netherlands Organization for Scientific Research, Metabolic Health Research, Leiden, the Netherlands
| | - Peter Y. Wielinga
- TNO, the Netherlands Organization for Scientific Research, Metabolic Health Research, Leiden, the Netherlands
| |
Collapse
|
29
|
Viglino D, Jullian-Desayes I, Minoves M, Aron-Wisnewsky J, Leroy V, Zarski JP, Tamisier R, Joyeux-Faure M, Pépin JL. Nonalcoholic fatty liver disease in chronic obstructive pulmonary disease. Eur Respir J 2017; 49:49/6/1601923. [DOI: 10.1183/13993003.01923-2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/23/2017] [Indexed: 12/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is independently linked to cardiometabolic morbidity and mortality. Low-grade inflammation, oxidative stress and ectopic fat, common features of chronic obstructive pulmonary disease (COPD), might contribute to the development of NAFLD.We aimed to investigate the prevalence of NAFLD and to evaluate the relationship between various types of liver damage and COPD severity, comorbidities and circulating inflammatory cytokines. Validated noninvasive tests (FibroMax: SteatoTest, NashTest and FibroTest) were used to assess steatosis, nonalcoholic steatohepatitis (NASH) and liver fibrosis. Patients underwent an objective assessment of COPD comorbidities, including sleep studies. Biological parameters included a complete lipid profile and inflammatory markers.In COPD patients the prevalence of steatosis, NASH and fibrosis were 41.4%, 36.9% and 61.3%, respectively. In multivariate analysis, SteatoTest and FibroTest were significantly associated with sex, body mass index (BMI), untreated sleep apnoea and insulin resistance, and, in addition, COPD Global Initiative for Chronic Obstructive Lung Disease stage for SteatoTest. Patients with steatosis had higher tumour necrosis factor-α levels and those with NASH or a combination of liver damage types had raised leptin levels after adjustment for age, sex and BMI.We concluded that NAFLD is highly prevalent in COPD and might contribute to cardiometabolic comorbidities.
Collapse
|