1
|
Silva MGFR, Luchiari AC, Medeiros I, de Souza AM, Serquiz AC, Martins FF, de Moura SAB, Camillo CS, de Medeiros SRB, Pais TDS, Passos TS, Galeno DML, Morais AHDA. Evaluation of the Effects of Diet-Induced Obesity in Zebrafish ( Danio rerio): A Comparative Study. Nutrients 2024; 16:3398. [PMID: 39408365 PMCID: PMC11479130 DOI: 10.3390/nu16193398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
OBJECTIVES This study aimed to compare diet-induced obesity (DIO) models in zebrafish and investigate the complications and differences between sexes in biochemical and inflammatory parameters. METHODS Adult animals of both sexes were divided into four groups (n = 50) and fed for eight weeks: control group 1: Artemia sp. (15-30 mg/day/fish); control group 2: commercial fish food (3.5% of average weight); obesity group 1: pasteurized egg yolk powder + soybean oil (5% of average weight); obesity group 2: Artemia sp. (60-120 mg/day/fish). Dietary intake, caloric intake and efficiency, body mass index, biochemical, inflammatory, behavioral, histopathological, and stereological parameters, and inflammation-related gene expression were investigated. RESULTS Obesity group 1 was the most indicated to investigate changes in the anxious behavioral profile (p < 0.05), triglyceride elevation [52.67 (1.2) mg/dL], adipocyte hypertrophy [67.8 (18.1) µm2; p = 0.0004], and intestinal inflammation. Obesity group 2 was interesting to investigate in terms of weight gain [167 mg; p < 0.0001), changes in fasting glucose [48.33 (4.14) mg/dL; p = 0.003), and inflammatory parameters [IL-6: 4.24 (0.18) pg/mL; p = 0.0015]. CONCLUSIONS Furthermore, both DIO models evaluated in the present study were effective in investigating hepatic steatosis. The data also highlighted that sex influences inflammatory changes and fasting blood glucose levels, which were higher in males (p > 0.05). The results show new metabolic routes to be explored in relation to DIO in zebrafish.
Collapse
Affiliation(s)
- Maria Gabriela F. R. Silva
- Nutrition Postgraduate Program, Center of Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (M.G.F.R.S.); (T.S.P.)
| | - Ana Carolina Luchiari
- Psychobiology Postgraduate Program, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Isaiane Medeiros
- Biochemistry and Molecular Biology Postgraduate Program, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (I.M.); (S.R.B.d.M.); (T.d.S.P.)
| | - Augusto M. de Souza
- Biotechnology Program—Northeast Biotechnology Network (RENORBIO), Technology Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Alexandre C. Serquiz
- Department of Nutrition, Federal University of Paraíba, João Pessoa 58051-900, PB, Brazil;
| | - Fabiane F. Martins
- Department of Morphology, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (F.F.M.); (S.A.B.d.M.); (C.S.C.)
| | - Sérgio A. B. de Moura
- Department of Morphology, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (F.F.M.); (S.A.B.d.M.); (C.S.C.)
| | - Christina S. Camillo
- Department of Morphology, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (F.F.M.); (S.A.B.d.M.); (C.S.C.)
| | - Silvia Regina B. de Medeiros
- Biochemistry and Molecular Biology Postgraduate Program, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (I.M.); (S.R.B.d.M.); (T.d.S.P.)
- Department of Cell Biology and Genetics, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Tatiana dos S. Pais
- Biochemistry and Molecular Biology Postgraduate Program, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (I.M.); (S.R.B.d.M.); (T.d.S.P.)
| | - Thaís S. Passos
- Nutrition Postgraduate Program, Center of Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (M.G.F.R.S.); (T.S.P.)
- Department of Nutrition, Center of Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Denise M. L. Galeno
- Multicenter Postgraduate Program in Physiological Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil;
| | - Ana Heloneida de A. Morais
- Nutrition Postgraduate Program, Center of Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (M.G.F.R.S.); (T.S.P.)
- Biochemistry and Molecular Biology Postgraduate Program, Center of Biosciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil; (I.M.); (S.R.B.d.M.); (T.d.S.P.)
- Department of Nutrition, Center of Health Sciences, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| |
Collapse
|
2
|
Della Pepa G, Patrício BG, Carli F, Sabatini S, Astiarraga B, Ferrannini E, Camastra S, Gastaldelli A. GLP-1 Receptor Agonist Treatment Improves Fasting and Postprandial Lipidomic Profiles Independently of Diabetes and Weight Loss. Diabetes 2024; 73:1605-1614. [PMID: 38976482 DOI: 10.2337/db23-0972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
Treatment with glucagon-like peptide 1 receptor agonists reduces liver steatosis and cardiometabolic risk (CMR). Few data are available on lipid metabolism, and no information is available on the postprandial lipidomic profile. Thus, we investigated how exenatide treatment changes lipid metabolism and composition during fasting and after a mixed-meal tolerance test (MMTT) in adults with severe obesity without diabetes. Thirty individuals (26 females and 4 males, 30-60 years old, BMI >40 kg/m2, HbA1c 5.76%) were assigned (1:1) to diet with exenatide 10 μg twice daily treatment (n = 15) or without treatment as control (n = 15) for 3 months. Fasting and postprandial lipidomic profile (by liquid chromatography quadrupole time-of-flight mass spectrometry) and fatty acid metabolism (following a 6-h MMTT/tracer study) and composition (by gas chromatography-mass spectrometry) were evaluated before and after treatment. Both groups had slight weight loss (-5.5% vs. -1.9%, exenatide vs. control; P = 0.052). During fasting, exenatide, compared with control, reduced some ceramides (CERs) and lysophosphatidylcholines (LPCs) previously associated with CMR, while relatively increasing unsaturated phospholipid species (phosphatidylcholine [PC], LPC) with protective effects on CMR, although concentrations of total lipid species were unchanged. During MMTT, both groups showed suppressed lipolysis equal to baseline, but exenatide significantly lowered free fatty acid clearance and postprandial triacyclglycerol (TAG) concentrations, particularly saturated TAGs with 44-54 carbons. Exenatide also reduced some postprandial CERs, PCs, and LPCs previously linked to CMR. These changes in lipidomic profile remained statistically significant after adjusting for weight loss. Exenatide improved fasting and postprandial lipidomic profiles associated with CMR mainly by reducing saturated postprandial TAGs and CERs independently of weight loss and diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | - Bárbara G Patrício
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
- Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Fabrizia Carli
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
| | - Silvia Sabatini
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
| | - Brenno Astiarraga
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Pere Virgili Institute for Health Research, Terragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ele Ferrannini
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefania Camastra
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Amalia Gastaldelli
- National Research Council, Institute of Clinical Physiology, Pisa, Italy
- Sant'Anna School of Advanced Studies, Pisa, Italy
| |
Collapse
|
3
|
Beygi M, Ahi S, Zolghadri S, Stanek A. Management of Metabolic-Associated Fatty Liver Disease/Metabolic Dysfunction-Associated Steatotic Liver Disease: From Medication Therapy to Nutritional Interventions. Nutrients 2024; 16:2220. [PMID: 39064665 PMCID: PMC11279539 DOI: 10.3390/nu16142220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/30/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common long-lasting liver disease that affects millions of people around the world. It is best identified with a hepatic fat build-up that ultimately leads to inflammation and damage. The classification and nomenclature of NAFLD have long been a controversial topic, until 2020 when a group of international experts recommended substituting NAFLD with MAFLD (metabolic dysfunction-associated FLD). MAFLD was then terminologically complemented in 2023 by altering it to MASLD, i.e., metabolic dysfunction-associated steatotic liver disease (MASLD). Both the MAFLD and the MASLD terminologies comprise the metabolic element of the disorder, as they offer diagnostic benchmarks that are embedded in the metabolic risk factors that underlie the disease. MASLD (as a multisystemic disease) provides a comprehensive definition that includes a larger population of patients who are at risk of liver morbidity and mortality, as well as adverse cardiovascular and diabetes outcomes. MASLD highlights metabolic risks in lean or normal weight individuals, a factor that has not been accentuated or discussed in previous guidelines. Novel antihyperglycemic agents, anti-hyperlipidemic drugs, lifestyle modifications, nutritional interventions, and exercise therapies have not been extensively studied in MAFLD and MASLD. Nutrition plays a vital role in managing both conditions, where centralizing on a diet rich in whole vegetables, fruits, foods, healthy fats, lean proteins, and specific nutrients (e.g., omega-3 fatty acids and fibers) can improve insulin resistance and reduce inflammation. Thus, it is essential to understand the role of nutrition in managing these conditions and to work with patients to develop an individualized plan for optimal health. This review discusses prevention strategies for NAFLD/MAFLD/MASLD management, with particular attention to nutrition and lifestyle correction.
Collapse
Affiliation(s)
- Mohammad Beygi
- Department of Agricultural Biotechnology, College of Agriculture, Isfahan University of Technology (IUT), Isfahan 8415683111, Iran;
| | - Salma Ahi
- Research Center for Noncommunicable Diseases, Jahrom University of Medical Sciences, Jahrom 7414846199, Iran;
| | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran
| | - Agata Stanek
- Department of Internal Medicine, Angiology and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Batorego 15 St., 41-902 Bytom, Poland
| |
Collapse
|
4
|
Wang Y, Wu G, Xiao F, Yin H, Yu L, Chen Y, Shehzad Q, Xu L, Zhang H, Jin Q, Wang X. Fatty acid composition in erythrocytes and coronary artery disease risk: a case-control study in China. Food Funct 2024; 15:7174-7188. [PMID: 38895817 DOI: 10.1039/d4fo00016a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background and aims: There is limited and conflicting evidence about the association of erythrocyte fatty acids with coronary artery disease (CAD), particularly in China where the CAD rates are high. Our study aimed to explore the association between erythrocyte fatty acid composition and CAD risk in Chinese adults. Methods: Erythrocyte fatty acids of 314 CAD patients and 314 matched controls were measured by gas chromatography. Multivariable conditional logistic regression and restricted cubic spline models were used to explore the odds ratio with 95% confidence interval (OR, 95% CI) and potential association between erythrocyte fatty acids and CAD risk. Principal component analysis (PCA) was used to analyze further the potential role of various erythrocyte fatty acid patterns in relation to CAD risk. Results: Significant inverse associations were observed between high levels of erythrocyte total n-3 polyunsaturated fatty acids (n-3 PUFA) [ORT3-T1 = 0.18 (0.12, 0.28)], monounsaturated fatty acids (MUFA) [ORT3-T1 = 0.21 (0.13, 0.32)], and the risk of CAD. Conversely, levels of saturated fatty acids (SFAs) and n-6 polyunsaturated fatty acids (n-6 PUFAs) were positively associated with CAD risk [ORT3-T1 = 3.33 (2.18, 5.13), ORT3-T1 = 1.61 (1.06, 2.43)]. No significant association was observed between CAD risk and total trans fatty acids. Additionally, the PCA identifies four new fatty acid patterns (FAPs). The risk of CAD was significantly positively associated with FAP1 and FAP2, while being negatively correlated with FAP3 and FAP4. Conclusion: The different types of erythrocyte fatty acids may significantly alter susceptibility to CAD. Elevated levels of n-3-PUFAs and MUFAs are considered as protective biomarkers against CAD, while SFAs and n-6 PUFAs may be associated with higher CAD risk in Chinese adults. The risk of CAD was positively associated with FAP1 and FAP2, and negatively associated with FAP3 and FAP4. Combinations of erythrocyte fatty acids may be more important markers of CAD development than individual fatty acids or their subgroups.
Collapse
Affiliation(s)
- Yongjin Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Gangcheng Wu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Feng Xiao
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Hongming Yin
- Department of Laboratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Le Yu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
- National Center of Technology Innovation for Dairy, Hohhot 010000, China
| | - Yujia Chen
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Qayyum Shehzad
- School of Fundamental Sciences, Massey University, Palmerston North 4410, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| | - Lirong Xu
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, China
| | - Hui Zhang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Qingzhe Jin
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| | - Xingguo Wang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, International Joint Research Laboratory for Lipid Nutrition and Safety, National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
5
|
Armandi A, Bugianesi E. Dietary and pharmacological treatment in patients with metabolic-dysfunction associated steatotic liver disease. Eur J Intern Med 2024; 122:20-27. [PMID: 38262842 DOI: 10.1016/j.ejim.2024.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/30/2023] [Accepted: 01/07/2024] [Indexed: 01/25/2024]
Abstract
Metabolic-dysfunction Associated Steatotic Liver Disease (MASLD) is a disease spectrum encompassing liver injury with progressive severity, tightly connected to the metabolic syndrome. Management of MASLD mostly relies on lifestyle change aiming at improving metabolic homeostasis and insulin resistance. A Mediterranean-like dietary pattern and individualized lifestyle interventions are the cornerstone of MASLD treatment. A careful evaluation of alcohol intake and active treatment of all metabolic co-morbidities are recommended. In the MASLD spectrum, the population with liver inflammation and enhanced fibrogenesis (MASH - Metabolic-dysfunction associated steatohepatitis) can progress to advanced liver disease and has been addressed as "at-risk MASH", eligible to pharmacological treatment according to FDA and EMA. Currently there is a robust therapeutic pipeline across a variety of new targets to resolve MASH or reverse fibrosis, or both. Some of these therapies have beneficial effects that extend beyond the liver, such as effects on glycaemic control, lipid profile and weight loss. For "at-risk" MASH, reversal of fibrosis by one stage or resolution of MASH with no worsening in fibrosis as a surrogate end-point will need to be accompanied by overall survival benefits. In this review, we summarize the current evidence on lifestyle interventions in MASLD as well as pharmacological approaches for fibrosing MASH that have progressed to phase II and phase III clinical trials.
Collapse
Affiliation(s)
- Angelo Armandi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Italy.
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Italy
| |
Collapse
|
6
|
Jia X, Chen Q, Wu H, Liu H, Jing C, Gong A, Zhang Y. Exploring a novel therapeutic strategy: the interplay between gut microbiota and high-fat diet in the pathogenesis of metabolic disorders. Front Nutr 2023; 10:1291853. [PMID: 38192650 PMCID: PMC10773723 DOI: 10.3389/fnut.2023.1291853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024] Open
Abstract
In the past two decades, the rapid increase in the incidence of metabolic diseases, including obesity, diabetes, dyslipidemia, non-alcoholic fatty liver disease, hypertension, and hyperuricemia, has been attributed to high-fat diets (HFD) and decreased physical activity levels. Although the phenotypes and pathologies of these metabolic diseases vary, patients with these diseases exhibit disease-specific alterations in the composition and function of their gut microbiota. Studies in germ-free mice have shown that both HFD and gut microbiota can promote the development of metabolic diseases, and HFD can disrupt the balance of gut microbiota. Therefore, investigating the interaction between gut microbiota and HFD in the pathogenesis of metabolic diseases is crucial for identifying novel therapeutic strategies for these diseases. This review takes HFD as the starting point, providing a detailed analysis of the pivotal role of HFD in the development of metabolic disorders. It comprehensively elucidates the impact of HFD on the balance of intestinal microbiota, analyzes the mechanisms underlying gut microbiota dysbiosis leading to metabolic disruptions, and explores the associated genetic factors. Finally, the potential of targeting the gut microbiota as a means to address metabolic disturbances induced by HFD is discussed. In summary, this review offers theoretical support and proposes new research avenues for investigating the role of nutrition-related factors in the pathogenesis of metabolic disorders in the organism.
Collapse
Affiliation(s)
- Xiaokang Jia
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Qiliang Chen
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huiwen Wu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Hongbo Liu
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Chunying Jing
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Aimin Gong
- School of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Yuanyuan Zhang
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Meek CL, Stewart ZA, Feig DS, Furse S, Neoh SL, Koulman A, Murphy HR. Metabolomic insights into maternal and neonatal complications in pregnancies affected by type 1 diabetes. Diabetologia 2023; 66:2101-2116. [PMID: 37615689 PMCID: PMC10542716 DOI: 10.1007/s00125-023-05989-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/19/2023] [Indexed: 08/25/2023]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes in pregnancy is associated with suboptimal pregnancy outcomes, attributed to maternal hyperglycaemia and offspring hyperinsulinism (quantifiable by cord blood C-peptide). We assessed metabolomic patterns associated with risk factors (maternal hyperglycaemia, diet, BMI, weight gain) and perinatal complications (pre-eclampsia, large for gestational age [LGA], neonatal hypoglycaemia, hyperinsulinism) in the Continuous Glucose Monitoring in Women with Type 1 Diabetes in Pregnancy Trial (CONCEPTT). METHODS A total of 174 CONCEPTT participants gave ≥1 non-fasting serum sample for the biorepository at 12 gestational weeks (147 women), 24 weeks (167 women) and 34 weeks (160 women) with cord blood from 93 infants. Results from untargeted metabolite analysis (ultrahigh performance LC-MS) are presented as adjusted logistic/linear regression of maternal and cord blood metabolites, risk factors and perinatal complications using a modified Bonferroni limit of significance for dependent variables. RESULTS Maternal continuous glucose monitoring time-above-range (but not BMI or excessive gestational weight gain) was associated with increased triacylglycerols in maternal blood and increased carnitines in cord blood. LGA, adiposity, neonatal hypoglycaemia and offspring hyperinsulinism showed distinct metabolite profiles. LGA was associated with increased carnitines, steroid hormones and lipid metabolites, predominantly in the third trimester. However, neonatal hypoglycaemia and offspring hyperinsulinism were both associated with metabolite changes from the first trimester, featuring triacylglycerols or dietary phenols. Pre-eclampsia was associated with increased abundance of phosphatidylethanolamines, a membrane phospholipid, at 24 weeks. CONCLUSIONS/INTERPRETATION Altered lipid metabolism is a key pathophysiological feature of type 1 diabetes pregnancy. New strategies for optimising maternal diet and insulin dosing from the first trimester are needed to improve pregnancy outcomes in type 1 diabetes.
Collapse
Affiliation(s)
- Claire L Meek
- Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Zoe A Stewart
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Denice S Feig
- Mount Sinai Hospital, Sinai Health System, New York, NY, USA
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Samuel Furse
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Core Metabolomics and Lipidomics Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Sandra L Neoh
- Department of Endocrinology, Austin Health, Melbourne, VIC, Australia
- Department of Endocrinology, Northern Health, Melbourne, VIC, Australia
| | - Albert Koulman
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Core Metabolomics and Lipidomics Laboratory, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Helen R Murphy
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
8
|
Tuero C, Becerril S, Ezquerro S, Neira G, Frühbeck G, Rodríguez A. Molecular and cellular mechanisms underlying the hepatoprotective role of ghrelin against NAFLD progression. J Physiol Biochem 2023; 79:833-849. [PMID: 36417140 DOI: 10.1007/s13105-022-00933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
The underlying mechanisms for the development and progression of nonalcoholic fatty liver disease (NAFLD) are complex and multifactorial. Within the last years, experimental and clinical evidences support the role of ghrelin in the development of NAFLD. Ghrelin is a gut hormone that plays a major role in the short-term regulation of appetite and long-term regulation of adiposity. The liver constitutes a target for ghrelin, where this gut-derived peptide triggers intracellular pathways regulating lipid metabolism, inflammation, and fibrosis. Interestingly, circulating ghrelin levels are altered in patients with metabolic diseases, such as obesity, type 2 diabetes, and metabolic syndrome, which, in turn, are well-known risk factors for the pathogenesis of NAFLD. This review summarizes the molecular and cellular mechanisms involved in the hepatoprotective action of ghrelin, including the reduction of hepatocyte lipotoxicity via autophagy and fatty acid β-oxidation, mitochondrial dysfunction, endoplasmic reticulum stress and programmed cell death, the reversibility of the proinflammatory phenotype in Kupffer cells, and the inactivation of hepatic stellate cells. Together, the metabolic and inflammatory pathways regulated by ghrelin in the liver support its potential as a therapeutic target to prevent NAFLD in patients with metabolic disorders.
Collapse
Affiliation(s)
- Carlota Tuero
- Department of General Surgery, Clínica Universidad de Navarra, School of Medicine, University of Navarra, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Silvia Ezquerro
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008, Pamplona, Irunlarrea 1, Spain.
- CIBER Fisiopatología de La Obesidad Y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
9
|
Hendriks D, Brouwers JF, Hamer K, Geurts MH, Luciana L, Massalini S, López-Iglesias C, Peters PJ, Rodríguez-Colman MJ, Chuva de Sousa Lopes S, Artegiani B, Clevers H. Engineered human hepatocyte organoids enable CRISPR-based target discovery and drug screening for steatosis. Nat Biotechnol 2023; 41:1567-1581. [PMID: 36823355 PMCID: PMC10635827 DOI: 10.1038/s41587-023-01680-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 01/19/2023] [Indexed: 02/25/2023]
Abstract
The lack of registered drugs for nonalcoholic fatty liver disease (NAFLD) is partly due to the paucity of human-relevant models for target discovery and compound screening. Here we use human fetal hepatocyte organoids to model the first stage of NAFLD, steatosis, representing three different triggers: free fatty acid loading, interindividual genetic variability (PNPLA3 I148M) and monogenic lipid disorders (APOB and MTTP mutations). Screening of drug candidates revealed compounds effective at resolving steatosis. Mechanistic evaluation of effective drugs uncovered repression of de novo lipogenesis as the convergent molecular pathway. We present FatTracer, a CRISPR screening platform to identify steatosis modulators and putative targets using APOB-/- and MTTP-/- organoids. From a screen targeting 35 genes implicated in lipid metabolism and/or NAFLD risk, FADS2 (fatty acid desaturase 2) emerged as an important determinant of hepatic steatosis. Enhancement of FADS2 expression increases polyunsaturated fatty acid abundancy which, in turn, reduces de novo lipogenesis. These organoid models facilitate study of steatosis etiology and drug targets.
Collapse
Affiliation(s)
- Delilah Hendriks
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| | - Jos F Brouwers
- Research Group Analysis Techniques in the Life Sciences, School of Life Sciences and Technology, Avans University of Applied Sciences, Breda, The Netherlands
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Karien Hamer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Maarten H Geurts
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Léa Luciana
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Simone Massalini
- The Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Carmen López-Iglesias
- The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Peter J Peters
- The Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, The Netherlands
| | - Maria J Rodríguez-Colman
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Benedetta Artegiani
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands.
- The Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Utrecht, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
- The Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.
- University Medical Center Utrecht, Utrecht, The Netherlands.
- Pharma, Research and Early Development of F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
10
|
White SL, Koulman A, Ozanne SE, Furse S, Poston L, Meek CL. Towards Precision Medicine in Gestational Diabetes: Pathophysiology and Glycemic Patterns in Pregnant Women With Obesity. J Clin Endocrinol Metab 2023; 108:2643-2652. [PMID: 36950879 PMCID: PMC10807907 DOI: 10.1210/clinem/dgad168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/22/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
AIMS Precision medicine has revolutionized our understanding of type 1 diabetes and neonatal diabetes but has yet to improve insight into gestational diabetes mellitus (GDM), the most common obstetric complication and strongly linked to obesity. Here we explored if patterns of glycaemia (fasting, 1 hour, 2 hours) during the antenatal oral glucose tolerance test (OGTT), reflect distinct pathophysiological subtypes of GDM as defined by insulin secretion/sensitivity or lipid profiles. METHODS 867 pregnant women with obesity (body mass index ≥ 30 kg/m2) from the UPBEAT trial (ISRCTN 89971375) were assessed for GDM at 28 weeks' gestation (75 g oral glucose tolerance test OGTT; World Health Organization criteria). Lipid profiling of the fasting plasma OGTT sample was undertaken using direct infusion mass spectrometry and analyzed by logistic/linear regression, with and without adjustment for confounders. Insulin secretion and sensitivity were characterized by homeostatic model assessment 2b and 2s, respectively. RESULTS In women who developed GDM (n = 241), patterns of glycaemia were associated with distinct clinical and biochemical characteristics and changes to lipid abundance in the circulation. Severity of glucose derangement, rather than pattern of postload glycaemia, was most strongly related to insulin action and lipid abundance/profile. Unexpectedly, women with isolated postload hyperglycemia had comparable insulin secretion and sensitivity to euglycemic women, potentially indicative of a novel mechanistic pathway. CONCLUSIONS Patterns of glycemia during the OGTT may contribute to a precision approach to GDM as assessed by differences in insulin resistance/secretion. Further research is indicated to determine if isolated postload hyperglycemia reflects a different mechanistic pathway for targeted management.
Collapse
Affiliation(s)
- Sara L White
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, SE1 7EH, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
| | - Susan E Ozanne
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
| | - Samuel Furse
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
| | - Lucilla Poston
- Department of Women and Children’s Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, SE1 7EH, UK
| | - Claire L Meek
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Cambridge, CB2 0QQ, UK
- Department of Clinical Biochemistry/Wolfson Diabetes & Endocrine Clinic, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| |
Collapse
|
11
|
Taylor R, Barnes A, Hollingsworth K, Irvine K, Solovyova A, Clark L, Kelly T, Martin-Ruiz C, Romeres D, Koulman A, Meek C, Jenkins B, Cobelli C, Holman R. Aetiology of Type 2 diabetes in people with a 'normal' body mass index: testing the personal fat threshold hypothesis. Clin Sci (Lond) 2023; 137:1333-1346. [PMID: 37593846 PMCID: PMC10472166 DOI: 10.1042/cs20230586] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/19/2023]
Abstract
Weight loss in overweight or obese individuals with Type 2 diabetes (T2D) can normalize hepatic fat metabolism, decrease fatty acid oversupply to β cells and restore normoglycaemia. One in six people has BMI <27 kg/m2 at diagnosis, and their T2D is assumed to have different aetiology. The Personal Fat Threshold hypothesis postulated differing individual thresholds for lipid overspill and adverse effects on β-cell function. To test this hypothesis, people with Type 2 diabetes and body mass index <27kg/m2 (n = 20) underwent repeated 5% weight loss cycles. Metabolic assessments were carried out at stable weight after each cycle and after 12 months. To determine how closely metabolic features returned to normal, 20 matched normoglycemic controls were studied once. Between baseline and 12 months: BMI fell (mean ± SD), 24.8 ± 0.4 to 22.5 ± 0.4 kg/m2 (P<0.0001) (controls: 21.5 ± 0.5); total body fat, 32.1 ± 1.5 to 27.6 ± 1.8% (P<0.0001) (24.6 ± 1.5). Liver fat content and fat export fell to normal as did fasting plasma insulin. Post-meal insulin secretion increased but remained subnormal. Sustained diabetes remission (HbA1c < 48 mmol/mol off all glucose-lowering agents) was achieved by 70% (14/20) by initial weight loss of 6.5 (5.5-10.2)%. Correction of concealed excess intra-hepatic fat reduced hepatic fat export, with recovery of β-cell function, glycaemic improvement in all and return to a non-diabetic metabolic state in the majority of this group with BMI <27 kg/m2 as previously demonstrated for overweight or obese groups. The data confirm the Personal Fat Threshold hypothesis: aetiology of Type 2 diabetes does not depend on BMI. This pathophysiological insight has major implications for management.
Collapse
Affiliation(s)
- Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | - Alison C. Barnes
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | - Kieren G. Hollingsworth
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | - Keaton M. Irvine
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | | | - Lucy Clark
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | - Tara Kelly
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, U.K
| | - Carmen Martin-Ruiz
- BioScreening Core Facility, Campus for Ageing and Vitality, Faculty of Medical Sciences, Newcastle University, U.K
| | - Davide Romeres
- Department of Endocrinology, University of Virginia, Charlottesville, VA, U.S.A
| | - Albert Koulman
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Cambridge, U.K
| | - Claire M. Meek
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Cambridge, U.K
- Wolfson Diabetes and Endocrine Centre, Cambridge Universities NHS Foundation Trust, Cambridge, U.K
| | - Benjamin Jenkins
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Cambridge, U.K
| | - Claudio Cobelli
- Department of Woman and Child's Health, University of Padova, Italy
| | - Rury R. Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| |
Collapse
|
12
|
Soria-Gondek A, Fernández-García P, González L, Reyes-Farias M, Murillo M, Valls A, Real N, Pellitero S, Tarascó J, Jenkins B, Galán M, Villarroya F, Koulman A, Corrales P, Vidal-Puig A, Cereijo R, Sánchez-Infantes D. Lipidome Profiling in Childhood Obesity Compared to Adults: A Pilot Study. Nutrients 2023; 15:3341. [PMID: 37571279 PMCID: PMC10421258 DOI: 10.3390/nu15153341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/13/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The objective is to assess the circulating lipidome of children with obesity before and after lifestyle intervention and to compare the data to the circulating lipidome of adults with obesity before and after bariatric surgery. Ten pediatric (PE) and thirty adult (AD) patients with obesity were prospectively recruited at a referral single center. The PE cohort received lifestyle recommendations. The AD cohort underwent bariatric surgery. Clinical parameters and lipidome were analyzed in serum before and after six months of metabolic intervention. The abundance of phosphatidylinositols in the PE cohort and phosphatidylcholines in the AD significantly increased, while O-phosphatidylserines in the PE cohort and diacyl/triacylglycerols in the AD decreased. Fifteen lipid species were coincident in both groups after lifestyle intervention and bariatric surgery. Five species of phosphatidylinositols, sphingomyelins, and cholesteryl esters were upregulated. Eight species of diacylglycerols, glycerophosphoglycerols, glycerophosphoethanolamines, and phosphatidylcholines were downregulated. Most matching species were regulated in the same direction except for two phosphatidylinositols: PI(O-36:2) and PI(O-34:0). A specific set of lipid species regulated after bariatric surgery in adult individuals was also modulated in children undergoing lifestyle intervention, suggesting they may constitute a core circulating lipid profile signature indicative of early development of obesity and improvement after clinical interventions regardless of individual age.
Collapse
Affiliation(s)
- Andrea Soria-Gondek
- Pediatric Surgery Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Pablo Fernández-García
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
| | - Lorena González
- Fundació Institut Germans Trias i Pujol, 08916 Barcelona, Spain; (L.G.); (M.R.-F.)
| | - Marjorie Reyes-Farias
- Fundació Institut Germans Trias i Pujol, 08916 Barcelona, Spain; (L.G.); (M.R.-F.)
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Marta Murillo
- Pediatric Endocrinology Unit, Pediatric Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (M.M.); (A.V.)
| | - Aina Valls
- Pediatric Endocrinology Unit, Pediatric Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain; (M.M.); (A.V.)
| | - Nativitat Real
- Pediatric Nurse, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Silvia Pellitero
- Endocrinology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Jordi Tarascó
- General Surgery Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1GG, UK; (B.J.); (A.K.)
| | - María Galán
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
| | - Francesc Villarroya
- Biochemistry and Molecular Biomedicine Department, Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1GG, UK; (B.J.); (A.K.)
| | - Patricia Corrales
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
| | - Antonio Vidal-Puig
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 1GG, UK;
| | - Rubén Cereijo
- Biochemistry and Molecular Biomedicine Department, Institut de Biomedicina de la Universitat de Barcelona (IBUB), 08028 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
- Institut de Recerca Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - David Sánchez-Infantes
- Department of Health Sciences, Campus Alcorcón, University Rey Juan Carlos (URJC), 28922 Madrid, Spain; (P.F.-G.); (M.G.); (P.C.)
- Fundació Institut Germans Trias i Pujol, 08916 Barcelona, Spain; (L.G.); (M.R.-F.)
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain
| |
Collapse
|
13
|
Horakova O, Sistilli G, Kalendova V, Bardova K, Mitrovic M, Cajka T, Irodenko I, Janovska P, Lackner K, Kopecky J, Rossmeisl M. Thermoneutral housing promotes hepatic steatosis in standard diet-fed C57BL/6N mice, with a less pronounced effect on NAFLD progression upon high-fat feeding. Front Endocrinol (Lausanne) 2023; 14:1205703. [PMID: 37501785 PMCID: PMC10369058 DOI: 10.3389/fendo.2023.1205703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) can progress to more severe stages, such as steatohepatitis and fibrosis. Thermoneutral housing together with high-fat diet promoted NAFLD progression in C57BL/6J mice. Due to possible differences in steatohepatitis development between different C57BL/6 substrains, we examined how thermoneutrality affects NAFLD progression in C57BL/6N mice. Methods Male mice were fed standard or high-fat diet for 24 weeks and housed under standard (22°C) or thermoneutral (30°C) conditions. Results High-fat feeding promoted weight gain and hepatic steatosis, but the effect of thermoneutral environment was not evident. Liver expression of inflammatory markers was increased, with a modest and inconsistent effect of thermoneutral housing; however, histological scores of inflammation and fibrosis were generally low (<1.0), regardless of ambient temperature. In standard diet-fed mice, thermoneutrality increased weight gain, adiposity, and hepatic steatosis, accompanied by elevated de novo lipogenesis and changes in liver metabolome characterized by complex decreases in phospholipids and metabolites involved in urea cycle and oxidative stress defense. Conclusion Thermoneutrality appears to promote NAFLD-associated phenotypes depending on the C57BL/6 substrain and/or the amount of dietary fat.
Collapse
Affiliation(s)
- Olga Horakova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Gabriella Sistilli
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Veronika Kalendova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Marko Mitrovic
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- First Faculty of Medicine, Charles University, Prague, Czechia
| | - Tomas Cajka
- Laboratory of Translational Metabolism, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Ilaria Irodenko
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Petra Janovska
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Karoline Lackner
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
14
|
Mocciaro G, Allison M, Jenkins B, Azzu V, Huang-Doran I, Herrera-Marcos LV, Hall Z, Murgia A, Susan D, Frontini M, Vidal-Puig A, Koulman A, Griffin JL, Vacca M. Non-alcoholic fatty liver disease is characterised by a reduced polyunsaturated fatty acid transport via free fatty acids and high-density lipoproteins (HDL). Mol Metab 2023; 73:101728. [PMID: 37084865 PMCID: PMC10176260 DOI: 10.1016/j.molmet.2023.101728] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/25/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) develops due to impaired hepatic lipid fluxes and is a risk factor for chronic liver disease and atherosclerosis. Lipidomic studies consistently reported characteristic hepatic/VLDL "lipid signatures" in NAFLD; whole plasma traits are more debated. Surprisingly, the HDL lipid composition by mass spectrometry has not been characterised across the NAFLD spectrum, despite HDL being a possible source of hepatic lipids delivered from peripheral tissues alongside free fatty acids (FFA). This study characterises the HDL lipidomic signature in NAFLD, and its correlation with metabolic and liver disease markers. METHODS We used liquid chromatography-mass spectrometry to determine the whole serum and HDL lipidomic profile in 89 biopsy-proven NAFLD patients and 20 sex and age-matched controls. RESULTS In the whole serum of NAFLD versus controls, we report a depletion in polyunsaturated (PUFA) phospholipids (PL) and FFA; with PUFA PL being also lower in HDL, and negatively correlated with BMI, insulin resistance, triglycerides, and hepatocyte ballooning. In the HDL of the NAFLD group we also describe higher saturated ceramides, which positively correlate with insulin resistance and transaminases. CONCLUSION NAFLD features lower serum lipid species containing polyunsaturated fatty acids; the most affected lipid fractions are FFA and (HDL) phospholipids; our data suggest a possible defect in the transfer of PUFA from peripheral tissues to the liver in NAFLD. Mechanistic studies are required to explore the biological implications of our findings addressing if HDL composition can influence liver metabolism and damage, thus contributing to NAFLD pathophysiology.
Collapse
Affiliation(s)
- Gabriele Mocciaro
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; Roger Williams Institute of Hepatology, Foundation for Liver Research, London, SE5 9NT, United Kingdom
| | - Michael Allison
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Benjamin Jenkins
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Vian Azzu
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom; Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Isabel Huang-Doran
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Luis Vicente Herrera-Marcos
- Department of Biochemistry and Molecular and Cellular Biology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
| | - Zoe Hall
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Antonio Murgia
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom
| | - Davies Susan
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Mattia Frontini
- Faculty of Health and Life Sciences, Clinical and Biomedical Sciences, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - Antonio Vidal-Puig
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Albert Koulman
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom.
| | - Julian L Griffin
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; The Rowett Institute, Foresterhill Campus, University of Aberdeen, Aberdeen, AB25 2ZD, United Kingdom.
| | - Michele Vacca
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; Roger Williams Institute of Hepatology, Foundation for Liver Research, London, SE5 9NT, United Kingdom; Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom; Aldo Moro University of Bari, Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", Bari, 70124, Italy.
| |
Collapse
|
15
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
16
|
Noureddin M, Abdelmalek MF. Current Treatment Options, Including Diet, Exercise, and Medications: The Impact on Histology. Clin Liver Dis 2023; 27:397-412. [PMID: 37024215 DOI: 10.1016/j.cld.2023.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Paralleling the rise in obesity and diabetes, nonalcoholic fatty liver disease (NAFLD) is now the most prevalent chronic liver disease worldwide. Nonalcoholic steatohepatitis (NASH), the progressive form of NAFLD, may progress to cirrhosis, hepatic decompensation, and hepatocellular carcinoma. Despite its public health treat, no approved pharmacotherapies for NAFLD/NASH currently exist. Although the armamentarium of therapies for NASH is limited, current treatment options include life-style modification and the use of medications to treat metabolic comorbidities. This review addresses current approaches to the treatment of NAFLD/NASH, including the impact of diet, exercise, and available pharmacotherapies on the histologic features of liver injury.
Collapse
Affiliation(s)
- Mazen Noureddin
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Houston Methodist Hospital, Houston, TX, USA; Houston Research Institute and Houston Liver Institute, Houston, TX, USA
| | - Manal F Abdelmalek
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
17
|
Rinella ME, Neuschwander-Tetri BA, Siddiqui MS, Abdelmalek MF, Caldwell S, Barb D, Kleiner DE, Loomba R. AASLD Practice Guidance on the clinical assessment and management of nonalcoholic fatty liver disease. Hepatology 2023; 77:1797-1835. [PMID: 36727674 PMCID: PMC10735173 DOI: 10.1097/hep.0000000000000323] [Citation(s) in RCA: 594] [Impact Index Per Article: 594.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 02/03/2023]
Affiliation(s)
- Mary E. Rinella
- University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| | | | | | | | - Stephen Caldwell
- School of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Diana Barb
- University of Florida College of Medicine, Gainesville, Florida, USA
| | | | - Rohit Loomba
- University of California, San Diego, San Diego, California, USA
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW De novo lipogenesis (DNL) is a metabolic process occurring mainly within the liver, in humans. Insulin is a primary signal for promoting DNL; thus, nutritional state is a key determinant for upregulation of the pathway. However, the effects of dietary macronutrient composition on hepatic DNL remain unclear. Nor is it clear if a nutrition-induced increase in DNL results in accumulation of intra-hepatic triglyceride (IHTG); a mechanism often proposed for pathological IHTG. Here, we review the latest evidence surrounding the nutritional regulation of hepatic DNL. RECENT FINDINGS The role of carbohydrate intake on hepatic DNL regulation has been well studied, with only limited data on the effects of fats and proteins. Overall, increasing carbohydrate intake typically results in an upregulation of DNL, with fructose being more lipogenic than glucose. For fat, it appears that an increased intake of n-3 polyunsaturated fatty acids downregulates DNL, whilst, in contrast, an increased dietary protein intake may upregulate DNL. SUMMARY Although DNL is upregulated with high-carbohydrate or mixed-macronutrient meal consumption, the effects of fat and protein remain unclear. Additionally, the effects of different phenotypes (including sex, age, ethnicity, and menopause status) in combination with different diets (enriched in different macronutrients) on hepatic DNL requires elucidation.
Collapse
Affiliation(s)
- Eloise Cross
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford
| | - David J Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
19
|
Herrera-Marcos LV, Martínez-Beamonte R, Arnal C, Barranquero C, Puente-Lanzarote JJ, Herrero-Continente T, Lou-Bonafonte JM, Gonzalo-Romeo G, Mocciaro G, Jenkins B, Surra JC, Rodríguez-Yoldi MJ, Burillo JC, Lasheras R, García-Gil A, Güemes A, Koulman A, Osada J. Dietary squalene supplementation decreases triglyceride species and modifies phospholipid lipidomic profile in the liver of a porcine model of non-alcoholic steatohepatitis. J Nutr Biochem 2023; 112:109207. [PMID: 36402249 DOI: 10.1016/j.jnutbio.2022.109207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/07/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
Abstract
Squalene is a key minor component of virgin olive oil, the main source of fat in the Mediterranean diet, and had shown to improve the liver metabolism in rabbits and mice. The present research was carried out to find out whether this effect was conserved in a porcine model of hepatic steatohepatitis and to search for the lipidomic changes involved. The current study revealed that a 0.5% squalene supplementation to a steatotic diet for a month led to hepatic accumulation of squalene and decreased triglyceride content as well as area of hepatic lipid droplets without influencing cholesterol content or fiber areas. However, ballooning score was increased and associated with the hepatic squalene content. Of forty hepatic transcripts related to lipid metabolism and hepatic steatosis, only citrate synthase and a non-coding RNA showed decreased expressions. The hepatic lipidome, assessed by liquid chromatography-mass spectrometry in a platform able to analyze 467 lipids, revealed that squalene supplementation increased ceramide, Cer(36:2), and phosphatidylcholine (PC[32:0], PC[33:0] and PC[34:0]) species and decreased cardiolipin, CL(69:5), and triglyceride (TG[54:2], TG[55:0] and TG[55:2]) species. Plasma levels of interleukin 12p40 increased in pigs receiving the squalene diet. The latter also modified plasma lipidome by increasing TG(58:12) and decreasing non-esterified fatty acid (FA 14:0, FA 16:1 and FA 18:0) species without changes in total NEFA levels. Together this shows that squalene-induced changes in hepatic and plasma lipidomic profiles, non-coding RNA and anti-inflammatory interleukin are suggestive of an alleviation of the disease despite the increase in the ballooning score.
Collapse
Affiliation(s)
- Luis V Herrera-Marcos
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain
| | - Roberto Martínez-Beamonte
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Carmen Arnal
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Patología Animal, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Cristina Barranquero
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Juan J Puente-Lanzarote
- Servicio de Bioquímica Clínica. Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Tania Herrero-Continente
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - José M Lou-Bonafonte
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Gonzalo Gonzalo-Romeo
- Servicio General de Apoyo a la Investigación. División de Experimentación Animal, Universidad de Zaragoza, Zaragoza, Spain
| | - Gabriele Mocciaro
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Joaquín C Surra
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Producción Animal y Ciencia de los Alimentos, Escuela Politécnica Superior de Huesca, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Huesca, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - María J Rodríguez-Yoldi
- Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; Departamento de Farmacología, Fisiología, Medicina Legal y Forense, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - Juan Carlos Burillo
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Zaragoza, Spain
| | - Roberto Lasheras
- Laboratorio Agroambiental, Servicio de Seguridad Agroalimentaria de la Dirección General de Alimentación y Fomento Agroalimentario, Zaragoza, Spain
| | - Agustín García-Gil
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Antonio Güemes
- Departamento de Cirugía, Facultad de Medicina, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón-Universidad de Zaragoza, Zaragoza, Spain; Instituto Agroalimentario de Aragón, CITA-Universidad de Zaragoza, Zaragoza, Spain; CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
20
|
Hu H, Sun N, Du H, He Y, Pan K, Liu X, Lu X, Wei J, Liao M, Duan C. Mouse promyelocytic leukemia zinc finger protein (PLZF) regulates hepatic lipid and glucose homeostasis dependent on SIRT1. Front Pharmacol 2022; 13:1039726. [PMID: 36438786 PMCID: PMC9684722 DOI: 10.3389/fphar.2022.1039726] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022] Open
Abstract
Previous studies have demonstrated that promyelocytic leukemia zinc finger protein (PLZF) promotes the expression of gluconeogenic genes and hepatic glucose output, which leads to hyperglycemia. However, the role played by PLZF in regulating lipid metabolism is not known. In this study, we aimed to examine the function of PLZF in regulating hepatic lipid and glucose homeostasis and the underlying mechanisms. The expression of PLZF was determined in different mouse models with regard to non-alcoholic fatty liver disease (NAFLD). In the next step, adenoviruses that express PLZF (Ad-PLZF) or PLZF-specific shRNA (Ad-shPLZF) were utilized to alter PLZF expression in mouse livers and in primary hepatocytes. For the phenotype of the fatty liver, histologic and biochemical analyses of hepatic triglyceride (TG), serum TG and cholesterol levels were carried out. The underlying molecular mechanism for the regulation of lipid metabolism by PLZF was further explored using luciferase reporter gene assay and ChIP analysis. The results demonstrated that PLZF expression was upregulated in livers derived from ob/ob, db/db and diet-induced obesity (DIO) mice. Liver PLZF-overexpressing C57BL/6J mice showed fatty liver phenotype, liver inflammation, impaired glucose tolerance and insulin sensitivity. On the other hand, hepatic PLZF knockdown in db/db and DIO mice alleviated hepatic steatosis. Of note, we found that PLZF activates SREBP-1c gene transcription through binding directly to the promoter fragment of this gene, which would induce a repressor-to-activator conversion depending on its interaction with SIRT1 in the role played by PLZF in the transcription process through deacetylation. Thus, PLZF is identified as an essential regulator of hepatic lipid and glucose metabolism, where the modulation of its liver expression could open up a therapeutic path for treating NAFLD.
Collapse
Affiliation(s)
- Huiling Hu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Nannan Sun
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haiyan Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Yuqing He
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kunyi Pan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiuli Liu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxia Lu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Wei
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jie Wei, ; Mianmian Liao, ; Chaohui Duan,
| | - Mianmian Liao
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- *Correspondence: Jie Wei, ; Mianmian Liao, ; Chaohui Duan,
| | - Chaohui Duan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Jie Wei, ; Mianmian Liao, ; Chaohui Duan,
| |
Collapse
|
21
|
Li X, Li M, Xu L, Zeng X, Zhang T, Yang H, Li H, Zhou Z, Wang Y, Zhang C, Zhu Y, Huang Y, Zhang Z, Yang W. Associations between low-carbohydrate and low-fat diets and hepatic steatosis. Obesity (Silver Spring) 2022; 30:2317-2328. [PMID: 36058841 DOI: 10.1002/oby.23551] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/26/2022] [Accepted: 07/19/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE This study assessed the cross-sectional associations of low-carbohydrate diets (LCDs) and low-fat diets (LFDs) with hepatic steatosis in the National Health and Nutrition Examination Survey. METHODS Diet was measured using the 24-hour recalls. Hepatic steatosis was defined by vibration-controlled transient elastography. The odds ratios (OR) and 95% confidence intervals (CI) were estimated using logistic regression. Substitution analysis was performed using the leave-one-out model. RESULTS Participants with higher adherence scores (comparing extreme tertiles) for an overall (OR = 0.76, 95% CI: 0.61-0.96, ptrend = 0.049) or a healthful LCD (OR = 0.61, 95% CI: 0.43-0.87, ptrend < 0.001) exhibited lower odds of steatosis. Replacing 5% of the energy from carbohydrates with total fat and protein (OR = 0.91, 95% CI: 0.83-0.99) or unsaturated fat and plant protein (OR = 0.89, 95% CI: 0.84-0.94) was associated with lower steatosis prevalence. High overall (OR = 1.65, 95% CI: 1.13-2.40, ptrend = 0.006) or unhealthful (OR = 1.41, 95% CI: 1.10-1.80, ptrend < 0.001) LFD scores were associated with increased likelihood of steatosis. CONCLUSIONS These findings suggest that the associations between LCDs and LFDs and steatosis may depend on the quality and food sources of the macronutrients.
Collapse
Affiliation(s)
- Xiude Li
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Meiling Li
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Linsheng Xu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xueke Zeng
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Tengfei Zhang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Hu Yang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Haowei Li
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhihao Zhou
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Yu Wang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Chenghao Zhang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Yu Zhu
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Yong Huang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhuang Zhang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
| | - Wanshui Yang
- Department of Nutrition, School of Public Health, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Hefei, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics/Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| |
Collapse
|
22
|
Biolo G, Vinci P, Mangogna A, Landolfo M, Schincariol P, Fiotti N, Mearelli F, Di Girolamo FG. Mechanism of action and therapeutic use of bempedoic acid in atherosclerosis and metabolic syndrome. Front Cardiovasc Med 2022; 9:1028355. [PMID: 36386319 PMCID: PMC9650075 DOI: 10.3389/fcvm.2022.1028355] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 08/13/2023] Open
Abstract
Bempedoic acid is a new cholesterol-lowering drug, which has recently received US FDA and EMA approval. This drug targets lipid and glucose metabolism as well as inflammation via downregulation of ATP-citrate lyase and upregulation of AMP-activated protein kinase (AMPK). The primary effect is the reduction of cholesterol synthesis in the liver and its administration is generally not associated to unwanted muscle effects. Suppression of hepatic fatty acid synthesis leads to decreased triglycerides and, possibly, improved non-alcoholic fatty liver disease. Bempedoic acid may decrease gluconeogenesis leading to improved insulin sensitivity, glucose metabolism, and metabolic syndrome. The anti-inflammatory action of bempedoic acid is mainly achieved via activation of AMPK pathway in the immune cells, leading to decreased plasma levels of C-reactive protein. Effects of bempedoic acid on atherosclerotic cardiovascular disease, type 2 diabetes and chronic liver disease have been assessed in randomized clinical trials but require further confirmation. Safety clinical trials in phase III indicate that bempedoic acid administration is generally well-tolerated in combination with statins, ezetimibe, or proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors to achieve low-density lipoprotein cholesterol targets. The aim of this narrative review on bempedoic acid is to explore the underlying mechanisms of action and potential clinical targets, present existing evidence from clinical trials, and describe practical management of patients.
Collapse
Affiliation(s)
- Gianni Biolo
- Medical Clinic, Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Pierandrea Vinci
- Medical Clinic, Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Matteo Landolfo
- Medical Clinic, Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Paolo Schincariol
- Hospital Pharmacy, Cattinara Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Nicola Fiotti
- Medical Clinic, Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Filippo Mearelli
- Medical Clinic, Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Filippo Giorgio Di Girolamo
- Medical Clinic, Department of Medical, Surgical and Health Sciences, Cattinara Hospital, University of Trieste, Trieste, Italy
- Hospital Pharmacy, Cattinara Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| |
Collapse
|
23
|
Niu Z, Wu Q, Luo Y, Wang D, Zheng H, Wu Y, Yang X, Zeng R, Sun L, Lin X. Plasma Lipidomic Subclasses and Risk of Hypertension in Middle-Aged and Elderly Chinese. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:283-294. [PMID: 36939788 PMCID: PMC9590468 DOI: 10.1007/s43657-022-00057-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 10/18/2022]
Abstract
While disrupted lipid metabolism is a well-established risk factor for hypertension in animal models, the links between various lipidomic signatures and hypertension in human studies remain unclear. We aimed to examine associations between plasma lipidomic profiles and prevalence of hypertension among 2248 community-living Chinese aged 50-70 years. Hypertension was defined according to 2020 International Society of Hypertension global hypertension practice guidelines and 2018 Chinese guidelines. In total, 728 plasma lipidomic species were profiled using high-coverage targeted lipidomics. After multivariate adjustment, including lifestyle, body mass index, blood lipids, and sodium intake, 110 metabolites from nine lipidomic subclasses showed significant associations with hypertension, among which phosphatidylethanolamines (PEs) had the strongest association. Eleven lipidomic signals for hypertension risk were further identified from the nine subclasses, including PE(18:0/18:2) (OR per SD, 1.49; 95% confidence intervals, 1.30-1.69), phosphatidylcholine (PC) (18:0/18:2) (1.27; 1.13-1.43), phosphatidylserine (18:0/18:0) (1.24; 1.09-1.41), lysophosphatidylinositol (18:1) (1.17; 1.06-1.29), triacylglycerol (52:5) (1.38; 1.18-1.61), diacylglycerol (16:0/18:2) (1.42; 1.19-1.69), dihydroceramide (24:0) (1.25; 1.09-1.43), hydroxyl-sphingomyelins (SM[2OH])C34:1 (1.19; 1.07-1.33), lysophosphatidylcholine (20:1) (0.86; 0.78-0.95), SM(OH)C38:1 (0.87; 0.79-0.96), and PC (18:2/20:1) (0.84; 0.75-0.94). Principal component analysis also showed that a factor mainly containing specific PEs was positively associated with hypertension (1.20; 1.09-1.33). Collectively, our study revealed that disturbances in multiple circulating lipidomic subclasses and signatures, especially PEs, were significantly associated with the prevalence of hypertension in middle-aged and elderly Chinese. Future studies are warranted to confirm our findings and determine the mechanisms underlying these associations. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-022-00057-y.
Collapse
Affiliation(s)
- Zhenhua Niu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - Qingqing Wu
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yaogan Luo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - Di Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - He Zheng
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - Yanpu Wu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - Xiaowei Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - Rong Zeng
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031 China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024 China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024 China
| | - Liang Sun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-yang Rd., Shanghai, 200031 China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024 China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024 China
| |
Collapse
|
24
|
Furse S, Kusinski LC, Ray A, Glenn-Sansum C, Williams HEL, Koulman A, Meek CL. Relative Abundance of Lipid Metabolites in Spermatozoa across Three Compartments. Int J Mol Sci 2022; 23:ijms231911655. [PMID: 36232961 PMCID: PMC9569887 DOI: 10.3390/ijms231911655] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022] Open
Abstract
Male fertility, as manifest by the quantity and progressive motility of spermatozoa, is negatively impacted by obesity, dyslipidaemia and metabolic disease. However, the relative distribution of lipids in spermatozoa and the two compartments which supply lipids for spermatogenesis (seminal fluid and blood serum) has not been studied. We hypothesised that altered availability of lipids in blood serum and seminal fluid may affect the lipid composition and progressive motility of sperm. 60 men of age 35 years (median (range 20-45) and BMI 30.4 kg/m2 (24-36.5) under preliminary investigation for subfertility were recruited at an NHS clinic. Men provided samples of serum and semen, subject to strict acceptance criteria, for analysis of spermatozoa count and motility. Blood serum (n = 60), spermatozoa (n = 26) and seminal fluid (n = 60) were frozen for batch lipidomics analysis. Spermatozoa and seminal fluid had comparable lipid composition but showed marked differences with the serum lipidome. Spermatozoa demonstrated high abundance of ceramides, very-long-chain fatty acids (C20-22), and certain phospholipids (sphingomyelins, plasmalogens, phosphatidylethanolamines) with low abundance of phosphatidylcholines, cholesterol and triglycerides. Men with spermatozoa of low progressive motility had evidence of fewer concentration gradients for many lipid species between blood serum and spermatozoa compartments. Spermatozoa are abundant in multiple lipid species which are likely to contribute to key cellular functions. Lipid metabolism shows reduced regulation between compartments in men with spermatozoa with reduced progressive motility.
Collapse
Affiliation(s)
- Samuel Furse
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Laura C. Kusinski
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Alison Ray
- Department of Clinical Chemistry and Immunology, Peterborough City Hospital, North West Anglia NHS Foundation Trust, Bretton Gate, Peterborough PE3 9GZ, UK
| | - Coralie Glenn-Sansum
- R&D Department, Peterborough City Hospital, North West Anglia NHS Foundation Trust, Bretton Gate, Peterborough PE3 9GZ, UK
| | - Huw E. L. Williams
- Centre for Biomolecular Sciences, School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | - Albert Koulman
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Claire L. Meek
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Box 289, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
- Department of Clinical Chemistry and Immunology, Peterborough City Hospital, North West Anglia NHS Foundation Trust, Bretton Gate, Peterborough PE3 9GZ, UK
- Department of Clinical Biochemistry, Cambridge Universities NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
- Correspondence: ; Tel.: +44-(0)1223-767176
| |
Collapse
|
25
|
Fan X, Liu Y, Zhang Z, Zhang Z, Peng J, Gao Y, Zheng L, Chen J, Du J, Yan S, Zhou X, Shi X, Zhang D. Bta06987, Encoding a Peptide of the AKH/RPCH Family: A Role of Energy Mobilization in Bemisia tabaci. INSECTS 2022; 13:834. [PMID: 36135535 PMCID: PMC9502992 DOI: 10.3390/insects13090834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 06/16/2023]
Abstract
A neuropeptide precursor encoded by Bta06987 associates with AKH neuropeptide. In the AKH/RPCH family, these members have been demonstrated to participate in energy mobilization in many insects. In our research, the Bta06987 gene from Bemisia tabaci was cloned, and the amino acid sequence analysis was performed. During the starvation of B. tabaci, the mRNA level of Bta06987 showed a significant elevation. We investigated the functions of Bta06987 in B. tabaci using RNA interference (RNAi), and the adult females of B. tabaci after being fed with dsBta06987 showed a higher glycogen and triglyceride levels and lower trehalose content than the control. Furthermore, in the electrical penetration graph (EPG) experiment, B. tabaci showed changes in feeding behavior after feeding with dsBta06987, such as the reduction in parameters of E waveform percentage and total feeding time. Our findings might be helpful in developing strategies to control pest and plant virus transmission.
Collapse
Affiliation(s)
- Xiaofan Fan
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Yong Liu
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Zhuo Zhang
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Zhanhong Zhang
- Institute of Vegetable Crops, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Jing Peng
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Yang Gao
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Limin Zheng
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Jianbin Chen
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Jiao Du
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Shuo Yan
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Xuguo Zhou
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA
| | - Xiaobin Shi
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Deyong Zhang
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China
- Institute of Plant Protection, Hunan Academy of Agricultural Sciences, Changsha 410125, China
| |
Collapse
|
26
|
Kim JE, Kim E, Lee JW. TM4SF5-Mediated Regulation of Hepatocyte Transporters during Metabolic Liver Diseases. Int J Mol Sci 2022; 23:ijms23158387. [PMID: 35955521 PMCID: PMC9369364 DOI: 10.3390/ijms23158387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is found in up to 30% of the world’s population and can lead to hepatocellular carcinoma (HCC), which has a poor 5-year relative survival rate of less than 40%. Clinical therapeutic strategies are not very successful. The co-occurrence of metabolic disorders and inflammatory environments during the development of steatohepatitis thus needs to be more specifically diagnosed and treated to prevent fatal HCC development. To improve diagnostic and therapeutic strategies, the identification of molecules and/or pathways responsible for the initiation and progression of chronic liver disease has been explored in many studies, but further study is still required. Transmembrane 4 L six family member 5 (TM4SF5) has been observed to play roles in the regulation of metabolic functions and activities in hepatocytes using in vitro cell and in vivo animal models without or with TM4SF5 expression in addition to clinical liver tissue samples. TM4SF5 is present on the membranes of different organelles or vesicles and cooperates with transporters for fatty acids, amino acids, and monocarbohydrates, thus regulating nutrient uptake into hepatocytes and metabolism and leading to phenotypes of chronic liver diseases. In addition, TM4SF5 can remodel the immune environment by interacting with immune cells during TM4SF5-mediated chronic liver diseases. Because TM4SF5 may act as an NAFLD biomarker, this review summarizes crosstalk between TM4SF5 and nutrient transporters in hepatocytes, which is related to chronic liver diseases.
Collapse
|
27
|
Leslie J, Geh D, Elsharkawy AM, Mann DA, Vacca M. Metabolic dysfunction and cancer in HCV: Shared pathways and mutual interactions. J Hepatol 2022; 77:219-236. [PMID: 35157957 DOI: 10.1016/j.jhep.2022.01.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 12/16/2022]
Abstract
HCV hijacks many host metabolic processes in an effort to aid viral replication. The resulting hepatic metabolic dysfunction underpins many of the hepatic and extrahepatic manifestations of chronic hepatitis C (CHC). However, the natural history of CHC is also substantially influenced by the host metabolic status: obesity, insulin resistance and hepatic steatosis are major determinants of CHC progression toward hepatocellular carcinoma (HCC). Direct-acting antivirals (DAAs) have transformed the treatment and natural history of CHC. While DAA therapy effectively eradicates the virus, the long-lasting overlapping metabolic disease can persist, especially in the presence of obesity, increasing the risk of liver disease progression. This review covers the mechanisms by which HCV tunes hepatic and systemic metabolism, highlighting how systemic metabolic disturbance, lipotoxicity and chronic inflammation favour disease progression and a precancerous niche. We also highlight the therapeutic implications of sustained metabolic dysfunction following sustained virologic response as well as considerations for patients who develop HCC on the background of metabolic dysfunction.
Collapse
Affiliation(s)
- Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel Geh
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ahmed M Elsharkawy
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Queen Elizabeth Medical Centre, Birmingham, B15 2TH UK; National Institute for Health Research, Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey.
| | - Michele Vacca
- Interdisciplinary Department of Medicine, Università degli Studi di Bari "Aldo Moro", Bari, Italy.
| |
Collapse
|
28
|
Mocciaro G, D’Amore S, Jenkins B, Kay R, Murgia A, Herrera-Marcos LV, Neun S, Sowton AP, Hall Z, Palma-Duran SA, Palasciano G, Reimann F, Murray A, Suppressa P, Sabbà C, Moschetta A, Koulman A, Griffin JL, Vacca M. Lipidomic Approaches to Study HDL Metabolism in Patients with Central Obesity Diagnosed with Metabolic Syndrome. Int J Mol Sci 2022; 23:6786. [PMID: 35743227 PMCID: PMC9223701 DOI: 10.3390/ijms23126786] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
The metabolic syndrome (MetS) is a cluster of cardiovascular risk factors characterised by central obesity, atherogenic dyslipidaemia, and changes in the circulating lipidome; the underlying mechanisms that lead to this lipid remodelling have only been partially elucidated. This study used an integrated "omics" approach (untargeted whole serum lipidomics, targeted proteomics, and lipoprotein lipidomics) to study lipoprotein remodelling and HDL composition in subjects with central obesity diagnosed with MetS (vs. controls). Compared with healthy subjects, MetS patients showed higher free fatty acids, diglycerides, phosphatidylcholines, and triglycerides, particularly those enriched in products of de novo lipogenesis. On the other hand, the "lysophosphatidylcholines to phosphatidylcholines" and "cholesteryl ester to free cholesterol" ratios were reduced, pointing to a lower activity of lecithin cholesterol acyltransferase (LCAT) in MetS; LCAT activity (directly measured and predicted by lipidomic ratios) was positively correlated with high-density lipoprotein cholesterol (HDL-C) and negatively correlated with body mass index (BMI) and insulin resistance. Moreover, many phosphatidylcholines and sphingomyelins were significantly lower in the HDL of MetS patients and strongly correlated with BMI and clinical metabolic parameters. These results suggest that MetS is associated with an impairment of phospholipid metabolism in HDL, partially led by LCAT, and associated with obesity and underlying insulin resistance. This study proposes a candidate strategy to use integrated "omics" approaches to gain mechanistic insights into lipoprotein remodelling, thus deepening the knowledge regarding the molecular basis of the association between MetS and atherosclerosis.
Collapse
Affiliation(s)
- Gabriele Mocciaro
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK; (G.M.); (A.M.); (S.N.); (Z.H.)
- Department of Interdisciplinary Medicine, Clinica Medica “C. Frugoni”, Aldo Moro University of Bari, 70124 Bari, Italy; (P.S.); (C.S.); (A.M.)
- Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
| | - Simona D’Amore
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK;
- Clinica Medica “A. Murri”, “Aldo Moro” University of Bari, 70124 Bari, Italy;
| | - Benjamin Jenkins
- Welcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK; (B.J.); (R.K.); (F.R.); (A.K.)
| | - Richard Kay
- Welcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK; (B.J.); (R.K.); (F.R.); (A.K.)
| | - Antonio Murgia
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK; (G.M.); (A.M.); (S.N.); (Z.H.)
| | - Luis Vicente Herrera-Marcos
- Department of Biochemistry and Molecular and Cellular Biology, Veterinary Faculty, University of Zaragoza, 50013 Zaragoza, Spain;
| | - Stefanie Neun
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK; (G.M.); (A.M.); (S.N.); (Z.H.)
| | - Alice P. Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.P.S.); (A.M.)
| | - Zoe Hall
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK; (G.M.); (A.M.); (S.N.); (Z.H.)
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK;
| | - Susana Alejandra Palma-Duran
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK;
| | - Giuseppe Palasciano
- Clinica Medica “A. Murri”, “Aldo Moro” University of Bari, 70124 Bari, Italy;
| | - Frank Reimann
- Welcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK; (B.J.); (R.K.); (F.R.); (A.K.)
| | - Andrew Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.P.S.); (A.M.)
| | - Patrizia Suppressa
- Department of Interdisciplinary Medicine, Clinica Medica “C. Frugoni”, Aldo Moro University of Bari, 70124 Bari, Italy; (P.S.); (C.S.); (A.M.)
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, Clinica Medica “C. Frugoni”, Aldo Moro University of Bari, 70124 Bari, Italy; (P.S.); (C.S.); (A.M.)
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, Clinica Medica “C. Frugoni”, Aldo Moro University of Bari, 70124 Bari, Italy; (P.S.); (C.S.); (A.M.)
| | - Albert Koulman
- Welcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK; (B.J.); (R.K.); (F.R.); (A.K.)
| | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK; (G.M.); (A.M.); (S.N.); (Z.H.)
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK;
- Rowlett Institute, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Michele Vacca
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1GA, UK; (G.M.); (A.M.); (S.N.); (Z.H.)
- Department of Interdisciplinary Medicine, Clinica Medica “C. Frugoni”, Aldo Moro University of Bari, 70124 Bari, Italy; (P.S.); (C.S.); (A.M.)
- Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK
- Welcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 0QQ, UK; (B.J.); (R.K.); (F.R.); (A.K.)
| |
Collapse
|
29
|
Inhibition of ATP-citrate lyase improves NASH, liver fibrosis, and dyslipidemia. Cell Metab 2022; 34:919-936.e8. [PMID: 35675800 DOI: 10.1016/j.cmet.2022.05.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/25/2022] [Accepted: 05/16/2022] [Indexed: 01/04/2023]
Abstract
Elevated liver de novo lipogenesis contributes to non-alcoholic steatohepatitis (NASH) and can be inhibited by targeting acetyl-CoA carboxylase (ACC). However, hypertriglyceridemia limits the use of pharmacological ACC inhibitors as a monotherapy. ATP-citrate lyase (ACLY) generates acetyl-CoA and oxaloacetate from citrate, but whether inhibition is effective for treating NASH is unknown. Here, we characterize a new mouse model that replicates many of the pathological and molecular drivers of NASH and find that genetically inhibiting ACLY in hepatocytes reduces liver malonyl-CoA, oxaloacetate, steatosis, and ballooning as well as blood glucose, triglycerides, and cholesterol. Pharmacological inhibition of ACLY mirrors genetic inhibition but has additional positive effects on hepatic stellate cells, liver inflammation, and fibrosis. Mendelian randomization of human variants that mimic reductions in ACLY also associate with lower circulating triglycerides and biomarkers of NASH. These data indicate that inhibiting liver ACLY may be an effective approach for treatment of NASH and dyslipidemia.
Collapse
|
30
|
Comparison of the Lipidomic Signature of Fatty Liver in Children and Adults: A Cross-Sectional Study. J Pediatr Gastroenterol Nutr 2022; 74:734-741. [PMID: 35185113 PMCID: PMC7613028 DOI: 10.1097/mpg.0000000000003418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is an increasingly common condition in children characterised by insulin resistance and altered lipid metabolism. Affected patients are at increased risk of cardiovascular disease (CVD) and children with NAFLD are likely to be at risk of premature cardiac events. Evaluation of the plasma lipid profile of children with NAFLD offers the opportunity to investigate these perturbations and understand how closely they mimic the changes seen in adults with cardiometabolic disease. METHODS We performed untargeted liquid chromatography-mass spectrometry (LC-MS) plasma lipidomics on 287 children: 19 lean controls, 146 from an obese cohort, and 122 NAFLD cases who had undergone liver biopsy. Associations between lipid species and liver histology were assessed using regression adjusted for age and sex. Results were then replicated using data from 9500 adults with metabolic phenotyping. RESULTS More severe paediatric NAFLD was associated with lower levels of long chain, polyunsaturated phosphatidylcholines (pC) and triglycerides (TG). Similar trends in pC and TG chain length and saturation were seen in adults with hepatic steatosis; however, many of the specific lipids associated with NAFLD differed between children and adults. Five lipids replicated in adults (including PC(36:4)) have been directly linked to death and cardiometabolic disease, as well as indirectly via genetic variants. CONCLUSION These findings suggest that, whilst similar pathways of lipid metabolism are perturbed in paediatric NAFLD as in cardiometabolic disease in adults, the specific lipid signature in children is different.
Collapse
|
31
|
Abstract
Much evidence for diabetes mellitus being associated with dysregulated lipid metabolism has been accrued from studies using blood plasma. However, the systemic dysregulation these results point to is not understood. This study used Lipid Traffic Analysis on data from a mouse model of diabetes to test the hypothesis that the systemic control of lipid metabolism differed in a model of diabetes. This provided eidence for changes in the systemic control of both triglyceride and phospholipid metabolism that were not attributable to dietary intake. This supports the conclusion that diabetes is a systemic condition associated with dysregulated lipid metabolism through several pathways.
Collapse
Affiliation(s)
- Samuel Furse
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, Cambridge, CB2 0QQ, UK.
- Metabolic Disease Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, Cambridge, CB2 0QQ, UK.
- Biological Chemistry Group, Jodrell Laboratory, Royal Botanic Gardens Kew, Richmond, TW9 3SD, UK.
| |
Collapse
|
32
|
Liang H, Yan J, Song K. Comprehensive lipidomic analysis reveals regulation of glyceride metabolism in rat visceral adipose tissue by high-altitude chronic hypoxia. PLoS One 2022; 17:e0267513. [PMID: 35522648 PMCID: PMC9075645 DOI: 10.1371/journal.pone.0267513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/10/2022] [Indexed: 11/18/2022] Open
Abstract
Adipose tissue plays a central role in energy substrate homeostasis and is a key regulator of lipid flow throughout these processes. As hypoxia affects lipid metabolism in adipose tissue, we aimed to investigate the effects of high-altitude chronic hypoxia on lipid metabolism in the adipose tissue of rats using a lipidomic analysis approach. Visceral adipose tissues from rats housed in a high-altitude hypoxia environment representing 4,300 m with 14.07% oxygen (hypoxia group) and from rats housed in a low-altitude normoxia environment representing 41 m with 20.95% oxygen (normoxia group) for 8 weeks were analyzed using an ultra-performance liquid chromatography-Orbitrap mass spectrometry system. After 8 weeks, the body weight and visceral adipose tissue weight of the hypoxia group were significantly decreased compared to those of the normoxia group (p < 0.05). The area and diameter of visceral adipose cells in the hypoxia group were significantly smaller than those of visceral adipose cells in the normoxia group (p < 0.05). The results of lipidomic analysis showed a total of 21 lipid classes and 819 lipid species. The total lipid concentration of the hypoxia group was lower than that in the normoxia group (p < 0.05). Concentrations of diacylglycerols and triacylglycerols in the hypoxia group were significantly lower than those in the normoxia group (p < 0.05). Using univariate and multivariate analyses, we identified 74 lipids that were significantly altered between the normoxia and hypoxia groups. These results demonstrate that high-altitude chronic hypoxia changes the metabolism of visceral adipose glycerides, which may potentially modulate other metabolic processes.
Collapse
Affiliation(s)
- Hong Liang
- Department of Basic Medical Sciences, Medical College, Qinghai University, Xining, PR, China
| | - Jun Yan
- Cardiovascular Medicine Department, Xuzhou Medical University affiliated Hospital, Xuzhou, PR China
| | - Kang Song
- Endocrinology Department, Qinghai Provincial People’s Hospital, Xining, PR, China
- Qinghai University affiliated Provincial People’s Hospital, Xining, PR, China
- * E-mail:
| |
Collapse
|
33
|
Li P, Hu S, Zhu Y, Sun T, Huang Y, Xu Z, Liu H, Luo C, Zhou S, Tan A, Liu L. Associations of Plasma Fatty Acid Patterns During Pregnancy With Gestational Diabetes Mellitus. Front Nutr 2022; 9:836115. [PMID: 35600822 PMCID: PMC9121815 DOI: 10.3389/fnut.2022.836115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/15/2022] [Indexed: 11/21/2022] Open
Abstract
Background Limited studies have explored the difference of fatty acid profile between women with and without gestational diabetes mellitus (GDM), and the results were inconsistent. Individual fatty acids tend to be interrelated because of the shared food sources and metabolic pathways. Thus, whether fatty acid patters during pregnancy were related to GDM odds needs further exploration. Objective To identify plasma fatty acid patters during pregnancy and their associations with odds of GDM. Methods A hospital-based case-control study including 217 GDM cases and 217 matched controls was carried out in urban Wuhan, China from August 2012 to April 2015. All the participants were enrolled at the time of GDM screening and provided fasting blood samples with informed consent. We measured plasma concentrations of fatty acids by gas chromatography-mass spectrometry, and derived potential fatty acid patterns (FAPs) through principal components analysis. Conditional logistic regression and restricted cubic spline model were used to evaluate the associations between individual fatty acids or FAPs and odds of GDM. Results Twenty individual fatty acids with relative concentrations ≥0.05% were included in the analyses. Compared with control group, GDM group had significantly higher concentrations of total fatty acids, 24:1n-9, and relatively lower levels of 14:0, 15:0, 17:0, 18:0, 24:0, 16:1n-7, 20:1n-9,18:3n-6, 20:2n-6, 18:3n-3, 20:3n-3, 22:5n-3. Two novel patterns of fatty acids were identified to be associated with lower odds of GDM: (1) relatively higher odd-chain fatty acids, 14:0, 18:0, 18:3n-3, 20:2n-6, 20:3n-6 and lower 24:1n-9 and 18:2n-6 [adjusted odds ratio (OR) (95% confidence interval) (CI) for quartiles 4 vs. 1: 0.42 (0.23-0.76), P-trend = 0.002], (2) relatively higher n-3 polyunsaturated fatty acids, 24:0, 18:3n-6 and lower 16:0 and 20:4n-6 [adjusted OR (95% CI) for quartiles 4 vs. 1: 0.48 (0.26-0.90), P-trend = 0.018]. Conclusion Our findings suggested that two novel FAPs were inversely associated with GDM odds. The combination of circulating fatty acids could be a more significant marker of GDM development than individual fatty acids or their subgroups.
Collapse
Affiliation(s)
- Peiyun Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Nutrition, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan Hu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yalun Zhu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Taoping Sun
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Huang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihui Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjie Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Luo
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiqiong Zhou
- Department of Clinical Nutrition, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aijun Tan
- Zhuhai Center for Disease Control and Prevention, Zhuhai, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Furse S, Virtue S, Snowden SG, Vidal-Puig A, Stevenson PC, Chiarugi D, Koulman A. Dietary PUFAs drive diverse system-level changes in lipid metabolism. Mol Metab 2022; 59:101457. [PMID: 35150907 PMCID: PMC8894240 DOI: 10.1016/j.molmet.2022.101457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Polyunsaturated fatty acid (PUFA) supplements have been trialled as a treatment for a number of conditions and produced a variety of results. This variety is ascribed to the supplements, that often comprise a mixture of fatty acids, and to different effects in different organs. In this study, we tested the hypothesis that the supplementation of individual PUFAs has system-level effects that are dependent on the molecular structure of the PUFA. METHODS We undertook a network analysis using Lipid Traffic Analysis to identify both local and system-level changes in lipid metabolism using publicly available lipidomics data from a mouse model of supplementation with FA(20:4n-6), FA(20:5n-3), and FA(22:6n-3); arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid, respectively. Lipid Traffic Analysis is a new computational/bioinformatics tool that uses the spatial distribution of lipids to pinpoint changes or differences in control of metabolism, thereby suggesting mechanistic reasons for differences in observed lipid metabolism. RESULTS There was strong evidence for changes to lipid metabolism driven by and dependent on the structure of the supplemented PUFA. Phosphatidylcholine and triglycerides showed a change in the variety more than the total number of variables, whereas phosphatidylethanolamine and phosphatidylinositol showed considerable change in both which variables and the number of them, in a highly PUFA-dependent manner. There was also evidence for changes to the endogenous biosynthesis of fatty acids and to both the elongation and desaturation of fatty acids. CONCLUSIONS These results show that the full biological impact of PUFA supplementation is far wider than any single-organ effect and implies that supplementation and dosing with PUFAs require a system-level assessment.
Collapse
Affiliation(s)
- Samuel Furse
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK; Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK; Royal Botanic Gardens, Kew, Kew Green, Richmond, Surrey, TW9 3AE, UK.
| | - Samuel Virtue
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK
| | - Stuart G Snowden
- Biology Department, Royal Holloway College, University of London, UK; Centro de Investigacion Principe Felipe, 46012 Valencia, Spain
| | - Antonio Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK
| | - Philip C Stevenson
- Royal Botanic Gardens, Kew, Kew Green, Richmond, Surrey, TW9 3AE, UK; Natural Resources Institute, University of Greenwich, Chatham, Kent ME4 4TB, UK
| | - Davide Chiarugi
- Bioinformatics and Biostatistics Core, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK; Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road Cambridge, CB2 0QQ, UK.
| |
Collapse
|
35
|
Subramanian P, Gargani S, Palladini A, Chatzimike M, Grzybek M, Peitzsch M, Papanastasiou AD, Pyrina I, Ntafis V, Gercken B, Lesche M, Petzold A, Sinha A, Nati M, Thangapandi VR, Kourtzelis I, Andreadou M, Witt A, Dahl A, Burkhardt R, Haase R, Domingues AMDJ, Henry I, Zamboni N, Mirtschink P, Chung KJ, Hampe J, Coskun Ü, Kontoyiannis DL, Chavakis T. The RNA binding protein human antigen R is a gatekeeper of liver homeostasis. Hepatology 2022; 75:881-897. [PMID: 34519101 DOI: 10.1002/hep.32153] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.
Collapse
Affiliation(s)
- Pallavi Subramanian
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Sofia Gargani
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Alessandra Palladini
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Margarita Chatzimike
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Michal Grzybek
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Anastasios D Papanastasiou
- Department of Biomedical SciencesUniversity of West AtticaAthensGreece.,Histopathology UnitBiomedical Sciences Research Center "Alexander Fleming"VariGreece
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Vasileios Ntafis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Mathias Lesche
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Andreas Petzold
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Anupam Sinha
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Marina Nati
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Veera Raghavan Thangapandi
- Department of Internal Medicine IUniversity Hospital and Faculty of Medicine, Technische Universität DresdenDresdenGermany
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany.,National Center for Tumor DiseasesPartner Site Dresden, Dresden and German Cancer Research CenterHeidelbergGermany.,York Biomedical Research Institute, Hull York Medical SchoolUniversity of YorkYorkUK
| | - Margarita Andreadou
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Anke Witt
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Andreas Dahl
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Hospital RegensburgRegensburgGermany
| | - Robert Haase
- Scientific Computing FacilityMax Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | | | - Ian Henry
- Scientific Computing FacilityMax Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Nicola Zamboni
- Institute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Jochen Hampe
- Department of Internal Medicine IUniversity Hospital and Faculty of Medicine, Technische Universität DresdenDresdenGermany
| | - Ünal Coskun
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Dimitris L Kontoyiannis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece.,Department of Genetics, Development & Molecular Biology, School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany.,Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany.,National Center for Tumor DiseasesPartner Site Dresden, Dresden and German Cancer Research CenterHeidelbergGermany
| |
Collapse
|
36
|
Seyres D, Cabassi A, Lambourne JJ, Burden F, Farrow S, McKinney H, Batista J, Kempster C, Pietzner M, Slingsby O, Cao TH, Quinn PA, Stefanucci L, Sims MC, Rehnstrom K, Adams CL, Frary A, Ergüener B, Kreuzhuber R, Mocciaro G, D'Amore S, Koulman A, Grassi L, Griffin JL, Ng LL, Park A, Savage DB, Langenberg C, Bock C, Downes K, Wareham NJ, Allison M, Vacca M, Kirk PDW, Frontini M. Transcriptional, epigenetic and metabolic signatures in cardiometabolic syndrome defined by extreme phenotypes. Clin Epigenetics 2022; 14:39. [PMID: 35279219 PMCID: PMC8917653 DOI: 10.1186/s13148-022-01257-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This work is aimed at improving the understanding of cardiometabolic syndrome pathophysiology and its relationship with thrombosis by generating a multi-omic disease signature. METHODS/RESULTS We combined classic plasma biochemistry and plasma biomarkers with the transcriptional and epigenetic characterisation of cell types involved in thrombosis, obtained from two extreme phenotype groups (morbidly obese and lipodystrophy) and lean individuals to identify the molecular mechanisms at play, highlighting patterns of abnormal activation in innate immune phagocytic cells. Our analyses showed that extreme phenotype groups could be distinguished from lean individuals, and from each other, across all data layers. The characterisation of the same obese group, 6 months after bariatric surgery, revealed the loss of the abnormal activation of innate immune cells previously observed. However, rather than reverting to the gene expression landscape of lean individuals, this occurred via the establishment of novel gene expression landscapes. NETosis and its control mechanisms emerge amongst the pathways that show an improvement after surgical intervention. CONCLUSIONS We showed that the morbidly obese and lipodystrophy groups, despite some differences, shared a common cardiometabolic syndrome signature. We also showed that this could be used to discriminate, amongst the normal population, those individuals with a higher likelihood of presenting with the disease, even when not displaying the classic features.
Collapse
Affiliation(s)
- Denis Seyres
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
| | - Alessandra Cabassi
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - John J Lambourne
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Frances Burden
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Samantha Farrow
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Harriet McKinney
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Joana Batista
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carly Kempster
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Oliver Slingsby
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Thong Huy Cao
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Paulene A Quinn
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Luca Stefanucci
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew C Sims
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
- Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Karola Rehnstrom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Claire L Adams
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Amy Frary
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Bekir Ergüener
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Roman Kreuzhuber
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Gabriele Mocciaro
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Simona D'Amore
- Addenbrooke's Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
- National Cancer Research Center, IRCCS Istituto Tumori 'Giovanni Paolo II', Viale Orazio Flacco, 65, 70124, Bari, Italy
| | - Albert Koulman
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- MRC Elsie Widdowson Laboratory, Cambridge, UK
- National Institute for Health Research Biomedical Research Centres Core Nutritional Biomarker Laboratory, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- National Institute for Health Research Biomedical Research Centres Core Metabolomics and Lipidomics Laboratory, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Luigi Grassi
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Julian L Griffin
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Leong Loke Ng
- Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Adrian Park
- Addenbrooke's Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David B Savage
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Kate Downes
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- East Midlands and East of England Genomic Laboratory Hub, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Michael Allison
- Addenbrooke's Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Michele Vacca
- Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Puddicombe Way, Cambridge, CB2 0AW, UK.
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
- British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge, UK.
- Institute of Biomedical & Clinical Science, College of Medicine and Health, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
37
|
Guerra S, Mocciaro G, Gastaldelli A. Adipose tissue insulin resistance and lipidome alterations as the characterizing factors of non-alcoholic steatohepatitis. Eur J Clin Invest 2022; 52:e13695. [PMID: 34695228 DOI: 10.1111/eci.13695] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/16/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The prevalence of non-alcoholic fatty liver disease (NAFLD) is now 25% in the general population but increases to more than 55% in subjects with obesity and/or type 2 diabetes. Simple steatosis (NAFL) can develop into more severe forms, that is non-alcoholic steatohepatitis (NASH), cirrhosis and hepatocellular carcinoma leading to death. METHODS In this narrative review, we have discussed the current knowledge in the pathophysiology of fatty liver disease, including both metabolic and non-metabolic factors, insulin resistance, mitochondrial function, as well as the markers of liver damage, giving attention to the alterations in lipid metabolism and production of lipotoxic lipids. RESULTS Insulin resistance, particularly in the adipose tissue, is the main driver of NAFLD due to the excess release of fatty acids. Lipidome analyses have shown that several lipids, including DAGs and ceramides, and especially if they contain saturated lipids, act as bioactive compounds, toxic to the cells. Lipids can also affect mitochondrial function. Not only lipids, but also amino acid metabolism is impaired in NAFL/NASH, and some amino acids, as branched-chain and aromatic amino acids, glutamate, serine and glycine, have been linked to impaired metabolism, insulin resistance and severity of NAFLD and serine is a precursor of ceramides. CONCLUSIONS The measurement of lipotoxic species and adipose tissue dysfunction can help to identify individuals at risk of progression to NASH.
Collapse
Affiliation(s)
- Sara Guerra
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.,Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Gabriele Mocciaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy
| | - Amalia Gastaldelli
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.,Sant'Anna School of Advanced Studies, Pisa, Italy
| |
Collapse
|
38
|
A mouse model of gestational diabetes shows dysregulated lipid metabolism post-weaning, after return to euglycaemia. Nutr Diabetes 2022; 12:8. [PMID: 35169132 PMCID: PMC8847647 DOI: 10.1038/s41387-022-00185-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gestational diabetes is associated with increased risk of type 2 diabetes mellitus and cardiovascular disease for the mother in the decade after delivery. However, the molecular mechanisms that drive these effects are unknown. Recent studies in humans have shown that lipid metabolism is dysregulated before diagnosis of and during gestational diabetes and we have shown previously that lipid metabolism is also altered in obese female mice before, during and after pregnancy. These observations led us to the hypothesis that this persistent dysregulation reflects an altered control of lipid distribution throughout the organism. METHODS We tested this in post-weaning (PW) dams using our established mouse model of obese GDM (high fat, high sugar, obesogenic diet) and an updated purpose-built computational tool for plotting the distribution of lipid variables throughout the maternal system (Lipid Traffic Analysis v2.3). RESULTS This network analysis showed that unlike hyperglycaemia, lipid distribution and traffic do not return to normal after pregnancy in obese mouse dams. A greater range of phosphatidylcholines was found throughout the lean compared to obese post-weaning dams. A range of triglycerides that were found in the hearts of lean post-weaning dams were only found in the livers of obese post-weaning dams and the abundance of odd-chain FA-containing lipids differed locally in the two groups. We have therefore shown that the control of lipid distribution changed for several metabolic pathways, with evidence for changes to the regulation of phospholipid biosynthesis and FA distribution, in a number of tissues. CONCLUSIONS We conclude that the control of lipid metabolism is altered following an obese pregnancy. These results support the hypothesis that obese dams that developed GDM maintain dysregulated lipid metabolism after pregnancy even when glycaemia returned to normal, and that these alterations could contribute to the increased risk of later type 2 diabetes and cardiovascular disease.
Collapse
|
39
|
Furse S, Watkins AJ, Williams HEL, Snowden SG, Chiarugi D, Koulman A. Paternal nutritional programming of lipid metabolism is propagated through sperm and seminal plasma. Metabolomics 2022; 18:13. [PMID: 35141784 PMCID: PMC8828597 DOI: 10.1007/s11306-022-01869-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND The paternal diet affects lipid metabolism in offspring for at least two generations through nutritional programming. However, we do not know how this is propagated to the offspring. OBJECTIVES We tested the hypothesis that the changes in lipid metabolism that are driven by paternal diet are propagated through spermatozoa and not seminal plasma. METHODS We applied an updated, purpose-built computational network analysis tool to characterise control of lipid metabolism systemically (Lipid Traffic Analysis v2.3) on a known mouse model of paternal nutritional programming. RESULTS The analysis showed that the two possible routes for programming effects, the sperm (genes) and seminal plasma (influence on the uterine environment), both have a distinct effect on the offspring's lipid metabolism. Further, the programming effects in offspring suggest that changes in lipid distribution are more important than alterations in lipid biosynthesis. CONCLUSIONS These results show how the uterine environment and genes both affect lipid metabolism in offspring, enhancing our understanding of the link between parental diet and metabolism in offspring.
Collapse
Affiliation(s)
- Samuel Furse
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
- Metabolic Disease Unit, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
- Biological Chemistry Group, Jodrell Laboratory, Royal Botanic Gardens Kew, Richmond, UK.
| | - Adam J Watkins
- Division of Child Health, Obstetrics and Gynaecology, Faculty of Medicine, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Huw E L Williams
- Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Stuart G Snowden
- Department of Biological Sciences, Royal Holloway College, University of London, Egham, TW20 0EX, Surrey, UK
| | - Davide Chiarugi
- Bioinformatics and Biostatistics Core, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
- Metabolic Disease Unit, Wellcome Trust-MRL Institute of Metabolic Science, University of Cambridge, Addenbrooke's Treatment Centre, Keith Day Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
40
|
Charidemou E, Tsiarli MA, Theophanous A, Yilmaz V, Pitsouli C, Strati K, Griffin JL, Kirmizis A. Histone acetyltransferase NAA40 modulates acetyl-CoA levels and lipid synthesis. BMC Biol 2022; 20:22. [PMID: 35057804 PMCID: PMC8781613 DOI: 10.1186/s12915-021-01225-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 12/30/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Epigenetic regulation relies on the activity of enzymes that use sentinel metabolites as cofactors to modify DNA or histone proteins. Thus, fluctuations in cellular metabolite levels have been reported to affect chromatin modifications. However, whether epigenetic modifiers also affect the levels of these metabolites and thereby impinge on downstream metabolic pathways remains largely unknown. Here, we tested this notion by investigating the function of N-alpha-acetyltransferase 40 (NAA40), the enzyme responsible for N-terminal acetylation of histones H2A and H4, which has been previously implicated with metabolic-associated conditions such as age-dependent hepatic steatosis and calorie-restriction-mediated longevity. RESULTS Using metabolomic and lipidomic approaches, we found that depletion of NAA40 in murine hepatocytes leads to significant increase in intracellular acetyl-CoA levels, which associates with enhanced lipid synthesis demonstrated by upregulation in de novo lipogenesis genes as well as increased levels of diglycerides and triglycerides. Consistently, the increase in these lipid species coincide with the accumulation of cytoplasmic lipid droplets and impaired insulin signalling indicated by decreased glucose uptake. However, the effect of NAA40 on lipid droplet formation is independent of insulin. In addition, the induction in lipid synthesis is replicated in vivo in the Drosophila melanogaster larval fat body. Finally, supporting our results, we find a strong association of NAA40 expression with insulin sensitivity in obese patients. CONCLUSIONS Overall, our findings demonstrate that NAA40 affects the levels of cellular acetyl-CoA, thereby impacting lipid synthesis and insulin signalling. This study reveals a novel path through which histone-modifying enzymes influence cellular metabolism with potential implications in metabolic disorders.
Collapse
Affiliation(s)
- Evelina Charidemou
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus
| | - Maria A Tsiarli
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus
| | - Andria Theophanous
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus
| | - Vural Yilmaz
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus
| | - Katerina Strati
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, CB2 1GA, UK
- Hammersmith Campus, UK Dementia Research Institute at Imperial College, Burlington Danes Building, Imperial College London, Du Cane Road, London, W12 0NN, UK
- Section of Biomolecular Medicine, Department of Metabolism, Division of Systems Medicine, Digestion and Reproduction, The Sir Alexander Fleming Building, Exhibition Road, South Kensington, Imperial College London, London, SW7 2AZ, UK
| | - Antonis Kirmizis
- Department of Biological Sciences, University of Cyprus, 2109, Nicosia, Cyprus.
| |
Collapse
|
41
|
Stratifying individuals into non-alcoholic fatty liver disease risk levels using time series machine learning models. J Biomed Inform 2022; 126:103986. [PMID: 35007752 DOI: 10.1016/j.jbi.2022.103986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/01/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects 25% of the population worldwide, and its prevalence is anticipated to increase globally. While most NAFLD patients are asymptomatic, NAFLD may progress to fibrosis, cirrhosis, cardiovascular disease, and diabetes. Research reports, with daunting results, show the challenge that NAFLD's burden causes to global population health. The current process for identifying fibrosis risk levels is inefficient, expensive, does not cover all potential populations, and does not identify the risk in time. Instead of invasive liver biopsies, we implemented a non-invasive fibrosis assessment process calculated from clinical data (accessed via EMRs/EHRs). We stratified patients' risks for fibrosis from 2007 to 2017 by modeling the risk in 5579 individuals. The process involved time-series machine learning models (Hidden Markov Models and Group-Based Trajectory Models) profiled fibrosis risk by modeling patients' latent medical status resulted in three groups. The high-risk group had abnormal lab test values and a higher prevalence of chronic conditions. This study can help overcome the inefficient, traditional process of detecting fibrosis via biopsies (that are also medically unfeasible due to their invasive nature, the medical resources involved, and costs) at early stages. Thus longitudinal risk assessment may be used to make population-specific medical recommendations targeting early detection of high risk patients, to avoid the development of fibrosis disease and its complications as well as decrease healthcare costs.
Collapse
|
42
|
Sanz-Lamora H, Marrero PF, Haro D, Relat J. A Mixture of Pure, Isolated Polyphenols Worsens the Insulin Resistance and Induces Kidney and Liver Fibrosis Markers in Diet-Induced Obese Mice. Antioxidants (Basel) 2022; 11:120. [PMID: 35052623 PMCID: PMC8772794 DOI: 10.3390/antiox11010120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity is a worldwide epidemic with severe metabolic consequences. Polyphenols are secondary metabolites in plants and the most abundant dietary antioxidants, which possess a wide range of health effects. The most relevant food sources are fruit and vegetables, red wine, black and green tea, coffee, virgin olive oil, and chocolate, as well as nuts, seeds, herbs, and spices. The aim of this work was to evaluate the ability of a pure, isolated polyphenol supplementation to counteract the pernicious metabolic effects of a high-fat diet (HFD). Our results indicated that the administration of pure, isolated polyphenols under HFD conditions for 26 weeks worsened the glucose metabolism in diet-induced obese mice. The data showed that the main target organ for these undesirable effects were the kidneys, where we observed fibrotic, oxidative, and kidney-disease markers. This work led us to conclude that the administration of pure polyphenols as a food supplement would not be advisable. Instead, the ingestion of complete "whole" foods would be the best way to get the health effects of bioactive compounds such as polyphenols.
Collapse
Affiliation(s)
- Hèctor Sanz-Lamora
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute for Nutrition and Food Safety Research, University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
| | - Pedro F. Marrero
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute of Biomedicine, University of Barcelona (IBUB), E-08028 Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Diego Haro
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute of Biomedicine, University of Barcelona (IBUB), E-08028 Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - Joana Relat
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921 Santa Coloma de Gramenet, Spain; (H.S.-L.); (P.F.M.)
- Institute for Nutrition and Food Safety Research, University of Barcelona (INSA-UB), E-08921 Santa Coloma de Gramenet, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| |
Collapse
|
43
|
Jao J, Balmert LC, Sun S, McComsey GA, Brown TT, Tien PC, Currier JS, Stein JH, Qiu Y, LeRoith D, Kurland IJ. Distinct Lipidomic Signatures in People Living With HIV: Combined Analysis of ACTG 5260s and MACS/WIHS. J Clin Endocrinol Metab 2022; 107:119-135. [PMID: 34498048 PMCID: PMC8684537 DOI: 10.1210/clinem/dgab663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Disentangling contributions of HIV from antiretroviral therapy (ART) and understanding the effects of different ART on metabolic complications in persons living with HIV (PLHIV) has been challenging. OBJECTIVE We assessed the effect of untreated HIV infection as well as different antiretroviral therapy (ART) on the metabolome/lipidome. METHODS Widely targeted plasma metabolomic and lipidomic profiling was performed on HIV-seronegative individuals and people living with HIV (PLHIV) before and after initiating ART (tenofovir/emtricitabine plus atazanavir/ritonavir [ATV/r] or darunavir/ritonavir [DRV/r] or raltegravir [RAL]). Orthogonal partial least squares discriminant analysis was used to assess metabolites/lipid subspecies that discriminated between groups. Graphical lasso estimated group-specific metabolite/lipid subspecies networks associated with the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR). Correlations between inflammatory markers and metabolites/lipid subspecies were visualized using heat maps. RESULTS Of 435 participants, 218 were PLHIV. Compared to HIV-seronegative individuals, ART-naive PLHIV exhibited higher levels of saturated triacylglycerols/triglycerides (TAGs) and 3-hydroxy-kynurenine, lower levels of unsaturated TAGs and N-acetyl-tryptophan, and a sparser and less heterogeneous network of metabolites/lipid subspecies associated with HOMA-IR. PLHIV on RAL vs ATV/r or DRV/r had lower saturated and unsaturated TAGs. Positive correlations were found between medium-long chain acylcarnitines (C14-C6 ACs), palmitate, and HOMA-IR for RAL but not ATV/r or DRV/r. Stronger correlations were seen for TAGs with interleukin 6 and high-sensitivity C-reactive protein after RAL vs ATV/r or DRV/r initiation; these correlations were absent in ART-naive PLHIV. CONCLUSION Alterations in the metabolome/lipidome suggest increased lipogenesis for ART-naive PLHIV vs HIV-seronegative individuals, increased TAG turnover for RAL vs ATV/r or DRV/r, and increased inflammation associated with this altered metabolome/lipidome after initiating ART. Future studies are needed to understand cardiometabolic consequences of lipogenesis and inflammation in PLHIV.
Collapse
Affiliation(s)
- Jennifer Jao
- Northwestern University Feinberg School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Diseases, Department of Medicine, Division of Adult Infectious Diseases, Chicago, Illinois 60611, USA
| | - Lauren C Balmert
- Northwestern University Feinberg School of Medicine, Department of Preventive Medicine, Division of Biostatistics, Chicago, Illinois 60611, USA
| | - Shan Sun
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Department of Pediatrics, Division of Pediatric Infectious Diseases, Chicago, Illinois 60611, USA
| | - Grace A McComsey
- University Hospitals Cleveland Medical Center and Case Western Reserve University, Department of Pediatrics, Department of Medicine, Cleveland, Ohio 44106, USA
| | - Todd T Brown
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Phyllis C Tien
- University of California, San Francisco, Department of Medicine and Department of Veterans Affairs Medical Center, Division of Infectious Diseases, San Francisco, California 94121, USA
| | - Judith S Currier
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, California 90095, USA
| | - James H Stein
- University of Wisconsin School of Medicine and Public Health, Department of Medicine, Cardiovascular Medicine Division, Madison, Wisconsin 53726, USA
| | - Yunping Qiu
- Stable Isotope and Metabolomics Core Facility, Department of Medicine, Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Derek LeRoith
- Icahn School of Medicine at Mount Sinai, Department of Medicine, Division of Endocrinology, New York, New York 10029, USA
| | - Irwin J Kurland
- Stable Isotope and Metabolomics Core Facility, Department of Medicine, Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
44
|
O'Brien KA, McNally BD, Sowton AP, Murgia A, Armitage J, Thomas LW, Krause FN, Maddalena LA, Francis I, Kavanagh S, Williams DP, Ashcroft M, Griffin JL, Lyon JJ, Murray AJ. Enhanced hepatic respiratory capacity and altered lipid metabolism support metabolic homeostasis during short-term hypoxic stress. BMC Biol 2021; 19:265. [PMID: 34911556 PMCID: PMC8675474 DOI: 10.1186/s12915-021-01192-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 11/12/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Tissue hypoxia is a key feature of several endemic hepatic diseases, including alcoholic and non-alcoholic fatty liver disease, and organ failure. Hypoxia imposes a severe metabolic challenge on the liver, potentially disrupting its capacity to carry out essential functions including fuel storage and the integration of lipid metabolism at the whole-body level. Mitochondrial respiratory function is understood to be critical in mediating the hepatic hypoxic response, yet the time-dependent nature of this response and the role of the respiratory chain in this remain unclear. RESULTS Here, we report that hepatic respiratory capacity is enhanced following short-term exposure to hypoxia (2 days, 10% O2) and is associated with increased abundance of the respiratory chain supercomplex III2+IV and increased cardiolipin levels. Suppression of this enhanced respiratory capacity, achieved via mild inhibition of mitochondrial complex III, disrupted metabolic homeostasis. Hypoxic exposure for 2 days led to accumulation of plasma and hepatic long chain acyl-carnitines. This was observed alongside depletion of hepatic triacylglycerol species with total chain lengths of 39-53 carbons, containing palmitic, palmitoleic, stearic, and oleic acids, which are associated with de novo lipogenesis. The changes to hepatic respiratory capacity and lipid metabolism following 2 days hypoxic exposure were transient, becoming resolved after 14 days in line with systemic acclimation to hypoxia and elevated circulating haemoglobin concentrations. CONCLUSIONS The liver maintains metabolic homeostasis in response to shorter term hypoxic exposure through transient enhancement of respiratory chain capacity and alterations to lipid metabolism. These findings may have implications in understanding and treating hepatic pathologies associated with hypoxia.
Collapse
Affiliation(s)
- Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.
| | - Ben D McNally
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Sanger Building Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Antonio Murgia
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Sanger Building Tennis Court Road, Cambridge, CB2 1GA, UK
| | - James Armitage
- Global Investigative Safety, GlaxoSmithKline R&D, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Luke W Thomas
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Fynn N Krause
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Sanger Building Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Lucas A Maddalena
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Ian Francis
- Ultrastructure and Cellular Bioimaging, GlaxoSmithKline R&D, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Stefan Kavanagh
- Oncology Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, CB2 OAA, Cambridge, UK
| | - Dominic P Williams
- Functional and Mechanistic Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, CB2 OAA, Cambridge, UK
| | - Margaret Ashcroft
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0QQ, UK
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Sanger Building Tennis Court Road, Cambridge, CB2 1GA, UK
- Section of Biomolecular Medicine, Department of Digestion, Metabolism and Reproduction, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Jonathan J Lyon
- Global Investigative Safety, GlaxoSmithKline R&D, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.
| |
Collapse
|
45
|
TM6SF2/PNPLA3/MBOAT7 Loss-of-Function Genetic Variants Impact on NAFLD Development and Progression Both in Patients and in In Vitro Models. Cell Mol Gastroenterol Hepatol 2021; 13:759-788. [PMID: 34823063 PMCID: PMC8783129 DOI: 10.1016/j.jcmgh.2021.11.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS The I148M Patatin-like Phospholipase Domain-containing 3 (PNPLA3), the rs641738 in the Membrane bound O-acyltransferase domain containing 7-transmembrane channel-like 4 (MBOAT7-TMC4) locus, and the E167K Transmembrane 6 Superfamily Member 2 (TM6SF2) polymorphisms represent the main predisposing factors to nonalcoholic fatty liver disease (NAFLD) development and progression. We previously generated a full knockout of MBOAT7 in HepG2 cells (MBOAT7-/-), homozygous for I148M PNPLA3. Therefore, we aimed to investigate the synergic impact of the 3 at-risk variants on liver injury and hepatocellular carcinoma (HCC) in a large cohort of NAFLD patients, and create in vitro models of genetic NAFLD by silencing TM6SF2 in both HepG2 and MBOAT7-/- cells. METHODS NAFLD patients (n = 1380), of whom 121 had HCC, were stratified with a semiquantitative score ranging from 0 to 3 according to the number of PNPLA3, TM6SF2, and MBOAT7 at-risk variants. TM6SF2 was silenced in HepG2 (TM6SF2-/-) and MBOAT7-/- (MBOAT7-/-TM6SF2-/-) through Clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9). RESULTS In NAFLD patients, the additive weight of these mutations was associated with liver disease severity and an increased risk of developing HCC. In HepG2 cells, TM6SF2 silencing altered lipid composition and induced the accumulation of microvesicular lipid droplets (LDs), whereas the MBOAT7-/-TM6SF2-/- cells showed a mixed microvesicular/macrovesicular pattern of LDs. TM6SF2 deletion strongly affected endoplasmic reticulum and mitochondria ultrastructures, thus increasing endoplasmic reticulum/oxidative stress. The mitochondrial number was increased in both TM6SF2-/- and MBOAT7-/-TM6SF2-/- models, suggesting an unbalancing in mitochondrial dynamics, and the silencing of both MBOAT7 and TM6SF2 impaired mitochondrial activity with a shift toward anaerobic glycolysis. MBOAT7-/-TM6SF2-/- cells also showed the highest proliferation rate. Finally, the re-overexpression of MBOAT7 and/or TM6SF2 reversed the metabolic and tumorigenic features observed in the compound knockout model. CONCLUSIONS The co-presence of the 3 at-risk variants impacts the NAFLD course in both patients and experimental models, affecting LD accumulation, mitochondrial functionality, and metabolic reprogramming toward HCC.
Collapse
|
46
|
Machine Learning-Based Identification of Potentially Novel Non-Alcoholic Fatty Liver Disease Biomarkers. Biomedicines 2021; 9:biomedicines9111636. [PMID: 34829865 PMCID: PMC8615894 DOI: 10.3390/biomedicines9111636] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease that presents a great challenge for treatment and prevention.. This study aims to implement a machine learning approach that employs such datasets to identify potential biomarker targets. We developed a pipeline to identify potential biomarkers for NAFLD that includes five major processes, namely, a pre-processing step, a feature selection and a generation of a random forest model and, finally, a downstream feature analysis and a provision of a potential biological interpretation. The pre-processing step includes data normalising and variable extraction accompanied by appropriate annotations. A feature selection based on a differential gene expression analysis is then conducted to identify significant features and then employ them to generate a random forest model whose performance is assessed based on a receiver operating characteristic curve. Next, the features are subjected to a downstream analysis, such as univariate analysis, a pathway enrichment analysis, a network analysis and a generation of correlation plots, boxplots and heatmaps. Once the results are obtained, the biological interpretation and the literature validation is conducted over the identified features and results. We applied this pipeline to transcriptomics and lipidomic datasets and concluded that the C4BPA gene could play a role in the development of NAFLD. The activation of the complement pathway, due to the downregulation of the C4BPA gene, leads to an increase in triglyceride content, which might further render the lipid metabolism. This approach identified the C4BPA gene, an inhibitor of the complement pathway, as a potential biomarker for the development of NAFLD.
Collapse
|
47
|
Flores YN, Amoon AT, Su B, Velazquez-Cruz R, Ramírez-Palacios P, Salmerón J, Rivera-Paredez B, Sinsheimer JS, Lusis AJ, Huertas-Vazquez A, Saab S, Glenn BA, May FP, Williams KJ, Bastani R, Bensinger SJ. Serum lipids are associated with nonalcoholic fatty liver disease: a pilot case-control study in Mexico. Lipids Health Dis 2021; 20:136. [PMID: 34629052 PMCID: PMC8504048 DOI: 10.1186/s12944-021-01526-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver disease and cirrhosis. NAFLD is mediated by changes in lipid metabolism and known risk factors include obesity, metabolic syndrome, and diabetes. The aim of this study was to better understand differences in the lipid composition of individuals with NAFLD compared to controls, by performing direct infusion lipidomics on serum biospecimens from a cohort study of adults in Mexico. Methods A nested case-control study was conducted with a sample of 98 NAFLD cases and 100 healthy controls who are participating in an on-going, longitudinal study in Mexico. NAFLD cases were clinically confirmed using elevated liver enzyme tests and liver ultrasound or liver ultrasound elastography, after excluding alcohol abuse, and 100 controls were identified as having at least two consecutive normal alanine aminotransferase (ALT) and aspartate aminotransferase (AST) (< 40 U/L) results in a 6-month period, and a normal liver ultrasound elastography result in January 2018. Samples were analyzed on the Sciex Lipidyzer Platform and quantified with normalization to serum volume. As many as 1100 lipid species can be identified using the Lipidyzer targeted multiple-reaction monitoring list. The association between serum lipids and NAFLD was investigated using analysis of covariance, random forest analysis, and by generating receiver operator characteristic (ROC) curves. Results NAFLD cases had differences in total amounts of serum cholesterol esters, lysophosphatidylcholines, sphingomyelins, and triacylglycerols (TAGs), however, other lipid subclasses were similar to controls. Analysis of individual TAG species revealed increased incorporation of saturated fatty acyl tails in serum of NAFLD cases. After adjusting for age, sex, body mass index, and PNPLA3 genotype, a combined panel of ten lipids predicted case or control status better than an area under the ROC curve of 0.83. Conclusions These preliminary results indicate that the serum lipidome differs in patients with NAFLD, compared to healthy controls, and suggest that assessing the desaturation state of TAGs or a specific lipid panel may be useful clinical tools for the diagnosis of NAFLD. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01526-5.
Collapse
Affiliation(s)
- Yvonne N Flores
- Department of Health Policy and Management, Fielding School of Public Health, University of California, Los Angeles (UCLA), Los Angeles, CA, USA. .,UCLA Center for Cancer Prevention and Control and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA. .,Unidad de Investigación Epidemiológica y en Servicios de Salud, Morelos, Instituto Mexicano del Seguro Social, Cuernavaca, Morelos, Mexico.
| | - Aryana T Amoon
- UCLA Center for Cancer Prevention and Control and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Baolong Su
- UCLA Lipidomics Laboratory, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Rafael Velazquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), Ciudad de México, Mexico
| | - Paula Ramírez-Palacios
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Morelos, Instituto Mexicano del Seguro Social, Cuernavaca, Morelos, Mexico
| | - Jorge Salmerón
- Centro de Investigación en Políticas, Población y Salud, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Berenice Rivera-Paredez
- Centro de Investigación en Políticas, Población y Salud, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Janet S Sinsheimer
- UCLA Department of Human Genetics and Computational Medicine, Los Angeles, CA, USA.,Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - Aldons J Lusis
- UCLA Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA.,UCLA Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Adriana Huertas-Vazquez
- UCLA Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sammy Saab
- UCLA Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA, USA.,Pfleger Liver Institute, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Beth A Glenn
- Department of Health Policy and Management, Fielding School of Public Health, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,UCLA Center for Cancer Prevention and Control and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Folasade P May
- Department of Health Policy and Management, Fielding School of Public Health, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,UCLA Center for Cancer Prevention and Control and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.,UCLA Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA, USA.,Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Kevin J Williams
- UCLA Lipidomics Laboratory, David Geffen School of Medicine, Los Angeles, CA, USA.,UCLA Department of Biological Chemistry, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Roshan Bastani
- Department of Health Policy and Management, Fielding School of Public Health, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,UCLA Center for Cancer Prevention and Control and UCLA-Kaiser Permanente Center for Health Equity, Fielding School of Public Health and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Steven J Bensinger
- UCLA Lipidomics Laboratory, David Geffen School of Medicine, Los Angeles, CA, USA.,UCLA Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
48
|
Lana-Elola E, Cater H, Watson-Scales S, Greenaway S, Müller-Winkler J, Gibbins D, Nemes M, Slender A, Hough T, Keskivali-Bond P, Scudamore CL, Herbert E, Banks GT, Mobbs H, Canonica T, Tosh J, Noy S, Llorian M, Nolan PM, Griffin JL, Good M, Simon M, Mallon AM, Wells S, Fisher EMC, Tybulewicz VLJ. Comprehensive phenotypic analysis of the Dp1Tyb mouse strain reveals a broad range of Down syndrome-related phenotypes. Dis Model Mech 2021; 14:dmm049157. [PMID: 34477842 PMCID: PMC8543064 DOI: 10.1242/dmm.049157] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022] Open
Abstract
Down syndrome (DS), trisomy 21, results in many complex phenotypes including cognitive deficits, heart defects and craniofacial alterations. Phenotypes arise from an extra copy of human chromosome 21 (Hsa21) genes. However, these dosage-sensitive causative genes remain unknown. Animal models enable identification of genes and pathological mechanisms. The Dp1Tyb mouse model of DS has an extra copy of 63% of Hsa21-orthologous mouse genes. In order to establish whether this model recapitulates DS phenotypes, we comprehensively phenotyped Dp1Tyb mice using 28 tests of different physiological systems and found that 468 out of 1800 parameters were significantly altered. We show that Dp1Tyb mice have wide-ranging DS-like phenotypes, including aberrant erythropoiesis and megakaryopoiesis, reduced bone density, craniofacial changes, altered cardiac function, a pre-diabetic state, and deficits in memory, locomotion, hearing and sleep. Thus, Dp1Tyb mice are an excellent model for investigating complex DS phenotype-genotype relationships for this common disorder.
Collapse
Affiliation(s)
| | - Heather Cater
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | | | | | | | | | - Amy Slender
- The Francis Crick Institute, London NW1 1AT, UK
| | - Tertius Hough
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | | | | | | | - Helene Mobbs
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1QW, UK
| | - Tara Canonica
- School of Psychology, Cardiff University, Cardiff CF10 3AT, UK
| | - Justin Tosh
- The Francis Crick Institute, London NW1 1AT, UK
- UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Suzanna Noy
- UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | | | | | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge CB2 1QW, UK
- Imperial College Dementia Research Institute, Imperial College London, London W12 7TA, UK
| | - Mark Good
- School of Psychology, Cardiff University, Cardiff CF10 3AT, UK
| | - Michelle Simon
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | - Sara Wells
- MRC Harwell Institute, Harwell Campus, Didcot, OX11 0RD, UK
| | | | - Victor L. J. Tybulewicz
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| |
Collapse
|
49
|
Harshfield EL, Fauman EB, Stacey D, Paul DS, Ziemek D, Ong RMY, Danesh J, Butterworth AS, Rasheed A, Sattar T, Zameer-Ul-Asar, Saleem I, Hina Z, Ishtiaq U, Qamar N, Mallick NH, Yaqub Z, Saghir T, Rizvi SNH, Memon A, Ishaq M, Rasheed SZ, Memon FUR, Jalal A, Abbas S, Frossard P, Saleheen D, Wood AM, Griffin JL, Koulman A. Genome-wide analysis of blood lipid metabolites in over 5000 South Asians reveals biological insights at cardiometabolic disease loci. BMC Med 2021; 19:232. [PMID: 34503513 PMCID: PMC8431908 DOI: 10.1186/s12916-021-02087-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 08/04/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Genetic, lifestyle, and environmental factors can lead to perturbations in circulating lipid levels and increase the risk of cardiovascular and metabolic diseases. However, how changes in individual lipid species contribute to disease risk is often unclear. Moreover, little is known about the role of lipids on cardiovascular disease in Pakistan, a population historically underrepresented in cardiovascular studies. METHODS We characterised the genetic architecture of the human blood lipidome in 5662 hospital controls from the Pakistan Risk of Myocardial Infarction Study (PROMIS) and 13,814 healthy British blood donors from the INTERVAL study. We applied a candidate causal gene prioritisation tool to link the genetic variants associated with each lipid to the most likely causal genes, and Gaussian Graphical Modelling network analysis to identify and illustrate relationships between lipids and genetic loci. RESULTS We identified 253 genetic associations with 181 lipids measured using direct infusion high-resolution mass spectrometry in PROMIS, and 502 genetic associations with 244 lipids in INTERVAL. Our analyses revealed new biological insights at genetic loci associated with cardiometabolic diseases, including novel lipid associations at the LPL, MBOAT7, LIPC, APOE-C1-C2-C4, SGPP1, and SPTLC3 loci. CONCLUSIONS Our findings, generated using a distinctive lipidomics platform in an understudied South Asian population, strengthen and expand the knowledge base of the genetic determinants of lipids and their association with cardiometabolic disease-related loci.
Collapse
Affiliation(s)
- Eric L Harshfield
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK. .,Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, Massachusetts, 02139, USA
| | - David Stacey
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Dirk S Paul
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB2 0QQ, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK.,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK.,Department of Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Daniel Ziemek
- Inflammation and Immunology, Pfizer Worldwide Research, Development and Medical, 10785, Berlin, Germany
| | - Rachel M Y Ong
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB2 0QQ, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK.,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK.,Department of Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB2 0QQ, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK.,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK.,Department of Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan
| | - Taniya Sattar
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan
| | - Zameer-Ul-Asar
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan
| | - Imran Saleem
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan
| | - Zoubia Hina
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan
| | - Unzila Ishtiaq
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan
| | - Nadeem Qamar
- National Institute of Cardiovascular Diseases, Karachi, 75510, Pakistan
| | | | - Zia Yaqub
- National Institute of Cardiovascular Diseases, Karachi, 75510, Pakistan
| | - Tahir Saghir
- National Institute of Cardiovascular Diseases, Karachi, 75510, Pakistan
| | | | - Anis Memon
- National Institute of Cardiovascular Diseases, Karachi, 75510, Pakistan
| | - Mohammad Ishaq
- Karachi Institute of Heart Diseases, Karachi, 75950, Pakistan
| | | | | | - Anjum Jalal
- Faisalabad Institute of Cardiology, Faisalabad, 38000, Pakistan
| | - Shahid Abbas
- Faisalabad Institute of Cardiology, Faisalabad, 38000, Pakistan
| | | | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, 75300, Pakistan.,Department of Biostatistics & Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Angela M Wood
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.,British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB2 0QQ, UK.,National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK.,National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK.,Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK.,Department of Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Julian L Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, CB2 1GA, UK. .,Section of Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, SW7 2AZ, UK.
| | - Albert Koulman
- Core Metabolomics and Lipidomics Laboratory, National Institute for Health Research, Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
50
|
Furse S, Fernandez-Twinn DS, Chiarugi D, Koulman A, Ozanne SE. Lipid Metabolism Is Dysregulated before, during and after Pregnancy in a Mouse Model of Gestational Diabetes. Int J Mol Sci 2021; 22:7452. [PMID: 34299070 PMCID: PMC8306994 DOI: 10.3390/ijms22147452] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/29/2022] Open
Abstract
The aim of the current study was to test the hypothesis that maternal lipid metabolism was modulated during normal pregnancy and that these modulations are altered in gestational diabetes mellitus (GDM). We tested this hypothesis using an established mouse model of diet-induced obesity with pregnancy-associated loss of glucose tolerance and a novel lipid analysis tool, Lipid Traffic Analysis, that uses the temporal distribution of lipids to identify differences in the control of lipid metabolism through a time course. Our results suggest that the start of pregnancy is associated with several changes in lipid metabolism, including fewer variables associated with de novo lipogenesis and fewer PUFA-containing lipids in the circulation. Several of the changes in lipid metabolism in healthy pregnancies were less apparent or occurred later in dams who developed GDM. Some changes in maternal lipid metabolism in the obese-GDM group were so late as to only occur as the control dams' systems began to switch back towards the non-pregnant state. These results demonstrate that lipid metabolism is modulated in healthy pregnancy and the timing of these changes is altered in GDM pregnancies. These findings raise important questions about how lipid metabolism contributes to changes in metabolism during healthy pregnancies. Furthermore, as alterations in the lipidome are present before the loss of glucose tolerance, they could contribute to the development of GDM mechanistically.
Collapse
Affiliation(s)
- Samuel Furse
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK; (S.F.); (D.S.F.-T.)
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK
- Biological Chemistry Group, Jodrell Laboratory, Royal Botanic Gardens Kew, London TW9 3AD, UK
| | - Denise S. Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK; (S.F.); (D.S.F.-T.)
| | - Davide Chiarugi
- Bioinformatics and Biostatistics Core, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK;
| | - Albert Koulman
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK; (S.F.); (D.S.F.-T.)
- Core Metabolomics and Lipidomics Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke’s Treatment Centre, Keith Day Road, Cambridge CB2 0QQ, UK; (S.F.); (D.S.F.-T.)
| |
Collapse
|