1
|
Mateo-Marín MA, Alves-Bezerra M. Targeting acetyl-CoA carboxylases for the treatment of MASLD. J Lipid Res 2024:100676. [PMID: 39461620 DOI: 10.1016/j.jlr.2024.100676] [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: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Hepatic accumulation of triglycerides is a hallmark feature of metabolic dysfunction-associated steatotic liver disease (MASLD). Growing evidence indicates that increased rates of de novo lipogenesis (DNL) is one of the earliest metabolic changes promoting hepatic steatosis in the onset of MASLD. The first step in DNL is catalyzed by acetyl-CoA carboxylases (ACC), which mediate the conversion of acetyl-CoA into malonyl-CoA. Given the critical role of ACC enzymes on DNL, ACC-based therapies have emerged as an attractive approach to address MASLD, leading to the development of pharmacologic inhibitors of ACC. In clinical trials, several of those compounds led to improved DNL rates and hepatic steatosis in MASLD patients. In this review, we describe the development of ACC dual inhibitors and isoform-specific inhibitors along with their clinical testing using monotherapy and combination therapy approaches. We also discuss their efficacy and safety profiles, identifying potential directions for future research. It is anticipated that advances in ACC-based therapies will be critical to the management of MASLD.
Collapse
Affiliation(s)
- María Antonia Mateo-Marín
- Department of Biomedicine, Biotechnology and Public Health, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain
| | - Michele Alves-Bezerra
- Department of Biomedicine, Biotechnology and Public Health, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Faculty of Medicine, University of Cadiz, 11002 Cadiz, Spain.
| |
Collapse
|
2
|
Klingbeil EA, Schade R, Lee SH, Kirkland R, de La Serre CB. Manipulation of feeding patterns in high fat diet fed rats improves microbiota composition dynamics, inflammation and gut-brain signaling. Physiol Behav 2024; 285:114643. [PMID: 39059597 DOI: 10.1016/j.physbeh.2024.114643] [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: 03/07/2024] [Revised: 06/29/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024]
Abstract
Chronic consumption of high fat (HF) diets has been shown to increase meal size and meal frequency in rodents, resulting in overeating. Reducing meal frequency and establishing periods of fasting, independently of caloric intake, may improve obesity-associated metabolic disorders. Additionally, diet-driven changes in microbiota composition have been shown to play a critical role in the development and maintenance of metabolic disorders. In this study, we used a pair-feeding paradigm to reduce meal frequency and snacking episodes while maintaining overall intake and body weight in HF fed rats. We hypothesized that manipulation of feeding patterns would improve microbiota composition and metabolic outcomes. Male Wistar rats were placed in three groups consuming either a HF, low fat diet (LF, matched for sugar), or pair-fed HF diet for 7 weeks (n = 11-12/group). Pair-fed animals received the same amount of food consumed by the HF fed group once daily before dark onset (HF-PF). Rats underwent oral glucose tolerance and gut peptide cholecystokinin sensitivity tests. Bacterial DNA was extracted from the feces collected during both dark and light cycles and sequenced via Illumina MiSeq sequencing of the 16S V4 region. Our pair-feeding paradigm reduced meal numbers, especially small meals in the inactive phase, without changing total caloric intake. This shift in feeding patterns reduced relative abundances of obesity-associated bacteria and maintained circadian fluctuations in microbial abundances. These changes were associated with improved gastrointestinal (GI) function, reduced inflammation, and improved glucose tolerance and gut to brain signaling. We concluded from these data that targeting snacking may help improve metabolic outcomes, independently of energy content of the diet and hyperphagia.
Collapse
Affiliation(s)
- E A Klingbeil
- Department of Nutritional Sciences, The University of Texas at Austin, United States
| | - R Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, United States
| | - S H Lee
- Department of Food Sciences, Sun Moon University, South Korea
| | - R Kirkland
- Office of Research, University of Georgia, United States
| | - C B de La Serre
- Department of Nutritional Sciences, University of Georgia, United States; Department of Biomedical Sciences, Colorado State University, United States.
| |
Collapse
|
3
|
Dusilová T, Kovář J, Laňková I, Thieme L, Hubáčková M, Šedivý P, Pajuelo D, Burian M, Dezortová M, Miklánková D, Malínská H, Svobodová Šťastná P, Poledne R, Hájek M, Haluzík M. Semaglutide Treatment Effects on Liver Fat Content in Obese Subjects with Metabolic-Associated Steatotic Liver Disease (MASLD). J Clin Med 2024; 13:6100. [PMID: 39458050 PMCID: PMC11508983 DOI: 10.3390/jcm13206100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/30/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) represents a major clinical complication of obesity. Methods: In this study, we used magnetic resonance (MR) methods to determine the effect of obesity treatment with semaglutide, a GLP-1 receptor agonist, on the liver fat content and selected metabolic variables. We investigated whether treatment would affect the acute response of liver fat to glucose and fructose administration and whether it would affect the fatty acid profile of VLDL-triglycerides. Sixteen obese non-diabetic men underwent a 16-week dietary intervention and 16-week treatment with subcutaneous semaglutide in a crossover design without a washout period. The order of the interventions was randomized. Results: After treatment, body weight of the subjects decreased by 5% and liver fat by a third, whereas dietary intervention had no impact on these parameters. The decrease in liver fat with semaglutide did not correlate with changes in body weight and other measures of adiposity and was unrelated to improved insulin sensitivity. Conclusions: The proportion of palmitic and palmitoleic acids in VLDL-triglycerides decreased after treatment, suggesting that the beneficial effects of semaglutide on liver fat are mediated by the suppression of de novo lipogenesis.
Collapse
Affiliation(s)
- Tereza Dusilová
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
- Department of Physiology, Faculty of Science, Charles University, 128 44 Prague, Czech Republic
| | - Jan Kovář
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Ivana Laňková
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Lenka Thieme
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Monika Hubáčková
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Petr Šedivý
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Dita Pajuelo
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Martin Burian
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Monika Dezortová
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Denisa Miklánková
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Hana Malínská
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Petra Svobodová Šťastná
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Rudolf Poledne
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Milan Hájek
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| | - Martin Haluzík
- Institute for Clinical and Experimental Medicine, 140 21 Prague, Czech Republic; (T.D.); (I.L.); (L.T.); (M.H.); (P.Š.); (D.P.); (M.B.); (M.D.); (D.M.); (H.M.); (P.S.Š.); (R.P.); (M.H.); (M.H.)
| |
Collapse
|
4
|
Srnic N, Dearlove D, Johnson E, MacLeod C, Krupa A, McGonnell A, Frazer-Morris C, O'Rourke P, Parry S, Hodson L. Greater oxidation of dietary linoleate compared to palmitate in humans following an acute high-carbohydrate diet. Clin Nutr 2024; 43:2305-2315. [PMID: 39226718 DOI: 10.1016/j.clnu.2024.08.028] [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: 04/30/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND We have previously demonstrated that dietary saturated fatty acids (SFA), when compared to polyunsaturated fatty acids (PUFA), are preferentially partitioned into oxidation pathways. However, it remains unclear if this preferential handling is maintained when hepatocellular metabolism is shifted toward fatty acid (FA) esterification and away from oxidation, such as when hepatic de novo lipogenesis (DNL) is upregulated. AIM To investigate whether an acute upregulation of hepatic DNL influences dietary FA partitioning into oxidation pathways. METHODS 20 healthy volunteers (11 females) underwent a fasting baseline visit followed by two study days, 2-weeks apart. Prior to each study day, participants consumed an isocaloric high-carbohydrate diet (to upregulate hepatic DNL) for 3-days. On the two study days, participants consumed an identical standardised test meal that contained either [U13C]palmitate or [U13C]linoleate, in random order, to trace the fate of dietary FA. Blood and breath samples were collected over a 6h postprandial period and 13C enrichment in breath CO2 and plasma lipid fractions were measured using gas-chromatography-combustion-isotope ratio mass spectrometry. RESULTS Compared to the baseline visit, fasting plasma triglyceride concentrations and markers of hepatic DNL, the lipogenic and stearyl-CoA desaturase indices, were significantly (p < 0.05) increased after consumption of the high-carbohydrate diet. Appearance of 13C in expired CO2 and tracer recovery were significantly (p < 0.05) higher after consumption of the meal containing [U13C]linoleate compared to [U13C]palmitate (5.1 ± 0.5% vs. 3.7 ± 0.4%), respectively. Incorporation of 13C into the plasma triglyceride and non-esterified fatty acid pool was significantly (p < 0.001) greater for [U13C]palmitate compared to [U13C]linoleate. CONCLUSION Dietary PUFA compared to SFA appear to be preferentially partitioned into oxidation pathways during an acute upregulation of hepatic DNL, thus consumption of a PUFA-enriched diet may help mitigate intrahepatic triglyceride accumulation in individuals at risk of cardiometabolic disease.
Collapse
Affiliation(s)
- Nikola Srnic
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - David Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Elspeth Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Cameron MacLeod
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Antoni Krupa
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Alice McGonnell
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Charlotte Frazer-Morris
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Paige O'Rourke
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Sion Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Aston Medical School, Aston University, Birmingham B4 7ET, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
5
|
Gaigé S, Abysique A, Barbouche R, Tonetto A, Di Maio A, Robin M, Lormier AT, Troadec JD. 3,5-Dimethyl-2,4,6-trimethoxychalcone Lessens Obesity and MAFLD in Leptin-Deficient ob/ob Mice. Int J Mol Sci 2024; 25:9838. [PMID: 39337328 PMCID: PMC11432508 DOI: 10.3390/ijms25189838] [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: 07/19/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Chalcones constitute an important group of natural compounds abundant in fruits and comestible plants. They are a subject of increasing interest because of their biological activities, including anti-diabetic and anti-obesity effects. The simple chalcone structural scaffold can be modified at multiple sites with different chemical moieties. Here, we generated an artificial chalcone, i.e., 3,5-dimethyl-2,4,6-trimethoxychalcone (TriMetChalc), derived from 2',4'-Dihydroxy-6'-methoxy-3',5'-dimethylchalcone (DMC). DMC is a major compound of Cleistocalyx operculatus, a plant widely used in Asia for its anti-hyperglycemic activity. Using ob/ob mice as an obesity model, we report that, after 3 weeks of per os administration, TriMetChalc modified food intake through the specific activation of brain structures dedicated to the regulation of energy balance. TriMetChalc also decreased weight gain, glucose intolerance, and hepatic steatosis. Moreover, through extensive liver lipidomic analysis, we identified TriMetChalc-induced modifications that could contribute to improving the liver status of the animals. Hence, TriMetChalc is a chalcone derivative capable of reducing food intake and the addition of glucose intolerance and hepatic steatosis in a mouse model of obesity. In light of these results, we believe that TriMetChalc action deserves to be more deeply evaluated over longer treatment periods and/or in combination with other chalcones with protective effects on the liver.
Collapse
Affiliation(s)
- Stéphanie Gaigé
- Centre de Recherche en Psychologie et Neurosciences (CRPN), UMR CNRS UMR 7077, Aix-Marseille University, 13331 Marseille, France; (S.G.); (A.A.); (R.B.)
| | - Anne Abysique
- Centre de Recherche en Psychologie et Neurosciences (CRPN), UMR CNRS UMR 7077, Aix-Marseille University, 13331 Marseille, France; (S.G.); (A.A.); (R.B.)
| | - Rym Barbouche
- Centre de Recherche en Psychologie et Neurosciences (CRPN), UMR CNRS UMR 7077, Aix-Marseille University, 13331 Marseille, France; (S.G.); (A.A.); (R.B.)
| | - Alain Tonetto
- PRATIM, FSCM (FR1739), Centrale Marseille, CNRS, Aix-Marseille University, 13397 Marseille, France;
| | - Attilio Di Maio
- Mediterranean Institute of Marine and Terrestrial Biodiversity and Ecology, IRD, CNRS UMR7263, Aix-Marseille University, 13013 Marseille, France; (A.D.M.); (M.R.)
- Mediterranean Institute of Marine and Terrestrial Biodiversity and Ecology, IRD, NRS UMR7263, Avignon University, 84029 Avignon, France
| | - Maxime Robin
- Mediterranean Institute of Marine and Terrestrial Biodiversity and Ecology, IRD, CNRS UMR7263, Aix-Marseille University, 13013 Marseille, France; (A.D.M.); (M.R.)
- Mediterranean Institute of Marine and Terrestrial Biodiversity and Ecology, IRD, NRS UMR7263, Avignon University, 84029 Avignon, France
- CAYLAB, Contract Research Organization, 13180 Istres, France
| | | | - Jean-Denis Troadec
- Centre de Recherche en Psychologie et Neurosciences (CRPN), UMR CNRS UMR 7077, Aix-Marseille University, 13331 Marseille, France; (S.G.); (A.A.); (R.B.)
| |
Collapse
|
6
|
Westcott F, Dearlove DJ, Hodson L. Hepatic fatty acid and glucose handling in metabolic disease: Potential impact on cardiovascular disease risk. Atherosclerosis 2024; 394:117237. [PMID: 37633797 DOI: 10.1016/j.atherosclerosis.2023.117237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/28/2023]
Abstract
The prevalence of metabolic diseases, including type 2 diabetes mellitus (T2DM) and metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing. Although invariably associated with obesity, the importance of fat deposition in non-adipose tissue organs has yet to be fully explored. Pathological ectopic fat deposition within the liver (known as (MASLD)) has been suggested to underlie the development of T2DM and is now emerging as an independent risk factor for cardiovascular disease (CVD). The process of hepatic de novo lipogenesis (DNL), that is the synthesis of fatty acids from non-lipid precursors (e.g. glucose), has received much attention as it sits at the intersect of hepatic glucose and fatty acid handling. An upregulation of the DNL pathway has been suggested to be central in the development of metabolic diseases (including MASLD, insulin resistance, and T2DM). Here we review the evidence to determine if hepatic DNL may play a role in the development of MASLD and T2DM and therefore underlie an increased risk of CVD.
Collapse
Affiliation(s)
- Felix Westcott
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, UK
| | - David J Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
7
|
Morelli AM, Scholkmann F. Should the standard model of cellular energy metabolism be reconsidered? Possible coupling between the pentose phosphate pathway, glycolysis and extra-mitochondrial oxidative phosphorylation. Biochimie 2024; 221:99-109. [PMID: 38307246 DOI: 10.1016/j.biochi.2024.01.018] [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: 08/07/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
The process of cellular respiration occurs for energy production through catabolic reactions, generally with glucose as the first process step. In the present work, we introduce a novel concept for understanding this process, based on our conclusion that glucose metabolism is coupled to the pentose phosphate pathway (PPP) and extra-mitochondrial oxidative phosphorylation in a closed-loop process. According to the current standard model of glycolysis, glucose is first converted to glucose 6-phosphate (glucose 6-P) and then to fructose 6-phosphate, glyceraldehyde 3-phosphate and pyruvate, which then enters the Krebs cycle in the mitochondria. However, it is more likely that the pyruvate will be converted to lactate. In the PPP, glucose 6-P is branched off from glycolysis and used to produce NADPH and ribulose 5-phosphate (ribulose 5-P). Ribulose 5-P can be converted to fructose 6-P and glyceraldehyde 3-P. In our view, a circular process can take place in which the ribulose 5-P produced by the PPP enters the glycolysis pathway and is then retrogradely converted to glucose 6-P. This process is repeated several times until the complete degradation of glucose 6-P. The role of mitochondria in this process is to degrade lipids by beta-oxidation and produce acetyl-CoA; the function of producing ATP appears to be only secondary. This proposed new concept of cellular bioenergetics allows the resolution of some previously unresolved controversies related to cellular respiration and provides a deeper understanding of metabolic processes in the cell, including new insights into the Warburg effect.
Collapse
Affiliation(s)
| | - Felix Scholkmann
- Neurophotonics and Biosignal Processing Research Group, Biomedical Optics Research Laboratory, Department of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Della Pepa G, Carli F, Sabatini S, Pezzica S, Russo M, Vitale M, Masulli M, Riccardi G, Rivellese AA, Vaccaro O, Bozzetto L, Gastaldelli A. Clusters of adipose tissue dysfunction in adults with type 2 diabetes identify those with worse lipidomic profile despite similar glycaemic control. Diabetes Metab Res Rev 2024; 40:e3798. [PMID: 38558269 DOI: 10.1002/dmrr.3798] [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: 07/18/2023] [Revised: 10/30/2023] [Accepted: 12/31/2023] [Indexed: 04/04/2024]
Abstract
AIMS To investigate clusters of adipose tissue dysfunction, that is, with adipose tissue insulin resistance (ADIPO-IR) and large waist circumference (WC), identify a worse lipidomic profile characterised by a high proportion of lipids rich in saturated fatty acids (SFA). MATERIALS AND METHODS Hierarchical clustering based on WC and ADIPO-IR (calculated as fasting plasma non-esterified fatty acids times fasting plasma insulin, FFA×INS), was performed in 192 adults with overweight/obesity and type 2 diabetes (T2D) treated with metformin (HbA1c = 7.8%). Free fatty acid composition and lipidomic profile were measured by mass spectrometry (GC-MS and LC-MSQTOF). Indexes of fatty acid desaturation (stearoyl-coA desaturase-1 activity, SCD116 = palmitoleic acid/palmitic acid and SCD118 = oleic acid/stearic acid) and of insulin resistance (HOMA-IR) were also calculated. RESULTS Three clusters were identified: CL1 (ADIPO-IR = 4.9 ± 2.4 and WC = 96±7 cm, mean ± SD), CL2 (ADIPO-IR = 6.5 ± 2.5 and WC = 114 ± 7 cm), and CL3 (ADIPO-IR = 15.0 ± 4.7 and WC = 107 ± 8 cm). Insulin concentrations, ADIPO-IR, and HOMA-IR significantly increased from CL1 to CL3 (all p < 0.001), while fasting glucose concentrations, HbA1c, dietary lipids and caloric intake were similar. Moreover, CL3 showed significantly higher concentrations of monounsaturated free fatty acids, oleic and palmitoleic acids, triglycerides (TAG) rich in saturated FA and associated with de novo lipogenesis (i.e., TAG 46-50), higher SCD116, SCD118, ceramide (d18:0/18:0), and phosphatidylcholine aa(36:5) compared with CL1/CL2 (all p < 0.005). CONCLUSIONS High ADIPO-IR and large WC identify a worse lipid profile in T2D characterised by complex lipids rich in SFA, likely due to de novo synthesis given higher plasma monounsaturated FFA and increased desaturase activity indexes. REGISTRATION NUMBER TRIAL ID NCT00700856 https://clinicaltrials.gov.
Collapse
Affiliation(s)
- Giuseppe Della Pepa
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Fabrizia Carli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Silvia Sabatini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Marco Russo
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Marilena Vitale
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Masulli
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Olga Vaccaro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lutgarda Bozzetto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council-CNR, Pisa, Italy
| |
Collapse
|
9
|
Gijbels A, Erdős B, Trouwborst I, Jardon KM, Adriaens ME, Goossens GH, Blaak EE, Feskens EJM, Afman LA. Hepatic insulin resistance and muscle insulin resistance are characterized by distinct postprandial plasma metabolite profiles: a cross-sectional study. Cardiovasc Diabetol 2024; 23:97. [PMID: 38493102 PMCID: PMC10944619 DOI: 10.1186/s12933-024-02188-0] [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: 09/19/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Tissue-specific insulin resistance (IR) predominantly in muscle (muscle IR) or liver (liver IR) has previously been linked to distinct fasting metabolite profiles, but postprandial metabolite profiles have not been investigated in tissue-specific IR yet. Given the importance of postprandial metabolic impairments in the pathophysiology of cardiometabolic diseases, we compared postprandial plasma metabolite profiles in response to a high-fat mixed meal between individuals with predominant muscle IR or liver IR. METHODS This cross-sectional study included data from 214 women and men with BMI 25-40 kg/m2, aged 40-75 years, and with predominant muscle IR or liver IR. Tissue-specific IR was assessed using the muscle insulin sensitivity index (MISI) and hepatic insulin resistance index (HIRI), which were calculated from the glucose and insulin responses during a 7-point oral glucose tolerance test. Plasma samples were collected before (T = 0) and after (T = 30, 60, 120, 240 min) consumption of a high-fat mixed meal and 247 metabolite measures, including lipoproteins, cholesterol, triacylglycerol (TAG), ketone bodies, and amino acids, were quantified using nuclear magnetic resonance spectroscopy. Differences in postprandial plasma metabolite iAUCs between muscle and liver IR were tested using ANCOVA with adjustment for age, sex, center, BMI, and waist-to-hip ratio. P-values were adjusted for a false discovery rate (FDR) of 0.05 using the Benjamini-Hochberg method. RESULTS Sixty-eight postprandial metabolite iAUCs were significantly different between liver and muscle IR. Liver IR was characterized by greater plasma iAUCs of large VLDL (p = 0.004), very large VLDL (p = 0.002), and medium-sized LDL particles (p = 0.026), and by greater iAUCs of TAG in small VLDL (p = 0.025), large VLDL (p = 0.003), very large VLDL (p = 0.002), all LDL subclasses (all p < 0.05), and small HDL particles (p = 0.011), compared to muscle IR. In liver IR, the postprandial plasma fatty acid (FA) profile consisted of a higher percentage of saturated FA (p = 0.013), and a lower percentage of polyunsaturated FA (p = 0.008), compared to muscle IR. CONCLUSION People with muscle IR or liver IR have distinct postprandial plasma metabolite profiles, with more unfavorable postprandial metabolite responses in those with liver IR compared to muscle IR.
Collapse
Grants
- AF-16505 The project was organized by and executed under the auspices of TiFN, a public-private partnership on precompetitive research in food and nutrition. Funding for this research was obtained from the industry partners DSM Nutritional Products, FrieslandCampina, Danone Nutricia Research, AMRA Medical AB, and the Top-sector Agri&Food.
- The project was organized by and executed under the auspices of TiFN, a public-private partnership on precompetitive research in food and nutrition. Funding for this research was obtained from the industry partners DSM Nutritional Products, FrieslandCampina, Danone Nutricia Research, AMRA Medical AB, and the Top-sector Agri&Food.
Collapse
Affiliation(s)
- Anouk Gijbels
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands.
| | - Balázs Erdős
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, The Netherlands
| | - Inez Trouwborst
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Kelly M Jardon
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Michiel E Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Paul-Henri Spaaklaan 1, 6229 EN, Maastricht, The Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Ellen E Blaak
- TI Food and Nutrition (TiFN), Nieuwe Kanaal 9A, 6709 PA, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| |
Collapse
|
10
|
Phimphila A, Aung TM, Wongwattanakul M, Maraming P, Tavichakorntrakool R, Proungvitaya T, Daduang J, Proungvitaya S. Serum CCDC25 Levels as a Potential Marker for Metabolic Syndrome. In Vivo 2024; 38:785-793. [PMID: 38418150 PMCID: PMC10905474 DOI: 10.21873/invivo.13502] [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: 11/29/2023] [Revised: 01/13/2024] [Accepted: 01/24/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND/AIM Metabolic syndrome (MetS) stands as a significant risk for developing various severe health problems. Therefore, the discovery of biomarkers capable of predicting the progression of metabolic conditions is crucial for improving overall health outcomes. Recently, we reported that coiled-coil domain containing 25 (CCDC25) might be associated with key proteins involved in metabolic pathways, by bioinformatics analysis. Thus, we assumed that serum CCDC25 levels might have an association with MetS status. PATIENTS AND METHODS In this study, based on the modified National Cholesterol Education Program-Adult Treatment Panel III (modified NCEP-ATP III) criteria, the participants who had three or more of abnormal criteria were defined as MetS, and those who had 1 or 2 abnormal criteria as pre-MetS groups; those who had no abnormal criteria were classified as the healthy control (HC) group. Serum CCDC25 levels were measured using the dot blot assay. RESULTS The results showed that serum CCDC25 levels of the MetS group (0.072±0.026 ng/μl) were significantly higher (p<0.001) than that of pre-MetS (0.031±0.011 ng/μl) or HC groups (0.018±0.007 ng/μl). We can discern a consistent trend indicating that serum CCDC25 level is well correlated with the number of abnormal criteria of MetS of each participant. Although serum CCDC25 levels correlated with the distribution of all 5 MetS criteria, the highest correlation was seen in serum CCDC25 levels and triglyceride (TG) levels, with r=0.563, followed by systolic blood pressure (SBP) levels (r=0.557) and high-density lipoprotein-cholesterol (HDL-C) levels (r=-0.545). CONCLUSION CCDC25 showed correlations with all MetS parameters, particularly with TG, SBP, and HDL-C. This prompts speculation that heightened CCDC25 levels may indicate the development and/or progression of those MetS-associated diseases.
Collapse
Affiliation(s)
- Anousone Phimphila
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Tin May Aung
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Molin Wongwattanakul
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
- Center for Innovation and Standard for Medical Technology and Physical Therapy (CISMaP), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Pornsuda Maraming
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Ratree Tavichakorntrakool
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Tanakorn Proungvitaya
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Jureerut Daduang
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Siriporn Proungvitaya
- Centre of Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand;
- Cholangiocarcinoma Research Institute (CARI), Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
11
|
Ma Q, Liu Z, Wang T, Zhao P, Liu M, Wang Y, Zhao W, Yuan Y, Li S. Resensitizing Paclitaxel-Resistant Ovarian Cancer via Targeting Lipid Metabolism Key Enzymes CPT1A, SCD and FASN. Int J Mol Sci 2023; 24:16503. [PMID: 38003694 PMCID: PMC10671839 DOI: 10.3390/ijms242216503] [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: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is a lethal gynecological cancer, of which paclitaxel resistance is the major factor limiting treatment outcomes, and identification of paclitaxel resistance-related genes is arduous. We obtained transcriptomic data from seven paclitaxel-resistant ovarian cancer cell lines and corresponding sensitive cell lines. Define genes significantly up-regulated in at least three resistant cell lines, meanwhile they did not down-regulate in the other resistant cell lines as candidate genes. Candidate genes were then ranked according to the frequencies of significant up-regulation in resistant cell lines, defining genes with the highest rankings as paclitaxel resistance-related genes (PRGs). Patients were grouped based on the median expression of PRGs. The lipid metabolism-related gene set and the oncological gene set were established and took intersections with genes co-upregulated with PRGs, obtaining 229 co-upregulated genes associated with lipid metabolism and tumorigenesis. The PPI network obtained 19 highly confidential synergistic targets (interaction score > 0.7) that directly associated with CPT1A. Finally, FASN and SCD were up-stream substrate provider and competitor of CPT1A, respectively. Western blot and qRT-PCR results confirmed the over-expression of CPT1A, SCD and FASN in the A2780/PTX cell line. The inhibition of CPT1A, SCD and FASN down-regulated cell viability and migration, pharmacological blockade of CPT1A and SCD increased apoptosis rate and paclitaxel sensitivity of A2780/PTX. In summary, our novel bioinformatic methods can overcome difficulties in drug resistance evaluation, providing promising therapeutical strategies for paclitaxel-resistant EOC via taregting lipid metabolism-related enzymes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ying Yuan
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang 110122, China; (Q.M.); (Z.L.); (T.W.); (P.Z.); (M.L.); (Y.W.); (W.Z.)
| | - Shuo Li
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang 110122, China; (Q.M.); (Z.L.); (T.W.); (P.Z.); (M.L.); (Y.W.); (W.Z.)
| |
Collapse
|
12
|
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
|
13
|
Tang T, Jiang G, Shao J, Wang M, Zhang X, Xia S, Sun W, Jia X, Wang J, Lai S. lncRNA MSTRG4710 Promotes the Proliferation and Differentiation of Preadipocytes through miR-29b-3p/IGF1 Axis. Int J Mol Sci 2023; 24:15715. [PMID: 37958699 PMCID: PMC10649235 DOI: 10.3390/ijms242115715] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Obesity, a major global health issue, is increasingly associated with the integral role of long non-coding RNA (lncRNA) in adipogenesis. Recently, we found that lncRNA-MSTRG4710 was highly expressed in the liver of rabbits fed a high-fat diet, but whether it is involved in lipid metabolism remains unclear. A series of experiments involving CCK-8, EDU, qPCR, and Oil Red O staining demonstrated that the overexpression of MSTRG4710 stimulated the proliferation and differentiation of preadipocytes while its knockdown inhibited these processes. Bioinformatics analysis showed that miR-29b-3p was a potential target gene of MSTRG4710, and IGF1 was a downstream target gene of miR-29b-3p. Luciferase reporter gene analysis and qPCR analysis confirmed that miR-29b-3p was a potential target gene of MSTRG4710, and miR-29b-3p directly targeted the 3'UTR of IGF1. The overexpression of miR-29b-3p was observed to regulate IGF1 protein and mRNA levels negatively. Additionally, a total of 414 known differentially expressed genes between the miR-29b-3p mimic, miR-29b-3p negative control (NC), siMSTRG4710, and siMSTRG4710-NC group were screened via transcriptome sequencing technology. The GO- and KEGG-enriched pathways were found to be related to lipid metabolism. The study also established that miR-29b-3p targets IGF1 to inhibit preadipocyte proliferation and differentiation. Notably, IGF1 knockdown significantly reduced preadipocyte proliferation and differentiation. Furthermore, co-transfection of pcDNA3.1(+)-MSTRG4710 and mimics into rabbit preadipocytes revealed that the mimics reversed the promotional effect of pcDNA3.1(+)-MSTRG4710. In conclusion, these results uncover that MSTRG4710 positively regulated cell proliferation and adipogenesis by the miR-29b-3p/IGF1 axis. Our findings might provide a new target for studying adipogenesis in rabbit preadipocytes and obesity.
Collapse
Affiliation(s)
- Tao Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Genglong Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiahao Shao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Meigui Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoxiao Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Siqi Xia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Wenqiang Sun
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China (J.W.)
| | - Xianbo Jia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China (J.W.)
| | - Jie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China (J.W.)
| | - Songjia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China (J.W.)
| |
Collapse
|
14
|
Gu W, Wang R, Chai Y, Zhang L, Chen R, Li R, Pan J, Zhu J, Sun Q, Liu C. β3 adrenergic receptor activation alleviated PM 2.5-induced hepatic lipid deposition in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 907:168167. [PMID: 39491202 DOI: 10.1016/j.scitotenv.2023.168167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Increasing energy expenditure through activation of hepatocytes is a potential approach to treat fine particulate matter (PM2.5) induced metabolic-associated fatty liver disease (MAFLD). Beta-3 adrenergic receptor (β3-AR) agonists could stimulate brown adipose tissue (BAT) energy expenditure, but it has never been investigated in MAFLD. The objective of this study is to explore the therapeutic effects of administering CL-316,243, a selective agonist of β3-AR, on hepatic lipid metabolism disturbances induced by PM2.5. Firstly, C57BL/6 N mice were intraperitoneally injected with CL-316,243 for one week. CL-316,243 significantly upregulated expression of β3-AR in the liver, accompanied with reduced serum triglyceride (TG) and free fatty acids (FFA). Next, mice were subjected to PM2.5 exposure for 4 weeks, and CL-316,243 was daily intraperitoneally injected in the fourth week of PM2.5 exposure. Exposure to PM2.5 led to a significant increase in hepatic TG and monounsaturated fatty acids (MUFAs), accompanied with elevated activity of SCD1, increased levels of TG synthesis enzymes and inhibited COX4 activity. Furthermore, the administration of CL-316,243 alleviated PM2.5-induced hepatic lipid deposition by enhancing SCD1 activity, TG lipolysis, fatty acid oxidation and TG synthesis via β3-AR/PKA/CREB/PPAR signaling pathway. Therefore, β3-AR activation may serve as a potential therapeutic approach for PM2.5 exposure-induced MAFLD.
Collapse
Affiliation(s)
- Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Ruiqing Wang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanxi Chai
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Jing Pan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Junyao Zhu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, China.
| |
Collapse
|
15
|
Khandelwal M, Krishna G, Ying Z, Gomez-Pinilla F. Liver acts as a metabolic gate for the traumatic brain injury pathology: Protective action of thyroid hormone. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166728. [PMID: 37137432 PMCID: PMC10601893 DOI: 10.1016/j.bbadis.2023.166728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/16/2023] [Accepted: 04/25/2023] [Indexed: 05/05/2023]
Abstract
Clinical evidence indicates that injury to the brain elicits systemic metabolic disturbances that contributes to the brain pathology. Since dietary fructose is metabolized in the liver, we explored mechanisms by which traumatic brain injury (TBI) and dietary fructose influence liver function and their possible repercussions to brain. Consumption of fructose contributed to the detrimental effects of TBI on liver operation, in terms of glucose and lipid metabolism, de novo lipogenesis, lipid peroxidation. Thyroid hormone (T4) is metabolized in the liver and found that T4 supply improved lipid metabolism by reducing de novo lipogenesis, lipid accumulation, lipogenic enzymes (ACC, AceCS1, FAS), lipid peroxidation in liver in response to fructose and fructose-TBI. T4 supply also helped to normalize glucose metabolism and improve insulin sensitivity. Furthermore, T4 counteracted elevations of the pro-inflammatory cytokines, Tnfα and Mcp-1 after TBI and/or fructose intake in liver and circulation. T4 also exerted an effect on isolated primary hepatocytes by potentiating phosphorylation of AMPKα and AKT substrate, AS160, leading to increased glucose uptake. In addition, T4 restored the metabolism of DHA in the liver disrupted by TBI and fructose, adding important information to optimize the action of DHA in therapeutics. The overall evidence seems to indicate that the liver works as a gate for the regulation of the effects of brain injury and foods on brain pathologies.
Collapse
Affiliation(s)
- Mayuri Khandelwal
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Gokul Krishna
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Zhe Ying
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA; Department of Neurosurgery, UCLA Brain Injury Research Center, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Chaturvedi S, Biswas M, Sadhukhan S, Sonawane A. Role of EGFR and FASN in breast cancer progression. J Cell Commun Signal 2023:10.1007/s12079-023-00771-w. [PMID: 37490191 DOI: 10.1007/s12079-023-00771-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/22/2023] [Indexed: 07/26/2023] Open
Abstract
Breast cancer (BC) emerged as one of the life-threatening diseases among females. Despite notable improvements made in cancer detection and treatment worldwide, according to GLOBACAN 2020, BC is the fifth leading cancer, with an estimated 1 in 6 cancer deaths, in a majority of countries. However, the exact cause that leads to BC progression still needs to be determined. Here, we reviewed the role of two novel biomarkers responsible for 50-70% of BC progression. The first one is epidermal growth factor receptor (EGFR) which belongs to the ErbB tyrosine kinases family, signalling pathways associated with it play a significant role in regulating cell proliferation and division. Another one is fatty acid synthase (FASN), a key enzyme responsible for the de novo lipid synthesis required for cancer cell development. This review presents a rationale for the EGFR-mediated pathways, their interaction with FASN, communion of these two biomarkers with BC, and improvements to overcome drug resistance caused by them.
Collapse
Affiliation(s)
- Suchi Chaturvedi
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India
| | - Mainak Biswas
- School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, 751024, India
| | - Sushabhan Sadhukhan
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India.
- Physical & Chemical Biology Laboratory and Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678623, India.
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Madhya Pradesh, 453552, India.
| |
Collapse
|
17
|
Li LJ, Du R, Ouidir M, Lu R, Chen Z, Weir NL, Tsai MY, Albert PS, Zhang C. Early Pregnancy Maternal Plasma Phospholipid Saturated Fatty Acids and Fetal Growth: Findings from a Multi-Racial/Ethnic Birth Cohort in US. Nutrients 2023; 15:3287. [PMID: 37571228 PMCID: PMC10420908 DOI: 10.3390/nu15153287] [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: 06/05/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Saturated fatty acids (SFAs) during pregnancy are associated with disrupted metabolic programming among offspring at birth and later growth. We examined plasma phospholipid SFAs in early pregnancy and fetal growth throughout pregnancy. We enrolled 321 pregnant women from the NICHD Fetal Growth Studies-Singleton Cohort at gestational weeks 8-13. Ultrasonogram schedules were randomly assigned to capture weekly fetal growth. We measured plasma phospholipid SFAs at early pregnancy using blood samples and modeled fetal growth trajectories across tertiles of SFAs with cubic splines using linear mixed models after full adjustment. We then compared pairwise weekly fetal growth biometrics referencing the lowest tertile in each SFA using the Wald test. We found that even-chain and very long even-chain SFAs were inversely associated, whereas odd-chain SFAs were positively associated with fetal weight and size. Compared with the lowest tertile, the highest tertile of pentadecanoic acid (15:0) had a greater fetal weight and size, starting from week 13 until late pregnancy (at week 39: 3429.89 vs. 3269.08 g for estimated fetal weight; 328.14 vs. 323.00 mm for head circumference). Our findings could inspire future interventions using an alternative high-fat diet rich in odd-chain SFAs for optimal fetal growth.
Collapse
Affiliation(s)
- Ling-Jun Li
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore;
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- NUS Bia-Echo Asia Centre for Reproductive Longevity and Equality (ARCLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- Human Potential Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
| | - Ruochen Du
- Biostatics Unit, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore;
| | - Marion Ouidir
- Institute for Advanced Biosciences, Grenoble Aples University, Site Santé, Allée des Alpes, 38700 La Tronche, France;
| | - Ruijin Lu
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA;
| | - Zhen Chen
- Division of Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Natalie L. Weir
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (N.L.W.); (M.Y.T.)
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA; (N.L.W.); (M.Y.T.)
| | - Paul S. Albert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Cuilin Zhang
- Department of O&G, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore;
- Global Centre for Asian Women’s Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- NUS Bia-Echo Asia Centre for Reproductive Longevity and Equality (ARCLE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
- Human Potential Translation Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117575, Singapore
| |
Collapse
|
18
|
Geidl-Flueck B, Gerber PA. Fructose drives de novo lipogenesis affecting metabolic health. J Endocrinol 2023; 257:e220270. [PMID: 36753292 PMCID: PMC10083579 DOI: 10.1530/joe-22-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/09/2023]
Abstract
Despite the existence of numerous studies supporting a pathological link between fructose consumption and the development of the metabolic syndrome and its sequelae, such as non-alcoholic fatty liver disease (NAFLD), this link remains a contentious issue. With this article, we shed a light on the impact of sugar/fructose intake on hepatic de novo lipogenesis (DNL), an outcome parameter known to be dysregulated in subjects with type 2 diabetes and/or NAFLD. In this review, we present findings from human intervention studies using physiological doses of sugar as well as mechanistic animal studies. There is evidence from both human and animal studies that fructose is a more potent inducer of hepatic lipogenesis than glucose. This is most likely due to the liver's prominent physiological role in fructose metabolism, which may be disrupted under pathological conditions by increased hepatic expression of fructolytic and lipogenic enzymes. Increased DNL may not only contribute to ectopic fat deposition (i.e. in the liver), but it may also impair several metabolic processes through DNL-related fatty acids (e.g. beta-cell function, insulin secretion, or insulin sensitivity).
Collapse
Affiliation(s)
- Bettina Geidl-Flueck
- 1Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| | - Philipp A Gerber
- 1Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| |
Collapse
|
19
|
Otoki Y, Yu D, Shen Q, Sahlas DJ, Ramirez J, Gao F, Masellis M, Swartz RH, Chan PC, Pettersen JA, Kato S, Nakagawa K, Black SE, Swardfager W, Taha AY. Quantitative Lipidomic Analysis of Serum Phospholipids Reveals Dissociable Markers of Alzheimer's Disease and Subcortical Cerebrovascular Disease. J Alzheimers Dis 2023; 93:665-682. [PMID: 37092220 DOI: 10.3233/jad-220795] [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: 04/25/2023]
Abstract
BACKGROUND Circulating phospholipid species have been shown to predict Alzheimer's disease (AD) prognosis but the link between phospholipid disturbances and subcortical small vessel cerebrovascular disease (CeVD) common in AD patients is not known. OBJECTIVE This study used quantitative lipidomics to measure serum diacyl, alkenyl (ether), alkyl, and lyso phospholipid species in individuals with extensive CeVD (n = 29), AD with minimal CeVD (n = 16), and AD with extensive CeVD (n = 14), and compared them to age-matched controls (n = 27). Memory was assessed using the California Verbal Learning Test. 3.0T MRI was used to assess hippocampal volume, atrophy, and white matter hyperintensity (WMH) volumes as manifestations of CeVD. RESULTS AD was associated with significantly higher concentrations of choline plasmalogen 18:0_18:1 and alkyl-phosphocholine 18:1. CeVD was associated with significantly lower lysophospholipids containing 16:0. Phospholipids containing arachidonic acid (AA) were associated with poorer memory in controls, whereas docosahexaenoic acid (DHA)-containing phospholipids were associated with better memory in individuals with AD+CeVD. In controls, DHA-containing phospholipids were associated with more atrophy and phospholipids containing linoleic acid and AA were associated with less atrophy. Lysophospholipids containing 16:0, 18:0, and 18:1 were correlated with less atrophy in controls, and of these, alkyl-phosphocholine 18:1 was correlated with smaller WMH volumes. Conversely, 16:0_18:1 choline plasmalogen was correlated with greater WMH volumes in controls. CONCLUSION This study demonstrates discernable differences in circulating phospholipids in individuals with AD and CeVD, as well as new associations between phospholipid species with memory and brain structure that were specific to contexts of commonly comorbid vascular and neurodegenerative pathologies.
Collapse
Affiliation(s)
- Yurika Otoki
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Di Yu
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada
- Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada
- LC Campbell Cognitive Neurology Unit, Sunnybrook Research Institute, Toronto, Canada
| | - Qing Shen
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA
| | - Demetrios J Sahlas
- Department of Medicine (Neurology Division), McMaster University, Hamilton, Canada
| | - Joel Ramirez
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
| | - Fuqiang Gao
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Medicine (Neurology Division) and the Northern Medical Program, University of British Columbia, Vancouver, Canada
| | - Richard H Swartz
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Pak Cheung Chan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Jacqueline A Pettersen
- Department of Medicine (Neurology Division) and the Northern Medical Program, University of British Columbia, Vancouver, Canada
| | - Shunji Kato
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi, Japan
| | - Sandra E Black
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada
- LC Campbell Cognitive Neurology Unit, Sunnybrook Research Institute, Toronto, Canada
- Department of Medicine (Neurology Division), University of Toronto, Toronto, Canada
| | - Walter Swardfager
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Canada
- Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Canada
- LC Campbell Cognitive Neurology Unit, Sunnybrook Research Institute, Toronto, Canada
- University Health Network Toronto Rehabilitation Institute, Toronto, Canada
| | - Ameer Y Taha
- Department of Food Science and Technology, College of Agriculture and Environmental Sciences, University of California, Davis, CA, USA
- West Coast Metabolomics Center, Genome Center, University of California - Davis, Davis, CA, USA
- Center for Neuroscience, University of California - Davis, Davis, CA, USA
| |
Collapse
|
20
|
Wu Q, Shi D, Dong T, Zhang Z, Ou Q, Fang Y, Zhang C. Serum Saturated Fatty Acids including Very Long-Chain Saturated Fatty Acids and Colorectal Cancer Risk among Chinese Population. Nutrients 2023; 15:nu15081917. [PMID: 37111137 PMCID: PMC10141165 DOI: 10.3390/nu15081917] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The association between circulating saturated fatty acids (SFAs) including very long-chain SFAs (VLCSFAs) and colorectal cancer (CRC) risk has not been clearly established. To investigate the association between serum SFAs and CRC risk in Chinese population, 680 CRC cases and 680 sex and age-matched (5-year interval) controls were recruited in our study. Serum levels of SFAs were detected by gas chromatography. Unconditional logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for the association between serum SFAs and CRC risk. Results showed that total SFAs were positively associated with the risk of CRC (adjusted OR quartile 4 vs. 1 = 2.64, 95%CI: 1.47-4.74). However, VLCSFAs were inversely associated with CRC risk (adjusted OR quartile 4 vs. 1 = 0.51, 95%CI: 0.36-0.72). Specifically, lauric acid, myristic acid, palmitic acid, heptadecanoic acid, and arachidic acid were positively associated with CRC risk, while behenic acid and lignoceric acid were inversely associated with CRC risk. This study indicates that higher levels of total serum SFAs and lower levels of serum VLCSFAs were associated with an increased risk of CRC in Chinese population. To reduce the risk of CRC, we recommend reducing the intake of foods containing palmitic acid and heptadecanoic acid such as animal products and dairy products, and moderately increasing the intake of foods containing VLCSFAs such as peanuts and canola oil.
Collapse
Affiliation(s)
- Qixin Wu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Dandan Shi
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Dong
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhuolin Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qingjian Ou
- Department of Experimental Research, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Yujing Fang
- Department of Experimental Research, Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangzhou 510060, China
| | - Caixia Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
21
|
Rowland LA, Guilherme A, Henriques F, DiMarzio C, Munroe S, Wetoska N, Kelly M, Reddig K, Hendricks G, Pan M, Han X, Ilkayeva OR, Newgard CB, Czech MP. De novo lipogenesis fuels adipocyte autophagosome and lysosome membrane dynamics. Nat Commun 2023; 14:1362. [PMID: 36914626 PMCID: PMC10011520 DOI: 10.1038/s41467-023-37016-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Adipocytes robustly synthesize fatty acids (FA) from carbohydrate through the de novo lipogenesis (DNL) pathway, yet surprisingly DNL contributes little to their abundant triglyceride stored in lipid droplets. This conundrum raises the hypothesis that adipocyte DNL instead enables membrane expansions to occur in processes like autophagy, which requires an abundant supply of phospholipids. We report here that adipocyte Fasn deficiency in vitro and in vivo markedly impairs autophagy, evident by autophagosome accumulation and severely compromised degradation of the autophagic substrate p62. Our data indicate the impairment occurs at the level of autophagosome-lysosome fusion, and indeed, loss of Fasn decreases certain membrane phosphoinositides necessary for autophagosome and lysosome maturation and fusion. Autophagy dependence on FA produced by Fasn is not fully alleviated by exogenous FA in cultured adipocytes, and interestingly, imaging studies reveal that Fasn colocalizes with nascent autophagosomes. Together, our studies identify DNL as a critical source of FAs to fuel autophagosome and lysosome maturation and fusion in adipocytes.
Collapse
Affiliation(s)
- Leslie A Rowland
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| | - Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Sean Munroe
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Nicole Wetoska
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Keith Reddig
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Gregory Hendricks
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
22
|
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
|
23
|
Mitchell CM, Oxtoby LE, Shaw PA, Budge SM, Wooller MJ, Cabeza de Baca T, Krakoff J, Votruba S, O'Brien DM. Carbon Isotope Ratios of Plasma and RBC Fatty Acids Identify Meat Consumers in a 12-Week Inpatient Feeding Study of 32 Men. J Nutr 2023; 152:2847-2855. [PMID: 36095134 PMCID: PMC9839995 DOI: 10.1093/jn/nxac213] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Molecular stable isotope ratios are a novel type of dietary biomarker with high sensitivity and specificity for certain foods. Among these, fatty acid carbon isotope ratios (CIRs) have strong potential but have not been investigated as dietary biomarkers. OBJECTIVES We evaluated whether fatty acid CIRs and mass proportions were associated with meat, fish, and sugar-sweetened beverage (SSB) intake. METHODS Thirty-two men [aged 46.2 ± 10.5 y; BMI (kg/m2): 27.2 ± 4.0] underwent a 12-wk inpatient dietary intervention at the National Institute of Diabetes and Digestive and Kidney Diseases in Phoenix, Arizona. Men were randomly assigned to 1 of 8 dietary treatments varying the presence/absence of dietary meat, fish, and SSBs in all combinations. Fatty acid CIRs and mass proportions were measured in fasting blood samples and adipose tissue biopsies that were collected pre- and postintervention. Dietary effects were analyzed using multivariable regression and receiver operating characteristic AUCs were calculated using logistic regression. RESULTS CIRs of the several abundant SFAs, MUFAs and arachidonic acid (20:4n-6) in plasma were strongly associated with meat, as were a subset of these fatty acids in RBCs. Effect sizes in plasma ranged from 1.01‰ to 1.93‰ and were similar but attenuated in RBCs. Mass proportions of those fatty acids were not associated with diet. CIRs of plasma dihomo-γ-linolenic acid (20:3n-6) and adipose palmitic acid (16:0) were weakly associated with SSBs. Mass proportions of plasma odd-chain fatty acids were associated with meat, and mass proportions of plasma EPA and DHA (20:5n-3 and 22:6n-3) were associated with fish. CONCLUSIONS CIRs of plasma and RBC fatty acids show promise as sensitive and specific measures of dietary meat. These provide different information from that provided by fatty acid mass proportions, and are informative where mass proportion is not. This trial is registered at www.clinicaltrials.gov as NCT01237093.
Collapse
Affiliation(s)
- Cassie M Mitchell
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Laura E Oxtoby
- Center for Alaska Native Health Research, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, USA
- Water and Environmental Research Center, Institute of Northern Engineering, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Pamela A Shaw
- Biostatistics Unit, Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Suzanne M Budge
- Department of Process Engineering and Applied Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Matthew J Wooller
- Water and Environmental Research Center, Institute of Northern Engineering, University of Alaska Fairbanks, Fairbanks, AK, USA
- Marine Biology Department, College of Fisheries and Ocean Sciences, University of Alaska Fairbanks, Fairbanks, AK, USA
| | - Tomás Cabeza de Baca
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Susanne Votruba
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Diane M O'Brien
- Center for Alaska Native Health Research, Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK, USA
| |
Collapse
|
24
|
Beebe J, Josephraj S, Wang CJ, Danielson J, Cui Q, Huang C, Barlow L, Zhang RH, Zhang T, Nakshatri H, Dong Z, Li X, Liu JY, Zhang JT. Therapeutic Activity of the Lansoprazole Metabolite 5-Hydroxy Lansoprazole Sulfide in Triple-Negative Breast Cancer by Inhibiting the Enoyl Reductase of Fatty Acid Synthase. J Med Chem 2022; 65:13681-13691. [PMID: 36257066 DOI: 10.1021/acs.jmedchem.2c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fatty acid synthase (FASN), a sole cytosolic enzyme responsible for de-novo lipid synthesis, is overexpressed in cancer but not in normal non-lipogenic tissues. FASN has been targeted, albeit no such inhibitor has been approved. Proton pump inhibitors (PPIs), approved for digestive disorders, were found to inhibit FASN with anticancer activities in attempting to repurpose Food and Drug Administration-approved drugs. Indeed, PPI usage benefited breast cancer patients and increased their response rate. Due to structural similarity, we thought that their metabolites might extend anticancer effects of PPIs by inhibiting FASN. Here, we tested this hypothesis and found that 5-hydroxy lansoprazole sulfide (5HLS), the end lansoprazole metabolite, was more active than lansoprazole in inhibiting FASN function and regulation of NHEJ repair of oxidative DNA damage via PARP1. Surprisingly, 5HLS inhibits the enoyl reductase, whereas lansoprazole inhibits the thioesterase of FASN. Thus, PPI metabolites may contribute to the lasting anticancer effects of PPIs by inhibiting FASN.
Collapse
Affiliation(s)
- Jenny Beebe
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Sophia Josephraj
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Chao J Wang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Jacob Danielson
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Qingbin Cui
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Caoqinglong Huang
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Lincoln Barlow
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Ryan H Zhang
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Taolan Zhang
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Zizheng Dong
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Xiaohong Li
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| | - Jian-Ting Zhang
- Department of Cell & Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio 43614, United States
| |
Collapse
|
25
|
Della Pepa G, Brancato V, Costabile G, Salamone D, Corrado A, Vitale M, Cavaliere C, Mancini M, Salvatore M, Luongo D, Riccardi G, Rivellese AA, Annuzzi G, Bozzetto L. An Isoenergetic Multifactorial Diet Reduces Pancreatic Fat and Increases Postprandial Insulin Response in Patients With Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2022; 45:1935-1942. [PMID: 35862001 DOI: 10.2337/dc22-0605] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/12/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To compare the effect of an isocaloric multifactorial diet with a diet rich in monounsaturated fatty acids (MUFA) and similar macronutrient composition on pancreatic fat (PF) and postprandial insulin response in type 2 diabetes (T2D). RESEARCH DESIGN AND METHODS According to a randomized controlled parallel-group design, 39 individuals with T2D, 35-75 years old, in satisfactory blood glucose control, were assigned to an 8 week isocaloric intervention with a multifactorial diet rich in MUFA, polyunsaturated fatty acids, fiber, polyphenols, and vitamins (n = 18) or a MUFA-rich diet (n = 21). Before/after the intervention, PF content was measured by the proton-density fat fraction using a three-dimensional mDIXON MRI sequence, and plasma insulin and glucose concentrations were measured over a 4 h test meal with a similar composition as the assigned diet. RESULTS After 8 weeks, PF significantly decreased after the multifactorial diet (from 15.7 ± 6.5% to 14.1 ± 6.3%; P = 0.024), while it did not change after the MUFA diet (from 17.1 ± 10.1% to 18.6 ± 10.6%; P = 0.139) with a significant difference between diets (P = 0.014). Postprandial glucose response was similar in the two groups. Early postprandial insulin response (incremental postprandial areas under the curve [iAUC0-120]) significantly increased with the multifactorial diet (from 36,340 ± 34,954 to 44,138 ± 31,878 pmol/L/min; P = 0.037), while it did not change significantly in the MUFA diet (from 31,754 ± 18,446 to 26,976 ± 12,265 pmol/L/min; P = 0.178), with a significant difference between diets (P = 0.023). Changes in PF inversely correlated with changes in early postprandial insulin response (r = -0.383; P = 0.023). CONCLUSIONS In patients with T2D, an isocaloric multifactorial diet, including several beneficial dietary components, markedly reduced PF. This reduction was associated with an improved postprandial insulin response.
Collapse
Affiliation(s)
- Giuseppe Della Pepa
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Giuseppina Costabile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| | - Dominic Salamone
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Alessandra Corrado
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Marilena Vitale
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Marcello Mancini
- Institute of Biostructure and Bioimaging of the National Research Council, Naples, Italy
| | | | - Delia Luongo
- Institute of Biostructure and Bioimaging of the National Research Council, Naples, Italy
| | - Gabriele Riccardi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Angela A Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| | - Giovanni Annuzzi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| | - Lutgarda Bozzetto
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Task Force on Microbiome Studies, Federico II University, Naples, Italy
| |
Collapse
|
26
|
Luo M, Guo J, Lu W, Fang X, Zhang R, Tang M, Luo Q, Liang W, Yu X, Hu C. The mediating role of maternal metabolites between lipids and adverse pregnancy outcomes of gestational diabetes mellitus. Front Med (Lausanne) 2022; 9:925602. [PMID: 36035400 PMCID: PMC9400014 DOI: 10.3389/fmed.2022.925602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common complications of pregnancy, and the demographics of pregnant women have changed in recent decades. GDM is a metabolic disease with short- and long-term adverse effects on both pregnant women and newborns. The metabolic changes and corresponding risk factors should be of great significance in understanding the pathological mechanism of GDM and reducing the incidence of adverse pregnancy outcomes in patients with GDM. The well-known GDM-associated lipids used in clinical tests, such as triglyceride (TG), are thought to play a major role in metabolic changes during GDM, which have a potential causal relationship with abnormal pregnancy outcomes of GDM. Therefore, this study analyzed the relationship between clinical lipid indicators, metabolic profiles, and abnormal pregnancy outcomes in GDM through mediation analysis. By constructing a metabolic atlas of 399 samples from GDM patients in different trimesters, we efficiently detected the key metabolites of adverse pregnancy outcomes and their mediating roles in bridging abnormal lipids and adverse pregnancy outcomes in patients with GDM. Our study confirmed that TG and total cholesterol were independent risk factors for adverse pregnancy outcomes in patients with GDM. Several key metabolites as mediators (e.g., gamma-linolenic acid, heptadecanoic acid, oleic acid, palmitic acid, and palmitoleic acid) have been identified as potential biomarkers for adverse pregnancy outcomes in patients with GDM. These metabolites mainly participate in the biosynthesis of unsaturated fatty acids, which may shed new light on the pathology of GDM and provide insights for further exploration of the molecular mechanisms underlying adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Mingjuan Luo
- Department of Endocrinology and Metabolism, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Jingyi Guo
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenqian Lu
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Xiangnan Fang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
- Department of Endocrinology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mengyang Tang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Qiong Luo
- Department of Obstetrics and Gynecology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wei Liang
- Department of Endocrinology and Metabolism, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- *Correspondence: Xiangtian Yu
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Cheng Hu
| |
Collapse
|
27
|
Sources of Variation in Food-Related Metabolites during Pregnancy. Nutrients 2022; 14:nu14122503. [PMID: 35745237 PMCID: PMC9227758 DOI: 10.3390/nu14122503] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
The extent to which variation in food-related metabolites are attributable to non-dietary factors remains unclear, which may explain inconsistent food-metabolite associations observed in population studies. This study examined the association between non-dietary factors and the serum concentrations of food-related biomarkers and quantified the amount of variability in metabolite concentrations explained by non-dietary factors. Pregnant women (n = 600) from two Canadian birth cohorts completed a validated semi-quantitative food frequency questionnaire, and serum metabolites were measured by multisegment injection-capillary electrophoresis-mass spectrometry. Hierarchical linear modelling and principal component partial R-square (PC-PR2) were used for data analysis. For proline betaine and DHA (mainly exogenous), citrus foods and fish/fish oil intake, respectively, explained the highest proportion of variability relative to non-dietary factors. The unique contribution of dietary factors was similar (15:0, 17:0, hippuric acid, TMAO) or lower (14:0, tryptophan betaine, 3-methylhistidine, carnitine) compared to non-dietary factors (i.e., ethnicity, maternal age, gestational age, pre-pregnancy BMI, physical activity, and smoking) for metabolites that can either be produced endogenously, biotransformed by gut microbiota, and/or derived from multiple food sources. The results emphasize the importance of adjusting for non-dietary factors in future analyses to improve the accuracy and precision of the measures of food intake and their associations with health and disease.
Collapse
|
28
|
Reduction of De Novo Lipogenesis Mediates Beneficial Effects of Isoenergetic Diets on Fatty Liver: Mechanistic Insights from the MEDEA Randomized Clinical Trial. Nutrients 2022; 14:nu14102178. [PMID: 35631319 PMCID: PMC9143579 DOI: 10.3390/nu14102178] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Non-alcoholic liver steatosis (NAS) results from an imbalance between hepatic lipid storage, disposal, and partitioning. A multifactorial diet high in fiber, monounsaturated fatty acids (MUFAs), n-6 and n-3 polyunsaturated fatty acids (PUFAs), polyphenols, and vitamins D, E, and C reduces NAS in people with type 2 diabetes (T2D) by 40% compared to a MUFA-rich diet. We evaluated whether dietary effects on NAS are mediated by changes in hepatic de novo lipogenesis (DNL), stearoyl-CoA desaturase (SCD1) activity, and/or β-oxidation. METHODS According to a randomized parallel group study design, 37 individuals with T2D completed an 8-week isocaloric intervention with a MUFA diet (n = 20) or multifactorial diet (n = 17). Before and after the intervention, liver fat content was evaluated by proton magnetic resonance spectroscopy, serum triglyceride fatty acid concentrations measured by gas chromatography, plasma β-hydroxybutyrate by enzymatic method, and DNL and SCD-1 activity assessed by calculating the palmitic acid/linoleic acid (C16:0/C18:2 n6) and palmitoleic acid/palmitic acid (C16:1/C16:0) ratios, respectively. RESULTS Compared to baseline, mean ± SD DNL significantly decreased after the multifactorial diet (2.2 ± 0.8 vs. 1.5 ± 0.5, p = 0.0001) but did not change after the MUFA diet (1.9 ± 1.1 vs. 1.9 ± 0.9, p = 0.949), with a significant difference between the two interventions (p = 0.004). The mean SCD-1 activity also decreased after the multifactorial diet (0.13 ± 0.05 vs. 0.10 ± 0.03; p = 0.001), but with no significant difference between interventions (p = 0.205). Fasting plasma β-hydroxybutyrate concentrations did not change significantly after the MUFA or multifactorial diet. Changes in the DNL index significantly and positively correlated with changes in liver fat (r = 0.426; p = 0.009). CONCLUSIONS A diet rich in multiple beneficial dietary components (fiber, polyphenols, MUFAs, PUFAs, and other antioxidants) compared to a diet rich only in MUFAs further reduces liver fat accumulation through the inhibition of DNL. Registered under ClinicalTrials.gov no. NCT03380416.
Collapse
|
29
|
Talenezhad N, Mirzavandi F, Rahimpour S, Amel Shahbaz AP, Mohammadi M, Hosseinzadeh M. Empirically derived dietary pattern and odds of non-alcoholic fatty liver diseases in overweight and obese adults: a case-control study. BMC Gastroenterol 2022; 22:158. [PMID: 35354433 PMCID: PMC8966273 DOI: 10.1186/s12876-022-02222-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 03/17/2022] [Indexed: 12/26/2022] Open
Abstract
Background The prevalence of non-alcoholic fatty liver disease (NAFLD) is rising at an exponential rate throughout the world. Given the confirmed association between nutritional status and NAFLD, this study aimed to investigate the relationship of dietary patterns with NAFLD in overweight and obese adults. Methods In this age- and gender-matched case–control study, 115 newly diagnosed cases and 102 control individuals participated. A validated 178-item semi-quantitative food frequency questionnaire was administered to assess the participants' dietary data. Dietary patterns were extracted from 24 predefined food groups by factor analysis. Multivariate logistic regression was run to evaluate the relationship between dietary patterns and NAFLD. Results Factor analysis resulted in: “western”, “traditional”, and “snack and sweets” dietary patterns. The NAFLD odds were greater in participants at the highest quintile of the “western” dietary pattern than the lowest quintile (OR: 3.52; 95% CI: 1.64, 8.61). A significant increasing trend was observed in NAFLD odds across increasing quintiles of the “western” dietary pattern (P-trend = 0.01). After adjusting for the potential confounders, this relationship remained significant (OR: 3.30; 95% CI: 1.06–10.27). After full adjustments, NAFLD had no association with “traditional” or “snack and sweets” dietary patterns. Conclusion The “western” dietary pattern containing fast food, refined grains, liquid oil, pickles, high-fat dairy, sweet desserts, red meat, tea, and coffee was associated with increased odds of NAFLD. However, further prospective studies are required to establish these results.
Collapse
Affiliation(s)
- Nasir Talenezhad
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Farhang Mirzavandi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Shahab Rahimpour
- Gastroentrology Department, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amir Pasha Amel Shahbaz
- Department of Radiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohammad Mohammadi
- Department of Community Medicine, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahdieh Hosseinzadeh
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. .,Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
30
|
Murru E, Manca C, Carta G, Banni S. Impact of Dietary Palmitic Acid on Lipid Metabolism. Front Nutr 2022; 9:861664. [PMID: 35399673 PMCID: PMC8983927 DOI: 10.3389/fnut.2022.861664] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Palmitic acid (PA) is ubiquitously present in dietary fat guaranteeing an average intake of about 20 g/d. The relative high requirement and relative content in the human body, which accounts for 20–30% of total fatty acids (FAs), is justified by its relevant nutritional role. In particular physiological conditions, such as in the fetal stage or in the developing brain, the respectively inefficient placental and brain blood–barrier transfer of PA strongly induces its endogenous biosynthesis from glucose via de novo lipogenesis (DNL) to secure a tight homeostatic control of PA tissue concentration required to exert its multiple physiological activities. However, pathophysiological conditions (insulin resistance) are characterized by a sustained DNL in the liver and aimed at preventing the excess accumulation of glucose, which result in increased tissue content of PA and disrupted homeostatic control of its tissue concentration. This leads to an overaccumulation of tissue PA, which results in dyslipidemia, increased ectopic fat accumulation, and inflammatory tone via toll-like receptor 4. Any change in dietary saturated FAs (SFAs) usually reflects a complementary change in polyunsaturated FA (PUFA) intake. Since PUFA particularly n-3 highly PUFA, suppress lipogenic gene expression, their reduction in intake rather than excess of dietary SFA may promote endogenous PA production via DNL. Thereby, the increase in tissue PA and its deleterious consequences from dysregulated DNL can be mistakenly attributed to dietary intake of PA.
Collapse
|
31
|
Cisbani G, Koppel A, Metherel AH, Smith ME, Aji KN, Andreazza AC, Mizrahi R, Bazinet RP. Serum lipid analysis and isotopic enrichment is suggestive of greater lipogenesis in young long-term cannabis users: A secondary analysis of a case-control study. Lipids 2022; 57:125-140. [PMID: 35075659 PMCID: PMC8923992 DOI: 10.1002/lipd.12336] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 01/15/2023]
Abstract
Cannabis is now legal in many countries and while numerous studies have reported on its impact on cognition and appetite regulation, none have examined fatty acid metabolism in young cannabis users. We conducted an exploratory analysis to evaluate cannabis impact on fatty acid metabolism in cannabis users (n = 21) and non-cannabis users (n = 16). Serum levels of some saturated and monounsaturated fatty acids, including palmitic, palmitoleic, and oleic acids were higher in cannabis users compared to nonusers. As palmitic acid can be derived from diet or lipogenesis from sugars, we evaluated lipogenesis using a de novo lipogenesis index (palmitate/linoleic acid) and carbon-specific isotope analysis, which allows for the determination of fatty acid 13 C signature. The significantly higher de novo lipogenesis index in the cannabis users group along with a more enriched 13 C signature of palmitic acid suggested an increase in lipogenesis. In addition, while serum glucose concentration did not differ between groups, pyruvate and lactate were lower in the cannabis user group, with pyruvate negatively correlating with palmitic acid. Furthermore, the endocannabinoid 2-arachidonoylglycerol was elevated in cannabis users and could contribute to lipogenesis by activating the cannabinoid receptor 1. Because palmitic acid has been suggested to increase inflammation, we measured peripheral cytokines and observed no changes in inflammatory cytokines. Finally, an anti-inflammatory metabolite of palmitic acid, palmitoylethanolamide was elevated in cannabis users. Our results suggest that lipogenic activity is increased in cannabis users; however, future studies, including prospective studies that control dietary intake are required.
Collapse
Affiliation(s)
- Giulia Cisbani
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Alex Koppel
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Adam H. Metherel
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Mackenzie E. Smith
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada
| | - Kankana N. Aji
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Ana C. Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario
| | - Romina Mizrahi
- Department of Psychiatry, McGill University, Montreal, Canada,Douglas Research Center, Montreal, Canada,Corresponding author: Richard P. Bazinet, Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada, Medical Sciences Building, 5th Floor, Room 5358, 1 King’s College Circle, Toronto, ON, M5S 1A8, , Phone number: (416) 946-8276, Romina Mizrahi, Department of Psychiatry, McGill University, 6875 Boulevard Lasalle, Montréal, QC H4H 1R3,
| | - Richard P. Bazinet
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada,Corresponding author: Richard P. Bazinet, Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Canada, Medical Sciences Building, 5th Floor, Room 5358, 1 King’s College Circle, Toronto, ON, M5S 1A8, , Phone number: (416) 946-8276, Romina Mizrahi, Department of Psychiatry, McGill University, 6875 Boulevard Lasalle, Montréal, QC H4H 1R3,
| |
Collapse
|
32
|
Effects of a low free sugar diet on the management of nonalcoholic fatty liver disease: a randomized clinical trial. Eur J Clin Nutr 2022; 76:987-994. [DOI: 10.1038/s41430-022-01081-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022]
|
33
|
Narimatsu Y, Iwakoshi-Ukena E, Fukumura K, Shikano K, Furumitsu M, Morishita M, Bentley GE, Kriegsfeld LJ, Ukena K. Hypothalamic Overexpression of Neurosecretory Protein GL Leads to Obesity in Male C57BL/6J Mice. Neuroendocrinology 2022; 112:606-620. [PMID: 34384081 DOI: 10.1159/000518969] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The mechanisms underlying obesity are not fully understood, necessitating the creation of novel animal models for the investigation of metabolic disorders. We have previously found that neurosecretory protein GL (NPGL), a newly identified hypothalamic neuropeptide, is involved in feeding behavior and fat accumulation in rats. However, the impact of NPGL on obesity remains unclear in any animal model. The present investigation sought to elucidate whether NPGL causes obesity in the obesity-prone mouse strain C57BL/6J. METHODS We overexpressed the NPGL-precursor gene (Npgl) in the hypothalamus using adeno-associated virus in male C57BL/6J mice fed normal chow (NC) or a high-calorie diet (HCD). After 9 weeks of Npgl overexpression, we measured adipose tissues, muscle, and several organ masses in addition to food intake and body mass. To assess the effects of Npgl overexpression on peripheral tissues, we analyzed mRNA expression of lipid metabolism-related genes by quantitative RT-PCR. Whole body energy consumption was assessed using an O2/CO2 metabolism measurement before an apparent increase in body mass. RESULTS Npgl overexpression increased food intake, body mass, adipose tissues and liver masses, and food efficiency under both NC and HCD, resulting in obesity observable within 8 weeks. Furthermore, we observed fat accumulation in adipose tissues and liver. Additionally, mRNA expression of lipid metabolism-related factors was increased in white adipose tissue and the liver after Npgl overexpression. Npgl overexpression inhibited energy expenditure during a dark period. CONCLUSION Taken together, the present study suggests that NPGL can act as an obesogenic factor that acts within a short period of time in mice. As a result, this Npgl overexpression-induced obesity can be widely applied to study the etiology of obesity from genes to behavior.
Collapse
Affiliation(s)
- Yuki Narimatsu
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Eiko Iwakoshi-Ukena
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Keisuke Fukumura
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Kenshiro Shikano
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
- Department of Neurophysiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Megumi Furumitsu
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Masahiro Morishita
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - George E Bentley
- Department of Integrative Biology and the Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| | - Lance J Kriegsfeld
- Department of Psychology, Integrative Biology, and the Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| | - Kazuyoshi Ukena
- Laboratory of Neurometabolism, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
34
|
Fretts AM, Jensen PN, Hoofnagle AN, McKnight B, Sitlani CM, Siscovick DS, King IB, Psaty BM, Sotoodehnia N, Lemaitre RN. Circulating Ceramides and Sphingomyelins and Risk of Mortality: The Cardiovascular Health Study. Clin Chem 2021; 67:1650-1659. [PMID: 34580702 PMCID: PMC8634404 DOI: 10.1093/clinchem/hvab182] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Recent studies suggest that associations of ceramides (Cer) and sphingomyelins (SM) with health outcomes differ according to the fatty acid acylated to the sphingoid backbone. The purpose of this study was to assess associations of Cer and SM species with mortality. METHODS The study population included participants from the Cardiovascular Health Study (CHS), a community-based cohort of adults aged ≥65 years who were followed from 1992-2015 (n = 4612). Associations of plasma Cer and SM species carrying long-chain (i.e., 16:0) and very-long-chain (i.e., 20:0, 22:0, 24:0) saturated fatty acids with mortality were assessed using Cox proportional hazards models. RESULTS During a median follow-up of 10.2 years, 4099 deaths occurred. High concentrations of Cer and SM carrying fatty acid 16:0 were each associated with an increased risk of mortality. Conversely, high concentrations of several ceramide and sphingomyelin species carrying longer fatty acids were each associated with a decreased risk of mortality. The hazard ratios for total mortality per 2-fold difference in each Cer and SM species were: 1.89 (95% CI), 1.65-2.17 for Cer-16, 0.79 (95% CI, 0.70-0.88) for Cer-22, 0.74 (95% CI, 0.65-0.84) for Cer-24, 2.51 (95% CI, 2.01-3.14) for SM-16, 0.68 (95% CI, 0.58-0.79) for SM-20, 0.57 (95% CI, 0.49-0.67) for SM-22, and 0.66 (0.57-0.75) for SM-24. We found no association of Cer-20 with risk of death. CONCLUSIONS Associations of Cer and SM with the risk of death differ according to the length of their acylated saturated fatty acid. Future studies are needed to explore mechanisms underlying these relationships.
Collapse
Affiliation(s)
- Amanda M Fretts
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Paul N Jensen
- Department of Medicine, University of Washington, Seattle, WA
| | - Andrew N Hoofnagle
- Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, WA
| | | | | | - Irena B King
- Department of Medicine, University of New Mexico, Albuquerque, NM
| | - Bruce M Psaty
- Department of Medicine , University of Washington, Seattle, WA
| | | | | |
Collapse
|
35
|
Zhang Z, Lai M, Piro AL, Alexeeff SE, Allalou A, Röst HL, Dai FF, Wheeler MB, Gunderson EP. Intensive lactation among women with recent gestational diabetes significantly alters the early postpartum circulating lipid profile: the SWIFT study. BMC Med 2021; 19:241. [PMID: 34620173 PMCID: PMC8499506 DOI: 10.1186/s12916-021-02095-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Women with a history of gestational diabetes mellitus (GDM) have a 7-fold higher risk of developing type 2 diabetes (T2D). It is estimated that 20-50% of women with GDM history will progress to T2D within 10 years after delivery. Intensive lactation could be negatively associated with this risk, but the mechanisms behind a protective effect remain unknown. METHODS In this study, we utilized a prospective GDM cohort of 1010 women without T2D at 6-9 weeks postpartum (study baseline) and tested for T2D onset up to 8 years post-baseline (n=980). Targeted metabolic profiling was performed on fasting plasma samples collected at both baseline and follow-up (1-2 years post-baseline) during research exams in a subset of 350 women (216 intensive breastfeeding, IBF vs. 134 intensive formula feeding or mixed feeding, IFF/Mixed). The relationship between lactation intensity and circulating metabolites at both baseline and follow-up were evaluated to discover underlying metabolic responses of lactation and to explore the link between these metabolites and T2D risk. RESULTS We observed that lactation intensity was strongly associated with decreased glycerolipids (TAGs/DAGs) and increased phospholipids/sphingolipids at baseline. This lipid profile suggested decreased lipogenesis caused by a shift away from the glycerolipid metabolism pathway towards the phospholipid/sphingolipid metabolism pathway as a component of the mechanism underlying the benefits of lactation. Longitudinal analysis demonstrated that this favorable lipid profile was transient and diminished at 1-2 years postpartum, coinciding with the cessation of lactation. Importantly, when stratifying these 350 women by future T2D status during the follow-up (171 future T2D vs. 179 no T2D), we discovered that lactation induced robust lipid changes only in women who did not develop incident T2D. Subsequently, we identified a cluster of metabolites that strongly associated with future T2D risk from which we developed a predictive metabolic signature with a discriminating power (AUC) of 0.78, superior to common clinical variables (i.e., fasting glucose, AUC 0.56 or 2-h glucose, AUC 0.62). CONCLUSIONS In this study, we show that intensive lactation significantly alters the circulating lipid profile at early postpartum and that women who do not respond metabolically to lactation are more likely to develop T2D. We also discovered a 10-analyte metabolic signature capable of predicting future onset of T2D in IBF women. Our findings provide novel insight into how lactation affects maternal metabolism and its link to future diabetes onset. TRIAL REGISTRATION ClinicalTrials.gov NCT01967030 .
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, Hangzhou, China
| | - Mi Lai
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anthony L Piro
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Stacey E Alexeeff
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Amina Allalou
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hannes L Röst
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Feihan F Dai
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Metabolism Research Group, Division of Advanced Diagnostics, Toronto General Research Institute, Toronto, Ontario, Canada.
| | - Erica P Gunderson
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA.
- Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, USA.
| |
Collapse
|
36
|
von Loeffelholz C, Coldewey SM, Birkenfeld AL. A Narrative Review on the Role of AMPK on De Novo Lipogenesis in Non-Alcoholic Fatty Liver Disease: Evidence from Human Studies. Cells 2021; 10:cells10071822. [PMID: 34359991 PMCID: PMC8306246 DOI: 10.3390/cells10071822] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/01/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
5′AMP-activated protein kinase (AMPK) is known as metabolic sensor in mammalian cells that becomes activated by an increasing adenosine monophosphate (AMP)/adenosine triphosphate (ATP) ratio. The heterotrimeric AMPK protein comprises three subunits, each of which has multiple phosphorylation sites, playing an important role in the regulation of essential molecular pathways. By phosphorylation of downstream proteins and modulation of gene transcription AMPK functions as a master switch of energy homeostasis in tissues with high metabolic turnover, such as the liver, skeletal muscle, and adipose tissue. Regulation of AMPK under conditions of chronic caloric oversupply emerged as substantial research target to get deeper insight into the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Evidence supporting the role of AMPK in NAFLD is mainly derived from preclinical cell culture and animal studies. Dysbalanced de novo lipogenesis has been identified as one of the key processes in NAFLD pathogenesis. Thus, the scope of this review is to provide an integrative overview of evidence, in particular from clinical studies and human samples, on the role of AMPK in the regulation of primarily de novo lipogenesis in human NAFLD.
Collapse
Affiliation(s)
- Christian von Loeffelholz
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany;
- Correspondence: ; Tel.: +49-3641-9323-177; Fax: +49-3641-9323-102
| | - Sina M. Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany;
- Septomics Research Center, Jena University Hospital, 07747 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Andreas L. Birkenfeld
- Department of Diabetology Endocrinology and Nephrology, University Hospital Tübingen, Eberhard Karls University Tübingen, 72074 Tübingen, Germany;
- Department of Therapy of Diabetes, Institute of Diabetes Research and Metabolic Diseases in the Helmholtz Center Munich, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
- Division of Diabetes and Nutritional Sciences, Rayne Institute, King’s College London, London SE5 9RJ, UK
| |
Collapse
|
37
|
Geidl-Flueck B, Hochuli M, Németh Á, Eberl A, Derron N, Köfeler HC, Tappy L, Berneis K, Spinas GA, Gerber PA. Fructose- and sucrose- but not glucose-sweetened beverages promote hepatic de novo lipogenesis: A randomized controlled trial. J Hepatol 2021; 75:46-54. [PMID: 33684506 DOI: 10.1016/j.jhep.2021.02.027] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 02/03/2021] [Accepted: 02/18/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Excessive fructose intake is associated with increased de novo lipogenesis, blood triglycerides, and hepatic insulin resistance. We aimed to determine whether fructose elicits specific effects on lipid metabolism independently of excessive caloric intake. METHODS A total of 94 healthy men were studied in this double-blind, randomized trial. They were assigned to daily consumption of sugar-sweetened beverages (SSBs) containing moderate amounts of fructose, sucrose (fructose-glucose disaccharide) or glucose (80 g/day) in addition to their usual diet or SSB abstinence (control group) for 7 weeks. De novo fatty acid (FA) and triglyceride synthesis, lipolysis and plasma free FA (FFA) oxidation were assessed by tracer methodology. RESULTS Daily intake of beverages sweetened with free fructose and fructose combined with glucose (sucrose) led to a 2-fold increase in basal hepatic fractional secretion rates (FSR) compared to control (median FSR %/day: sucrose 20.8 (p = 0.0015); fructose 19.7 (p = 0.013); control 9.1). Conversely, the same amounts of glucose did not change FSR (median of FSR %/day 11.0 (n.s.)). Fructose intake did not change basal secretion of newly synthesized VLDL-triglyceride, nor did it alter rates of peripheral lipolysis, nor total FA and plasma FFA oxidation. Total energy intake was similar across groups. CONCLUSIONS Regular consumption of both fructose- and sucrose-sweetened beverages in moderate doses - associated with stable caloric intake - increases hepatic FA synthesis even in a basal state; this effect is not observed after glucose consumption. These findings provide evidence of an adaptative response to regular fructose exposure in the liver. LAY SUMMARY This study investigated the metabolic effects of daily sugar-sweetened beverage consumption for several weeks in healthy lean men. It revealed that beverages sweetened with the sugars fructose and sucrose (glucose and fructose combined), but not glucose, increase the ability of the liver to produce lipids. This change may pave the way for further unfavorable effects on metabolic health. CLINICAL TRIAL REGISTRATION NUMBER NCT01733563.
Collapse
Affiliation(s)
- Bettina Geidl-Flueck
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland.
| | - Michel Hochuli
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland; Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital and University of Bern, Switzerland
| | - Ágota Németh
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| | - Anita Eberl
- Institute for Biomedicine and Health Sciences, Joanneum Research, Graz, Austria
| | - Nina Derron
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| | - Harald C Köfeler
- Core Facility Mass Spectrometry, Medical University of Graz, Austria
| | - Luc Tappy
- Department of Physiology, University of Lausanne, Switzerland
| | - Kaspar Berneis
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| | - Giatgen A Spinas
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| | - Philipp A Gerber
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Switzerland
| |
Collapse
|
38
|
Guo L, Chao X, Huang W, Li Z, Luan K, Ye M, Zhang S, Liu M, Li H, Luo W, Nie Q, Zhang X, Luo Q. Whole Transcriptome Analysis Reveals a Potential Regulatory Mechanism of LncRNA-FNIP2/miR-24-3p/ FNIP2 Axis in Chicken Adipogenesis. Front Cell Dev Biol 2021; 9:653798. [PMID: 34249911 PMCID: PMC8265275 DOI: 10.3389/fcell.2021.653798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 01/20/2023] Open
Abstract
Lipid biosynthesis is a complex process, which is regulated by multiple factors including lncRNA. However, the role of lncRNA in chicken abdominal fat accumulation is still unclear. In this research, we collected liver tissues from six high abdominal fat rate Sanhuang broilers and six low abdominal fat rate Sanhuang broilers to perform lncRNA sequencing and small RNA sequencing. A total of 2,265 lncRNAs, 245 miRNAs, and 5,315 mRNAs were differently expressed. Among of them, 1,136 differently expressed genes were enriched in the metabolic process. A total of 36 differently expressed genes, which were considered as differently expressed lncRNAs' targets, were enriched in the metabolic process. In addition, we also found out that eight differently expressed miRNAs could target 19 differently expressed genes. FNIP2 and PEX5L were shared in a cis-regulatory network and a differently expressed miRNA target relationship network. LncRNA-FNIP2/miR-24-3p/FNIP2 axis was considered as a potential candidate that may participate in lipid synthesis. Experimentally, the objective reality of lncRNA-FNIP2/miR-24-3p/FNIP2 axis was clarified and the regulation effect of lncRNA-FNIP2/miR-24-3p/FNIP2 axis on synthesis was validated. In brief, our study reveals a potential novel regulatory mechanism that lncRNA-FNIP2/miR-24-3p/FNIP2 axis was considered as being involved in lipid synthesis during chicken adipogenesis in liver.
Collapse
Affiliation(s)
- Lijin Guo
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaohuan Chao
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Weiling Huang
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Zhenhui Li
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Kang Luan
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mao Ye
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Siyu Zhang
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Manqing Liu
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Hongmei Li
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wen Luo
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qinghua Nie
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiquan Zhang
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qingbin Luo
- Guangdong Laboratory, Lingnan Modern Agricultural Science and Technology, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
39
|
Mathew J, Huang SC, Dumolt JH, Patel MS, Rideout TC. Maternal hypercholesterolemia programs dyslipidemia in adult male mouse progeny. Reproduction 2021; 160:1-10. [PMID: 32272447 DOI: 10.1530/rep-20-0065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/09/2020] [Indexed: 12/29/2022]
Abstract
As a collection of metabolic abnormalities including inflammation, insulin resistance, hypertension, hormone imbalance, and dyslipidemia, maternal obesity has been well-documented to program disease risk in adult offspring. Although hypercholesterolemia is strongly associated with obesity, less work has examined the programming influence of maternal hypercholesterolemia (MHC) independent of maternal obesity or high-fat feeding. This study was conducted to characterize how MHC per se impacts lipid metabolism in offspring. Female (n = 6/group) C57BL/6J mice were randomly assigned to: (1.) a standard chow diet (Control, CON) or (2.) the CON diet supplemented with exogenous cholesterol (CH) (0.15%, w/w) throughout mating and the gestation and lactation periods. At weaning (postnatal day (PND) 21) and adulthood (PND 84), male offspring were characterized for blood lipid and lipoprotein profile and hepatic lipid endpoints, namely cholesterol and triglyceride (TG) accumulation, fatty acid profile, TG production, and mRNA expression of lipid-regulatory genes. Both newly weaned and adult offspring from CH mothers demonstrated increased very low-density lipoprotein (VLDL) particle number and size and hepatic TG and n-6 polyunsaturated fatty acid accumulation. Further, adult CH offspring exhibited reduced fatty acid synthase (Fasn) and increased diglyceride acyltransferase (Dgat1) mRNA expression. These programming effects appear to be independent of changes in hepatic TG production and postprandial lipid clearance. Study results suggest that MHC, independent of obesity or high-fat feeding, can induce early changes to serum VLDL distribution and hepatic lipid profile that persist into adulthood.
Collapse
Affiliation(s)
- Joyce Mathew
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Sze-Chi Huang
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Jerad H Dumolt
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| | - Mulchand S Patel
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
40
|
Gonzalez-Soto M, Mutch DM. Diet Regulation of Long-Chain PUFA Synthesis: Role of Macronutrients, Micronutrients, and Polyphenols on Δ-5/Δ-6 Desaturases and Elongases 2/5. Adv Nutr 2021; 12:980-994. [PMID: 33186986 PMCID: PMC8166571 DOI: 10.1093/advances/nmaa142] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/04/2020] [Accepted: 10/01/2020] [Indexed: 01/08/2023] Open
Abstract
Deficiencies in the n-3 (ω-3) long-chain PUFAs (LC-PUFAs) EPA and DHA are associated with increased risk for the development of numerous diseases. Although n-3 LC-PUFAs can be obtained by consuming marine products, they are also synthesized endogenously through a biochemical pathway regulated by the Δ-5/Δ-6 desaturase and elongase 2/5 enzymes. This narrative review collates evidence from the past 40 y demonstrating that mRNA expression and activity of desaturase and elongase enzymes are influenced by numerous dietary components, including macronutrients, micronutrients, and polyphenols. Specifically, we highlight that both the quantity and the composition of dietary fats, carbohydrates, and proteins can differentially regulate desaturase pathway activity. Furthermore, desaturase and elongase mRNA levels and enzyme activities are also influenced by micronutrients (folate, vitamin B-12, vitamin A), trace minerals (iron, zinc), and polyphenols (resveratrol, isoflavones). Understanding how these various dietary components influence LC-PUFA synthesis will help further advance our understanding of how dietary patterns, ranging from caloric excesses to micronutrient deficiencies, influence disease risks.
Collapse
Affiliation(s)
- Melissa Gonzalez-Soto
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - David M Mutch
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| |
Collapse
|
41
|
Maj M, Harbottle B, Thomas PA, Hernandez GV, Smith VA, Edwards MS, Fanter RK, Glanz HS, Immoos C, Burrin DG, Santiago-Rodriguez TM, La Frano MR, Manjarín R. Consumption of High-Fructose Corn Syrup Compared with Sucrose Promotes Adiposity and Increased Triglyceridemia but Comparable NAFLD Severity in Juvenile Iberian Pigs. J Nutr 2021; 151:1139-1149. [PMID: 33693900 PMCID: PMC8112773 DOI: 10.1093/jn/nxaa441] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/11/2020] [Accepted: 12/16/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Fructose consumption has been linked to nonalcoholic fatty liver disease (NAFLD) in children. However, the effect of high-fructose corn syrup (HFCS) compared with sucrose in pediatric NAFLD has not been investigated. OBJECTIVES We tested whether the isocaloric substitution of dietary sucrose by HFCS would increase the severity of NAFLD in juvenile pigs, and whether this effect would be associated with changes in gut histology, SCFA production, and microbial diversity. METHODS Iberian pigs, 53-d-old and pair-housed in pens balanced for weight and sex, were randomly assigned to receive a mash diet top-dressed with increasing amounts of sucrose (SUC; n = 3 pens; 281.6-486.8 g/kg diet) or HFCS (n = 4; 444.3-724.8 g/kg diet) during 16 wk. Diets exceeded the animal's energy requirements by providing sugars in excess, but met the requirements for all other nutrients. Animals were killed at 165 d of age after blood sampling, and liver, muscle, and gut were collected for histology, metabolome, and microbiome analyses. Data were analyzed by multivariate and univariate statistics. RESULTS Compared with SUC, HFCS increased subcutaneous fat, triacylglycerides in plasma, and butyrate in colon (P ≤ 0.05). In addition, HFCS decreased UMP and short-chain acyl carnitines in liver, and urea nitrogen and creatinine in serum (P ≤ 0.05). Microbiome analysis showed a 24.8% average dissimilarity between HFCS and SUC associated with changes in SCFA-producing bacteria. Body weight gain, intramuscular fat, histological and serum markers of liver injury, and circulating hormones, glucose, and proinflammatory cytokines did not differ between diets. CONCLUSIONS Fructose consumption derived from HFCS promoted butyrate synthesis, triglyceridemia, and subcutaneous lipid deposition in juvenile Iberian pigs, but did not increase serum and histological markers of NAFLD compared with a sucrose-enriched diet. Longer studies could be needed to observe differences in liver injury among sugar types.
Collapse
Affiliation(s)
- Magdalena Maj
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA, USA,Center for Applications in Biotechnology, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Brooke Harbottle
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Payton A Thomas
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Gabriella V Hernandez
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Victoria A Smith
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Mark S Edwards
- Animal Science Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Rob K Fanter
- College of Agriculture, Food and Environmental Sciences, California Polytechnic State University, San Luis Obispo, CA, USA,Center for Health Research, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Hunter S Glanz
- Statistics Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Chad Immoos
- Chemistry and Biochemistry Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - Douglas G Burrin
- United States Department of Agriculture-Agricultural Research Services, Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Michael R La Frano
- Center for Health Research, California Polytechnic State University, San Luis Obispo, CA, USA,Food Science and Nutrition Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | | |
Collapse
|
42
|
Berger JM, Moon YA. Increased Hepatic Lipogenesis Elevates Liver Cholesterol Content. Mol Cells 2021; 44:116-125. [PMID: 33658436 PMCID: PMC7941001 DOI: 10.14348/molcells.2021.2147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/17/2020] [Accepted: 02/07/2021] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the most common cause of death in patients with nonalcoholic fatty liver disease (NAFLD) and dyslipidemia is considered at least partially responsible for the increased CVD risk in NAFLD patients. The aim of the present study is to understand how hepatic de novo lipogenesis influences hepatic cholesterol content as well as its effects on the plasma lipid levels. Hepatic lipogenesis was induced in mice by feeding a fat-free/high-sucrose (FF/HS) diet and the metabolic pathways associated with cholesterol were then analyzed. Both liver triglyceride and cholesterol contents were significantly increased in mice fed an FF/HS diet. Activation of fatty acid synthesis driven by the activation of sterol regulatory element binding protein (SREBP)-1c resulted in the increased liver triglycerides. The augmented cholesterol content in the liver could not be explained by an increased cholesterol synthesis, which was decreased by the FF/HS diet. HMGCoA reductase protein level was decreased in mice fed an FF/HS diet. We found that the liver retained more cholesterol through a reduced excretion of bile acids, a reduced fecal cholesterol excretion, and an increased cholesterol uptake from plasma lipoproteins. Very low-density lipoproteintriglyceride and -cholesterol secretion were increased in mice fed an FF/HS diet, which led to hypertriglyceridemia and hypercholesterolemia in Ldlr-/- mice, a model that exhibits a more human like lipoprotein profile. These findings suggest that dietary cholesterol intake and cholesterol synthesis rates cannot only explain the hypercholesterolemia associated with NAFLD, and that the control of fatty acid synthesis should be considered for the management of dyslipidemia.
Collapse
Affiliation(s)
- Jean-Mathieu Berger
- Departments of Internal Medicine and Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Young-Ah Moon
- Department of Molecular Medicine, Inha University College of Medicine, Incheon 22212, Korea
| |
Collapse
|
43
|
Roumans KH, Basset Sagarminaga J, Peters HP, Schrauwen P, Schrauwen-Hinderling VB. Liver fat storage pathways: methodologies and dietary effects. Curr Opin Lipidol 2021; 32:9-15. [PMID: 33234776 PMCID: PMC7810416 DOI: 10.1097/mol.0000000000000720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver is the result of an imbalance between lipid storage [from meal, de novo lipogenesis (DNL) and fatty acid (FA) uptake] and disposal (oxidation and VLDL output). Knowledge on the contribution of each of these pathways to liver fat content in humans is essential to develop tailored strategies to prevent and treat nonalcoholic fatty liver. Here, we review the techniques available to study the different storage pathways and review dietary modulation of these pathways. RECENT FINDINGS The type of carbohydrate and fat could be of importance in modulating DNL, as complex carbohydrates and omega-3 FAs have been shown to reduce DNL. No effects were found on the other pathways, however studies investigating this are scarce. SUMMARY Techniques used to assess storage pathways are predominantly stable isotope techniques, which require specific expertise and are costly. Validated biomarkers are often lacking. These methodological limitations also translate into a limited number of studies investigating to what extent storage pathways can be modulated by diet. Further research is needed to elucidate in more detail the impact that fat and carbohydrate type can have on liver fat storage pathways and content.
Collapse
Affiliation(s)
- Kay H.M. Roumans
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht
| | | | | | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht
| | - Vera B. Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
44
|
Goedecke JH, Chorell E, van Jaarsveld PJ, Risérus U, Olsson T. Fatty Acid Metabolism and Associations with Insulin Sensitivity Differs Between Black and White South African Women. J Clin Endocrinol Metab 2021; 106:e140-e151. [PMID: 32995848 DOI: 10.1210/clinem/dgaa696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/28/2020] [Indexed: 01/22/2023]
Abstract
PURPOSE Genetic differences in desaturase genes and consequently fatty acid metabolism have been reported. The aims were to examine ethnic differences in serum fatty acid composition and desaturase indices, and assess the ethnic-specific associations with insulin sensitivity (IS) and liver fat in black and white South African (SA) women. METHODS In this cross-sectional study including 92 premenopausal black (n = 46) and white (n = 46) SA women, serum fatty acid composition was measured in cholesteryl ester (CE) and nonesterified fatty acid (NEFA) fractions. Desaturase activities were estimated as product-to-precursor ratios: stearoyl-CoA desaturase-1 (SCD1-16, 16:1n-7/16:0); δ-5 desaturase (D5D, 20:4n-6/20:3n-6), and δ-6 desaturase (D6D, 18:3n-6/18:2n-6). Whole-body IS was estimated from an oral glucose tolerance test using the Matsuda index. In a subsample (n = 30), liver fat and hepatic IS were measured by 1H-magnetic resonance spectroscopy and hyperinsulinemic euglycemic clamp, respectively. RESULTS Despite lower whole-body IS (P = .006), black women had higher CE D5D and lower D6D and SCD1-16 indices than white women (P < .01). CE D6D index was associated with lower IS in white women only (r = -0.31, P = .045), whereas D5D index was associated with higher IS in black women only (r = 0.31, P = .041). In the subsample, D6D and SCD1-16 indices were positively and D5D was negatively associated with liver fat (P < .05). Conversely, CE SCD1-16 was negatively associated with hepatic IS (P < .05), but not independently of liver fat. CONCLUSIONS Ethnic differences in fatty acid-derived desaturation indices were observed, with insulin-resistant black SA women paradoxically showing a fatty acid pattern typical for higher insulin sensitivity in European populations.
Collapse
Affiliation(s)
- Julia H Goedecke
- Non-communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Elin Chorell
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Paul J van Jaarsveld
- Non-communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Tommy Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
45
|
Miyamoto JÉ, Reginato A, Portovedo M, Dos Santos RM, Stahl MA, Le Stunff H, Latorraca MQ, de Barros Reis MA, Arantes VC, Doneda DL, Ignacio-Souza LM, Torsoni AS, Grimaldi R, Ribeiro APB, Torsoni MA, Milanski M. Interesterified palm oil impairs glucose homeostasis and induces deleterious effects in liver of Swiss mice. Metabolism 2020; 112:154350. [PMID: 32910938 DOI: 10.1016/j.metabol.2020.154350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/30/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Interesterified fats have largely replaced the partially hydrogenated oils which are the main dietary source of trans fat in industrialized food. This process promotes a random rearrangement of the native fatty acids and the results are different triacylglycerol (TAG) molecules without generating trans isomers. The role of interesterified fats in metabolism remains unclear. We evaluated metabolic parameters, glucose homeostasis and inflammatory markers in mice fed with normocaloric and normolipidic diets or hypercaloric and high-fat diet enriched with interesterified palm oil. METHODS Male Swiss mice were randomly divided into four experimental groups and submitted to either normolipidic palm oil diet (PO), normolipidic interesterified palm oil diet (IPO), palm oil high-fat diet (POHF) or interesterified palm oil high-fat diet (IPOHF) during an 8 weeks period. RESULTS When compared to the PO group, IPO group presented higher body mass, hyperglycemia, impaired glucose tolerance, evidence of insulin resistance and greater production of glucose in basal state during pyruvate in situ assay. We also observed higher protein content of hepatic PEPCK and increased cytokine mRNA expression in the IPO group when compared to PO. Interestingly, IPO group showed similar parameters to POHF and IPOHF groups. CONCLUSION The results indicate that substitution of palm oil for interesterified palm oil even on normocaloric and normolipidic diet could negatively modulate metabolic parameters and glucose homeostasis as well as cytokine gene expression in the liver and white adipose tissue. This data support concerns about the effects of interesterified fats on health and could promote further discussions about the safety of the utilization of this unnatural fat by food industry.
Collapse
Affiliation(s)
- Josiane Érica Miyamoto
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Andressa Reginato
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Mariana Portovedo
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Raísa Magno Dos Santos
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Hervé Le Stunff
- Paris-Saclay Institute of Neuroscience, CNRS UMR 9197, Université Paris-Sud, University Paris Saclay, Orsay, France
| | | | | | | | - Diego Luiz Doneda
- Physiology Laboratory, Department of Basic Health Sciences, Federal University of Mato Grosso, Cuiabá, Brazil
| | - Leticia Martins Ignacio-Souza
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Adriana Souza Torsoni
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Renato Grimaldi
- School of Food Engineering, University of Campinas, UNICAMP, Campinas, Brazil
| | | | - Marcio Alberto Torsoni
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil
| | - Marciane Milanski
- School of Applied Sciences, University of Campinas, UNICAMP, Limeira, Brazil; Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Brazil.
| |
Collapse
|
46
|
Weir NL, Steffen BT, Guan W, Johnson LM, Djousse L, Mukamal KJ, Tsai MY. Circulating omega-7 fatty acids are differentially related to metabolic dysfunction and incident type II diabetes: The Multi-Ethnic Study of Atherosclerosis (MESA). DIABETES & METABOLISM 2020; 46:319-325. [PMID: 31706030 PMCID: PMC7200281 DOI: 10.1016/j.diabet.2019.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 12/16/2022]
Abstract
AIM Determine whether plasma omega-7 vaccenic acid and palmitoleic acid levels are related to homeostasis model of insulin resistance scores and incident type II diabetes, and whether race/ethnicity modifies these associations. METHODS Plasma phospholipid fatty acids were measured by gas chromatography with flame-ionization detection in Multi-Ethnic Study of Atherosclerosis participants. Linear regression determined associations of vaccenic acid and palmitoleic acid with log-transformed homeostasis model of insulin resistance scores (n=5689), and Cox regression determined associations with incident type II diabetes (n=5413, 660 cases). Race-interactions were tested. RESULTS Adjusting for typical risk factors, higher levels of plasma vaccenic acid were found to be inversely associated with insulin resistance scores across all four race/ethnicities, and a significant race-interaction was observed between Hispanics and Caucasians (P for interaction=0.03). Vaccenic acid was related to 17%, 32%, and 39% lower risks of incident type II diabetes in Black, Hispanic, and Chinese American participants, respectively. Differences in associations between races were detected (P for interactions<0.05). By contrast, higher levels of plasma palmitoleic acid were related to greater insulin resistance scores in Blacks (P<0.001) and Hispanics (P<0.001); significant race-based differences between associations were detected (P for interactions<0.05). Palmitoleic acid was correspondingly related to a 21% greater risk of incident type II diabetes in Black individuals. CONCLUSIONS Results suggest that plasma vaccenic acid and palmitoleic acid are markers of metabolic health and dysfunction, respectively. Coupled with previous evidence and the significant race-interactions, our findings have implications for future studies of the race-based differences in omega-7 fatty acids and their regulation in the context of deteriorating metabolic health.
Collapse
Affiliation(s)
- Natalie L. Weir
- University of Minnesota, Department of Laboratory Medicine & Pathology, Minneapolis, MN 55455
| | - Brian T. Steffen
- University of Minnesota, Department of Laboratory Medicine & Pathology, Minneapolis, MN 55455
| | - Weihua Guan
- University of Minnesota, School of Public Health Biostatistics Division, Minneapolis, MN 55455
| | - Lisa M. Johnson
- University of Utah, Department of Pathology, Salt Lake City, UT 84108
| | - Luc Djousse
- Brigham and Women’s Hospital, Department of Medicine Division of Aging, Boston, MA 02115
| | - Kenneth J. Mukamal
- Beth Israel Deaconess Medical Center, General Medicine, Brookline, MA 02446
| | - Michael Y. Tsai
- University of Minnesota, Department of Laboratory Medicine & Pathology, Minneapolis, MN 55455
| |
Collapse
|
47
|
Podszun MC, Alawad AS, Lingala S, Morris N, Huang WCA, Yang S, Schoenfeld M, Rolt A, Ouwerkerk R, Valdez K, Umarova R, Ma Y, Fatima SZ, Lin DD, Mahajan LS, Samala N, Violet PC, Levine M, Shamburek R, Gharib AM, Kleiner DE, Garraffo HM, Cai H, Walter PJ, Rotman Y. Vitamin E treatment in NAFLD patients demonstrates that oxidative stress drives steatosis through upregulation of de-novo lipogenesis. Redox Biol 2020; 37:101710. [PMID: 32920226 PMCID: PMC7494510 DOI: 10.1016/j.redox.2020.101710] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/23/2020] [Accepted: 08/27/2020] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress (OS) in non-alcoholic fatty liver disease (NAFLD) promotes liver injury and inflammation. Treatment with vitamin E (α-tocopherol, αT), a lipid-soluble antioxidant, improves liver injury but also decreases steatosis, thought to be upstream of OS, through an unknown mechanism. To elucidate the mechanism, we combined a mechanistic human trial interrogating pathways of intrahepatic triglyceride (IHTG) accumulation and in vitro experiments. 50% of NAFLD patients (n = 20) treated with αT (200-800 IU/d) for 24 weeks had a ≥ 25% relative decrease in IHTG by magnetic resonance spectroscopy. Paired liver biopsies at baseline and week 4 of treatment revealed a decrease in markers of hepatic de novo lipogenesis (DNL) that strongly predicted week 24 response. In vitro, using HepG2 cells and primary human hepatocytes, αT inhibited glucose-induced DNL by decreasing SREBP-1 processing and lipogenic gene expression. This mechanism is dependent on the antioxidant capacity of αT, as redox-silenced methoxy-αT is unable to inhibit DNL in vitro. OS by itself was sufficient to increase S2P expression in vitro, and S2P is upregulated in NAFLD livers. In summary, we utilized αT to demonstrate a vicious cycle in which NAFLD generates OS, which feeds back to augment DNL and increases steatosis. Clinicaltrials.gov: NCT01792115.
Collapse
Affiliation(s)
- Maren C Podszun
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ahmad S Alawad
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shilpa Lingala
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nevitt Morris
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wen-Chun A Huang
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- Nutrition Department, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Megan Schoenfeld
- Nutrition Department, NIH Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Adam Rolt
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ronald Ouwerkerk
- Biomedical and Metabolic Imaging Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kristin Valdez
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Regina Umarova
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yanling Ma
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Syeda Zaheen Fatima
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dennis D Lin
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lakshmi S Mahajan
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Niharika Samala
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pierre-Christian Violet
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert Shamburek
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ahmed M Gharib
- Biomedical and Metabolic Imaging Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - H Martin Garraffo
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hongyi Cai
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter J Walter
- Clinical Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yaron Rotman
- Liver and Energy Metabolism Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
48
|
Fatty Acid Synthase: An Emerging Target in Cancer. Molecules 2020; 25:molecules25173935. [PMID: 32872164 PMCID: PMC7504791 DOI: 10.3390/molecules25173935] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
In recent years, lipid metabolism has garnered significant attention as it provides the necessary building blocks required to sustain tumor growth and serves as an alternative fuel source for ATP generation. Fatty acid synthase (FASN) functions as a central regulator of lipid metabolism and plays a critical role in the growth and survival of tumors with lipogenic phenotypes. Accumulating evidence has shown that it is capable of rewiring tumor cells for greater energy flexibility to attain their high energy requirements. This multi-enzyme protein is capable of modulating the function of subcellular organelles for optimal function under different conditions. Apart from lipid metabolism, FASN has functional roles in other cellular processes such as glycolysis and amino acid metabolism. These pivotal roles of FASN in lipid metabolism make it an attractive target in the clinic with several new inhibitors currently being tested in early clinical trials. This article aims to present the current evidence on the emergence of FASN as a target in human malignancies.
Collapse
|
49
|
Lai M, Al Rijjal D, Röst HL, Dai FF, Gunderson EP, Wheeler MB. Underlying dyslipidemia postpartum in women with a recent GDM pregnancy who develop type 2 diabetes. eLife 2020; 9:59153. [PMID: 32748787 PMCID: PMC7417169 DOI: 10.7554/elife.59153] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/18/2020] [Indexed: 12/15/2022] Open
Abstract
Approximately, 35% of women with Gestational Diabetes (GDM) progress to Type 2 Diabetes (T2D) within 10 years. However, links between GDM and T2D are not well understood. We used a well-characterised GDM prospective cohort of 1035 women following up to 8 years postpartum. Lipidomics profiling covering >1000 lipids was performed on fasting plasma samples from participants 6–9 week postpartum (171 incident T2D vs. 179 controls). We discovered 311 lipids positively and 70 lipids negatively associated with T2D risk. The upregulation of glycerolipid metabolism involving triacylglycerol and diacylglycerol biosynthesis suggested activated lipid storage before diabetes onset. In contrast, decreased sphingomyelines, hexosylceramide and lactosylceramide indicated impaired sphingolipid metabolism. Additionally, a lipid signature was identified to effectively predict future diabetes risk. These findings demonstrate an underlying dyslipidemia during the early postpartum in those GDM women who progress to T2D and suggest endogenous lipogenesis may be a driving force for future diabetes onset.
Collapse
Affiliation(s)
- Mi Lai
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Dana Al Rijjal
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Hannes L Röst
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Ontario, Canada
| | - Feihan F Dai
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Erica P Gunderson
- Kaiser Permanente Northern California, Division of Research, Oakland, United States
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada.,Advanced Diagnostics, Metabolism, Toronto General Research Institute, Ontario, Canada
| |
Collapse
|
50
|
Fretts AM, Jensen PN, Hoofnagle A, McKnight B, Howard BV, Umans J, Yu C, Sitlani C, Siscovick DS, King IB, Sotoodehnia N, Lemaitre RN. Plasma Ceramide Species Are Associated with Diabetes Risk in Participants of the Strong Heart Study. J Nutr 2020; 150:1214-1222. [PMID: 31665380 PMCID: PMC7198314 DOI: 10.1093/jn/nxz259] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/09/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Few studies have assessed the associations of ceramides and sphingomyelins (SMs) with diabetes in humans. OBJECTIVE We assessed associations of 15 circulating ceramides and SM species with incident diabetes in 2 studies. METHODS The analysis included 435 American-Indian participants from the Strong Heart Study (nested case-control design for analyses; mean age: 57 y; 34% male; median time until diabetes 4.3 y for cases) and 1902 participants from the Strong Heart Family Study (prospective design for analyses; mean age: 37 y; 39% male; median 12.5 y of follow-up). Sphingolipid species were measured using stored plasma samples by sequential LC and MS. Using logistic regression and parametric survival models within studies, and an inverse-variance-weighted meta-analysis across studies, we examined associations of 15 ceramides and SM species with incident diabetes. RESULTS There were 446 cases of incident diabetes across the studies. Higher circulating concentrations of ceramides containing stearic acid (Cer-18), arachidic acid (Cer-20), and behenic acid (Cer-22) were each associated with a higher risk of diabetes. The RRs for incident diabetes per 1 SD of each log ceramide species (μM) were 1.22 (95% CI: 1.09, 1.37) for Cer-18, 1.18 (95% CI: 1.06, 1.31) for Cer-20, and 1.20 (95% CI: 1.08, 1.32) for Cer-22. Although the magnitude of the risk estimates for the association of ceramides containing lignoceric acid (Cer-24) with diabetes was similar to those for Cer-18, Cer-20, and Cer-22 (RR = 1.13; 95% CI: 1.01, 1.26), the association was not statistically significant after correction for multiple testing (P = 0.007). Ceramides carrying palmitic acid (Cer-16), SMs, glucosyl-ceramides, or a lactosyl-ceramide were not associated with diabetes risk. CONCLUSIONS Higher concentrations of circulating Cer-18, Cer-20, and Cer-22 were associated with a higher risk of developing diabetes in 2 studies of American-Indian adults. This trial was registered at clinicaltrials.gov as NCT00005134.
Collapse
Affiliation(s)
- Amanda M Fretts
- Department of Epidemiology, University of Washington, Seattle, WA, USA,Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Address correspondence to AMF (e-mail: )
| | - Paul N Jensen
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew Hoofnagle
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Barbara McKnight
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Barbara V Howard
- MedStar Health Research Institute, Hyattsville, MD, USA,Georgetown and Howard Universities Center for Translational Science, Washington, DC, USA
| | - Jason Umans
- MedStar Health Research Institute, Hyattsville, MD, USA
| | - Chaoyu Yu
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Colleen Sitlani
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Irena B King
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|