26 results on '"Panu K, Luukkonen"'
Search Results
2. Overfeeding Saturated Fat Increases LDL (Low-Density Lipoprotein) Aggregation Susceptibility While Overfeeding Unsaturated Fat Decreases Proteoglycan-Binding of Lipoproteins
- Author
-
Mari Lahelma, Martina B. Lorey, Leanne Hodson, Katariina Öörni, Panu K. Luukkonen, Hannele Yki-Järvinen, Sami Qadri, Petri T. Kovanen, Sanja Sädevirta, Tuulia Hyötyläinen, Maija Ruuth, Medicum, Research Programs Unit, University of Helsinki, HUS Internal Medicine and Rehabilitation, Department of Medicine, Clinicum, Doctoral Programme in Clinical Research, Molecular and Integrative Biosciences Research Programme, and Biosciences
- Subjects
proteoglykaanit ,Male ,intervention study ,Saturated fat ,030204 cardiovascular system & hematology ,chemistry.chemical_compound ,0302 clinical medicine ,Tandem Mass Spectrometry ,2. Zero hunger ,0303 health sciences ,rasvat ,sfingomyeliini ,aggregation ,aggregaatio ,ruokavalio ,LDL - Low density lipoprotein ,interventiotutkimus ,Middle Aged ,metabolomics ,3. Good health ,Lipoproteins, LDL ,nutrition ,Low-density lipoprotein ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Female ,Proteoglycans ,lipids (amino acids, peptides, and proteins) ,3143 Nutrition ,Cardiology and Cardiovascular Medicine ,Sphingomyelin ,Protein Binding ,Adult ,ateroskleroosi ,Spectrometry, Mass, Electrospray Ionization ,medicine.medical_specialty ,lipoproteiinit ,Diet, High-Fat ,Independent predictor ,LDL ,sphingomyelin ,ravinto ,Protein Aggregates ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,metabolomiikka ,Nuclear Magnetic Resonance, Biomolecular ,030304 developmental biology ,Proteoglycan binding ,Unsaturated fat ,dietary fats ,Fats, Unsaturated ,lipoproteins ,Endocrinology ,low-density lipoprotein ,chemistry ,3121 General medicine, internal medicine and other clinical medicine ,atherosclerosis ,diet ,Clinical and Population Studies ,Chromatography, Liquid ,Lipoprotein - Abstract
Supplemental Digital Content is available in the text., Objective: We recently showed that measurement of the susceptibility of LDL (low-density lipoprotein) to aggregation is an independent predictor of cardiovascular events. We now wished to compare effects of overfeeding different dietary macronutrients on LDL aggregation, proteoglycan-binding of plasma lipoproteins, and on the concentration of oxidized LDL in plasma, 3 in vitro parameters consistent with increased atherogenicity. Approach and Results: The participants (36 subjects; age, 48±10 years; body mass index, 30.9±6.2 kg/m2) were randomized to consume an extra 1000 kcal/day of either unsaturated fat, saturated fat, or simple sugars (CARB) for 3 weeks. We measured plasma proatherogenic properties (susceptibility of LDL to aggregation, proteoglycan-binding, oxidized LDL) and concentrations and composition of plasma lipoproteins using nuclear magnetic resonance spectroscopy, and in LDL using liquid chromatography mass spectrometry, before and after the overfeeding diets. LDL aggregation increased in the saturated fat but not the other groups. This change was associated with increased sphingolipid and saturated triacylglycerols in LDL and in plasma and reduction of clusterin on LDL particles. Proteoglycan binding of plasma lipoproteins decreased in the unsaturated fat group relative to the baseline diet. Lipoprotein properties remained unchanged in the CARB group. Conclusions: The type of fat during 3 weeks of overfeeding is an important determinant of the characteristics and functional properties of plasma lipoproteins in humans. Registration: URL: http://www.clinicaltrials.gov; Unique identifier NCT02133144.
- Published
- 2021
- Full Text
- View/download PDF
3. LPIAT1/MBOAT7 depletion increases triglyceride synthesis fueled by high phosphatidylinositol turnover
- Author
-
Takuya Kubo, Toshimasa Yamauchi, Naoto Kubota, Panu K. Luukkonen, Yuki Tanaka, Yanli Mao, Jussi Pihlajamäki, Luca Valenti, Yuta Shimanaka, Andrea Caddeo, Rosellina Margherita Mancina, Hiroyuki Arai, Nozomu Kono, Guido Baselli, Tetsuya Kubota, Hannele Yki-Järvinen, Stefano Romeo, HUS Internal Medicine and Rehabilitation, Department of Medicine, Department of Biochemistry and Developmental Biology, University of Helsinki, and Helsinki University Hospital Area
- Subjects
VARIANT ,Cell Culture Techniques ,Phosphatidylinositols ,GLUCOSE ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,hepatic fibrosis ,INOSITOL PHOSPHATES ,Mice, Knockout ,INSULIN-RESISTANCE ,0303 health sciences ,Chemistry ,Fatty liver ,Gastroenterology ,3. Good health ,ACID ,030211 gastroenterology & hepatology ,Arachidonic acid ,medicine.medical_specialty ,MBOAT7 ,lipids ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Humans ,Phosphatidylinositol ,DE-NOVO LIPOGENESIS ,Triglycerides ,FATTY LIVER-DISEASE ,fatty liver ,030304 developmental biology ,Hepatology ,Triglyceride ,Membrane Proteins ,Lipid metabolism ,Lipid Metabolism ,medicine.disease ,GENE ,INDIVIDUALS ,Disease Models, Animal ,Endocrinology ,3121 General medicine, internal medicine and other clinical medicine ,Hepatocytes ,Hepatic stellate cell ,Steatosis ,Hepatic fibrosis ,Acyltransferases - Abstract
ObjectiveNon-alcoholic fatty liver disease (NAFLD) is a common prelude to cirrhosis and hepatocellular carcinoma. The genetic rs641738 C>T variant in the lysophosphatidylinositol acyltransferase 1 (LPIAT1)/membrane bound O-acyltransferase domain-containing 7, which incorporates arachidonic acid into phosphatidylinositol (PI), is associated with the entire spectrum of NAFLD. In this study, we investigated the mechanism underlying this association in mice and cultured human hepatocytes.DesignWe generated the hepatocyte-specific Lpiat1 knockout mice to investigate the function of Lpiat1 in vivo. We also depleted LPIAT1 in cultured human hepatic cells using CRISPR-Cas9 systems or siRNA. The effect of LPIAT1-depletion on liver fibrosis was examined in mice fed high fat diet and in liver spheroids. Lipid species were measured using liquid chromatography-electrospray ionisation mass spectrometry. Lipid metabolism was analysed using radiolabeled glycerol or fatty acids.ResultsThe hepatocyte-specific Lpiat1 knockout mice developed hepatic steatosis spontaneously, and hepatic fibrosis on high fat diet feeding. Depletion of LPIAT1 in cultured hepatic cells and in spheroids caused triglyceride accumulation and collagen deposition. The increase in hepatocyte fat content was due to a higher triglyceride synthesis fueled by a non-canonical pathway. Indeed, reduction in the PI acyl chain remodelling caused a high PI turnover, by stimulating at the same time PI synthesis and breakdown. The degradation of PI was mediated by a phospholipase C, which produces diacylglycerol, a precursor of triglyceride.ConclusionWe found a novel pathway fueling triglyceride synthesis in hepatocytes, by a direct metabolic flow of PI into triglycerides. Our findings provide an insight into the pathogenesis and therapeutics of NAFLD.
- Published
- 2020
- Full Text
- View/download PDF
4. Effect of a ketogenic diet on hepatic steatosis and hepatic mitochondrial metabolism in nonalcoholic fatty liver disease
- Author
-
Gary W. Cline, Antti Hakkarainen, Hannele Yki-Järvinen, Xian-Man Zhang, Gerald I. Shulman, Panu K. Luukkonen, Kun Lyu, Sylvie Dufour, Kitt Falk Petersen, Tiina Lehtimäki, HUS Internal Medicine and Rehabilitation, Department of Medicine, University of Helsinki, HUS Medical Imaging Center, Department of Diagnostics and Therapeutics, Helsinki University Hospital Area, Yale University, Department of Neuroscience and Biomedical Engineering, Minerva Foundation Institute for Medical Research Helsinki, Aalto-yliopisto, and Aalto University
- Subjects
Male ,0301 basic medicine ,Citrate synthase ,Medical Sciences ,medicine.medical_treatment ,Fatty Acids, Nonesterified ,Lipoproteins, VLDL ,pyruvate carboxylase ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,insulin resistance ,Ketogenesis ,Nonalcoholic fatty liver disease ,Insulin ,INSULIN-RESISTANCE ,SPECTROSCOPY ,Multidisciplinary ,biology ,Chemistry ,Fatty Acids ,Middle Aged ,Biological Sciences ,LOW-CARBOHYDRATE DIET ,Mitochondria ,3. Good health ,ADIPOSE-TISSUE ,Liver ,redox ,Lipogenesis ,Body Composition ,Female ,Diet, Ketogenic ,Oxidation-Reduction ,medicine.medical_specialty ,WEIGHT-LOSS ,030209 endocrinology & metabolism ,Citrate (si)-Synthase ,Redox ,Carbohydrate restriction ,03 medical and health sciences ,NEFA ,Insulin resistance ,Internal medicine ,medicine ,Humans ,Obesity ,CYCLE ,Triglycerides ,DE-NOVO LIPOGENESIS ,Pyruvate carboxylase ,REDOX STATE ,TRIGLYCERIDE CONTENT ,Overweight ,ACIDS ,medicine.disease ,carbohydrate restriction ,Fatty Liver ,030104 developmental biology ,Endocrinology ,biology.protein ,3111 Biomedicine ,Steatosis ,citrate synthase ,Ketogenic diet - Abstract
Significance Ketogenic diet is an effective treatment for nonalcoholic fatty liver disease (NAFLD). Here, we present evidence that hepatic mitochondrial fluxes and redox state are markedly altered during ketogenic diet-induced reversal of NAFLD in humans. Ketogenic diet for 6 d markedly decreased liver fat content and hepatic insulin resistance. These changes were associated with increased net hydrolysis of liver triglycerides and decreased endogenous glucose production and serum insulin concentrations. Partitioning of fatty acids toward ketogenesis increased, which was associated with increased hepatic mitochondrial redox state and decreased hepatic citrate synthase flux. These data demonstrate heretofore undescribed adaptations underlying the reversal of NAFLD by ketogenic diet and highlight hepatic mitochondrial fluxes and redox state as potential treatment targets in NAFLD., Weight loss by ketogenic diet (KD) has gained popularity in management of nonalcoholic fatty liver disease (NAFLD). KD rapidly reverses NAFLD and insulin resistance despite increasing circulating nonesterified fatty acids (NEFA), the main substrate for synthesis of intrahepatic triglycerides (IHTG). To explore the underlying mechanism, we quantified hepatic mitochondrial fluxes and their regulators in humans by using positional isotopomer NMR tracer analysis. Ten overweight/obese subjects received stable isotope infusions of: [D7]glucose, [13C4]β-hydroxybutyrate and [3-13C]lactate before and after a 6-d KD. IHTG was determined by proton magnetic resonance spectroscopy (1H-MRS). The KD diet decreased IHTG by 31% in the face of a 3% decrease in body weight and decreased hepatic insulin resistance (−58%) despite an increase in NEFA concentrations (+35%). These changes were attributed to increased net hydrolysis of IHTG and partitioning of the resulting fatty acids toward ketogenesis (+232%) due to reductions in serum insulin concentrations (−53%) and hepatic citrate synthase flux (−38%), respectively. The former was attributed to decreased hepatic insulin resistance and the latter to increased hepatic mitochondrial redox state (+167%) and decreased plasma leptin (−45%) and triiodothyronine (−21%) concentrations. These data demonstrate heretofore undescribed adaptations underlying the reversal of NAFLD by KD: That is, markedly altered hepatic mitochondrial fluxes and redox state to promote ketogenesis rather than synthesis of IHTG.
- Published
- 2020
- Full Text
- View/download PDF
5. Distinct contributions of metabolic dysfunction and genetic risk factors in the pathogenesis of non-alcoholic fatty liver disease
- Author
-
Kimmo Porthan, Hannele Yki-Järvinen, Marju Orho-Melander, Panu K. Luukkonen, Sami Qadri, Anne K. Penttilä, Amalia Gastaldelli, Jussi Pihlajamäki, Henna Sammalkorpi, Anne Juuti, Ville Männistö, Melania Gaggini, Johanna Arola, A. Hakkarainen, Mika Ala-Korpela, Noora Ahlholm, Leanne Hodson, Tiina Lehtimäki, Yale University, University of Helsinki, University of Eastern Finland, Department of Neuroscience and Biomedical Engineering, National Research Council of Italy, University of Oulu, Lund University, University of Oxford, Aalto-yliopisto, Aalto University, Clinicum, HUS Internal Medicine and Rehabilitation, Department of Medicine, Kardiologian yksikkö, HUS Heart and Lung Center, HUS Abdominal Center, II kirurgian klinikka, Department of Surgery, HUS Medical Imaging Center, Department of Diagnostics and Therapeutics, Department of Pathology, and HUSLAB
- Subjects
Male ,Hepatic mitochondrial redox state ,Biopsy ,Mboat7 ,Pnpla3 ,Hsd17b13 ,chemistry.chemical_compound ,0302 clinical medicine ,High-density lipoprotein ,Non-alcoholic Fatty Liver Disease ,Risk Factors ,insulin resistance ,Marc1 ,Finland ,De novo lipogenesis ,0303 health sciences ,Fatty liver ,Middle Aged ,metabolomics ,Adipose tissue lipolysis ,3. Good health ,Liver ,Lipogenesis ,Female ,030211 gastroenterology & hepatology ,adipose tissue lipolysis ,Adult ,medicine.medical_specialty ,TM6SF2 ,HSD17B13 ,MBOAT7 ,03 medical and health sciences ,NEFA ,Insulin resistance ,Metabolic Diseases ,Internal medicine ,medicine ,Humans ,Metabolomics ,Obesity ,PNPLA3 ,030304 developmental biology ,Tm6sf2 ,Hepatology ,Triglyceride ,hepatic mitochondrial redox state ,MARC1 ,medicine.disease ,Mitochondrial amidoxime reducing component 1 ,Endocrinology ,de novo lipogenesis ,chemistry ,3121 General medicine, internal medicine and other clinical medicine - Abstract
Funding Information: This study was supported by Academy of Finland Grant 309263 (H.Y.-J.) and Grant 138006 (J.P.), EU H2020 project ?Elucidating Pathways of Steatohepatitis? EPoS Grant 634413 (H.Y.-J.), H2020-JTI-IMI2 EU project 777377-2 Liver Investigation: Testing Marker Utility in Steatohepatitis (LITMUS) (H.Y.-J.), Government Funding (H.Y.-J.), Novo Nordisk Foundation (H.Y.-J., P.K.L., M.A.-K.), Ralph Gr?sbeck Scholarship of the Minerva Foundation (P.K.L.), Novo Nordisk Foundation (P.K.L.), Juho Vainio Foundation (J.P.), Finnish Medical Foundation (V.M.), British Heart Foundation Senior Research Fellowship in Basic Science (FS/15/56/31645) (L.H.) and Kuopio University Hospital Project grant (J.P., EVO/VTR grants 2005-2019). Funding Information: This study was supported by Academy of Finland Grant 309263 (H.Y.-J.) and Grant 138006 (J.P.), EU H2020 project ‘Elucidating Pathways of Steatohepatitis’ EPoS Grant 634413 (H.Y.-J.), H2020-JTI-IMI2 EU project 777377-2 Liver Investigation: Testing Marker Utility in Steatohepatitis (LITMUS) (H.Y.-J.), Government Funding (H.Y.-J.), Novo Nordisk Foundation (H.Y.-J., P.K.L., M.A.-K.), Ralph Gräsbeck Scholarship of the Minerva Foundation (P.K.L.), Novo Nordisk Foundation (P.K.L.), Juho Vainio Foundation (J.P.), Finnish Medical Foundation (V.M.), British Heart Foundation Senior Research Fellowship in Basic Science (FS/15/56/31645) (L.H.) and Kuopio University Hospital Project grant (J.P., EVO/VTR grants 2005-2019). Publisher Copyright: © 2021 The Author(s) Background & Aims: There is substantial inter-individual variability in the risk of non-alcoholic fatty liver disease (NAFLD). Part of which is explained by insulin resistance (IR) (‘MetComp’) and part by common modifiers of genetic risk (‘GenComp’). We examined how IR on the one hand and genetic risk on the other contribute to the pathogenesis of NAFLD. Methods: We studied 846 individuals: 492 were obese patients with liver histology and 354 were individuals who underwent intrahepatic triglyceride measurement by proton magnetic resonance spectroscopy. A genetic risk score was calculated using the number of risk alleles in PNPLA3, TM6SF2, MBOAT7, HSD17B13 and MARC1. Substrate concentrations were assessed by serum NMR metabolomics. In subsets of participants, non-esterified fatty acids (NEFAs) and their flux were assessed by D5-glycerol and hyperinsulinemic-euglycemic clamp (n = 41), and hepatic de novo lipogenesis (DNL) was measured by D2O (n = 61). Results: We found that substrate surplus (increased concentrations of 28 serum metabolites including glucose, glycolytic intermediates, and amino acids; increased NEFAs and their flux; increased DNL) characterized the ‘MetComp’. In contrast, the ‘GenComp’ was not accompanied by any substrate excess but was characterized by an increased hepatic mitochondrial redox state, as determined by serum β-hydroxybutyrate/acetoacetate ratio, and inhibition of hepatic pathways dependent on tricarboxylic acid cycle activity, such as DNL. Serum β-hydroxybutyrate/acetoacetate ratio correlated strongly with all histological features of NAFLD. IR and hepatic mitochondrial redox state conferred additive increases in histological features of NAFLD. Conclusions: These data show that the mechanisms underlying ‘Metabolic’ and ‘Genetic’ components of NAFLD are fundamentally different. These findings may have implications with respect to the diagnosis and treatment of NAFLD. Lay summary: The pathogenesis of non-alcoholic fatty liver disease can be explained in part by a metabolic component, including obesity, and in part by a genetic component. Herein, we demonstrate that the mechanisms underlying these components are fundamentally different: the metabolic component is characterized by hepatic oversupply of substrates, such as sugars, lipids and amino acids. In contrast, the genetic component is characterized by impaired hepatic mitochondrial function, making the liver less able to metabolize these substrates.
- Published
- 2022
6. Exposure to environmental contaminants is associated with altered hepatic lipid metabolism in non-alcoholic fatty liver disease
- Author
-
Thomas F. Webster, Sirkku Jäntti, Panu K. Luukkonen, Hannele Yki-Järvinen, Sami Qadri, Jennifer J. Schlezinger, Partho Sen, Tuulia Hyötyläinen, Oddny Ragnarsdottir, Johanna Arola, Matej Orešič, Aidan McGlinchey, Anne Juuti, Department of Medicine, HUS Internal Medicine and Rehabilitation, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, HUS Abdominal Center, II kirurgian klinikka, Department of Pathology, and HUSLAB
- Subjects
Male ,Fatty Acids, Nonesterified ,SERUM ,Cohort Studies ,STEATOHEPATITIS ,Mice ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,Fibrosis ,Amino Acids ,0303 health sciences ,INSULIN-RESISTANCE ,Bile acid ,Fatty liver ,PFOA ,Middle Aged ,Lipidome ,3. Good health ,Female ,metabolome ,non-alcoholic steatohepatitis ,Adult ,EXPRESSION ,medicine.medical_specialty ,SEX-DIFFERENCES ,medicine.drug_class ,Carbohydrate metabolism ,exposome ,03 medical and health sciences ,lipidome ,Internal medicine ,medicine ,Metabolome ,Animals ,Humans ,bile acid ,NHANES ,chemical exposure ,030304 developmental biology ,Hepatology ,business.industry ,fibrosis ,Lipid metabolism ,Environmental Exposure ,Metabolism ,metabolic pathway ,Lipid Metabolism ,medicine.disease ,Disease Models, Animal ,perfluorinated alkyl substance ,Endocrinology ,3121 General medicine, internal medicine and other clinical medicine ,business ,030217 neurology & neurosurgery ,BILE-ACID - Abstract
Background & aims: Recent experimental models and epidemiological studies suggest that specific environmental contaminants (ECs) contribute to the initiation and pathology of nonalcoholic fatty liver disease (NAFLD). However, the underlying mechanisms linking EC exposure with NAFLD remain poorly understood and there is no data on their impact on the human liver metabolome. Herein, we hypothesized that exposure to ECs, particularly perfluorinated alkyl substances (PFAS), impacts liver metabolism, specifically bile acid metabolism. Methods: In a well-characterized human NAFLD cohort of 105 individuals, we investigated the effects of EC exposure on liver metabolism. We characterized the liver (via biopsy) and circulating metabolomes using 4 mass spectrometry-based analytical platforms, and measured PFAS and other ECs in serum. We subsequently compared these results with an exposure study in a PPARa-humanized mouse model. Results: PFAS exposure appears associated with perturbation of key hepatic metabolic pathways previously found altered in NAFLD, particularly those related to bile acid and lipid metabolism. We identified stronger associations between the liver metabolome, chemical exposure and NAFLD-associated clinical variables (liver fat content, HOMA-IR), in females than males. Specifically, we observed PFAS-associated upregulation of bile acids, triacylglycerols and ceramides, and association between chemical exposure and dysregulated glucose metabolism in females. The murine exposure study further corroborated our findings, vis-a-vis a sex-specific association between PFAS exposure and NAFLD-associated lipid changes. Conclusions: Females may be more sensitive to the harmful impacts of PFAS. Lipid-related changes subsequent to PFAS exposure may be secondary to the interplay between PFAS and bile acid metabolism. Lay summary: There is increasing evidence that specific environmental contaminants, such as perfluorinated alkyl substances (PFAS), contribute to the progression of non-alcoholic fatty liver disease (NAFLD). However, it is poorly understood how these chemicals impact human liver metabolism. Here we show that human exposure to PFAS impacts metabolic processes associated with NAFLD, and that the effect is different in females and males. (C) 2021 The Author(s). Published by Elsevier B.V. on behalf of European Association for the Study of the Liver.
- Published
- 2022
7. PSD3 downregulation confers protection against fatty liver disease
- Author
-
Rosellina M. Mancina, Kavitha Sasidharan, Anna Lindblom, Ying Wei, Ester Ciociola, Oveis Jamialahmadi, Piero Pingitore, Anne-Christine Andréasson, Giovanni Pellegrini, Guido Baselli, Ville Männistö, Jussi Pihlajamäki, Vesa Kärjä, Stefania Grimaudo, Ilaria Marini, Marco Maggioni, Barbara Becattini, Federica Tavaglione, Carly Dix, Marie Castaldo, Stephanie Klein, Mark Perelis, Francois Pattou, Dorothée Thuillier, Violeta Raverdy, Paola Dongiovanni, Anna Ludovica Fracanzani, Felix Stickel, Jochen Hampe, Stephan Buch, Panu K. Luukkonen, Daniele Prati, Hannele Yki-Järvinen, Salvatore Petta, Chao Xing, Clemens Schafmayer, Elmar Aigner, Christian Datz, Richard G. Lee, Luca Valenti, Daniel Lindén, Stefano Romeo, Mancina, Rosellina M, Sasidharan, Kavitha, Lindblom, Anna, Wei, Ying, Ciociola, Ester, Jamialahmadi, Ovei, Pingitore, Piero, Andréasson, Anne-Christine, Pellegrini, Giovanni, Baselli, Guido, Männistö, Ville, Pihlajamäki, Jussi, Kärjä, Vesa, Grimaudo, Stefania, Marini, Ilaria, Maggioni, Marco, Becattini, Barbara, Tavaglione, Federica, Dix, Carly, Castaldo, Marie, Klein, Stephanie, Perelis, Mark, Pattou, Francoi, Thuillier, Dorothée, Raverdy, Violeta, Dongiovanni, Paola, Fracanzani, Anna Ludovica, Stickel, Felix, Hampe, Jochen, Buch, Stephan, Luukkonen, Panu K, Prati, Daniele, Yki-Järvinen, Hannele, Petta, Salvatore, Xing, Chao, Schafmayer, Clemen, Aigner, Elmar, Datz, Christian, Lee, Richard G, Valenti, Luca, Lindén, Daniel, Romeo, Stefano, HUS Internal Medicine and Rehabilitation, Department of Medicine, and Department of Biochemistry and Developmental Biology
- Subjects
Genotype ,Endocrinology, Diabetes and Metabolism ,VARIANT ,SUSCEPTIBILITY ,Polymorphism, Single Nucleotide ,Article ,Cell Line ,Mice ,Ribonucleases ,Physiology (medical) ,Internal Medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,RNA-Seq ,Alleles ,Non-alcoholic steatohepatitis ,NONALCOHOLIC STEATOHEPATITIS ,HERITABILITY ,Gene Expression Profiling ,fungi ,NASH ,Genetic Variation ,Cell Biology ,Metabolic syndrome ,Fatty Liver ,Metabolism ,Gene Expression Regulation ,Liver ,EXOME-WIDE ASSOCIATION ,3121 General medicine, internal medicine and other clinical medicine ,ACID ,Hepatocytes ,SECRETION ,Disease Susceptibility ,VLDL ,Biomarkers ,TRIGLYCERIDES ,Non-alcoholic fatty liver disease - Abstract
Fatty liver disease (FLD) is a growing health issue with burdening unmet clinical needs. FLD has a genetic component but, despite the common variants already identified, there is still a missing heritability component. Using a candidate gene approach, we identify a locus (rs71519934) at the Pleckstrin and Sec7 domain-containing 3 (PSD3) gene resulting in a leucine to threonine substitution at position 186 of the protein (L186T) that reduces susceptibility to the entire spectrum of FLD in individuals at risk. PSD3 downregulation by short interfering RNA reduces intracellular lipid content in primary human hepatocytes cultured in two and three dimensions, and in human and rodent hepatoma cells. Consistent with this, Psd3 downregulation by antisense oligonucleotides in vivo protects against FLD in mice fed a non-alcoholic steatohepatitis-inducing diet. Thus, translating these results to humans, PSD3 downregulation might be a future therapeutic option for treating FLD., Employing a candidate gene approach, Mancina et al. identify a genetic variant of the Pleckstrin and Sec7 domain-containing 3 (PSD3) gene that reduces susceptibility to fatty liver disease. Functional studies in vitro and in vivo demonstrate that targeting PSD3 protects against fatty liver disease.
- Published
- 2022
8. Dietary carbohydrates and fats in nonalcoholic fatty liver disease
- Author
-
Panu K. Luukkonen, Hannele Yki-Järvinen, J B Moore, Leanne Hodson, Department of Medicine, HUS Internal Medicine and Rehabilitation, and University of Helsinki
- Subjects
medicine.medical_specialty ,Saturated fat ,WEIGHT-LOSS ,030209 endocrinology & metabolism ,Context (language use) ,DIFFERENTIAL OXIDATION ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Weight loss ,Internal medicine ,Nonalcoholic fatty liver disease ,Medicine ,HIGH-FRUCTOSE ,DE-NOVO LIPOGENESIS ,030304 developmental biology ,0303 health sciences ,Hepatology ,Triglyceride ,business.industry ,Gastroenterology ,HEPATIC INSULIN-RESISTANCE ,RANDOMIZED CONTROLLED-TRIAL ,medicine.disease ,Obesity ,3. Good health ,ADIPOSE-TISSUE ,Endocrinology ,chemistry ,VISCERAL FAT ,CARDIOVASCULAR-DISEASE ,3121 General medicine, internal medicine and other clinical medicine ,Lipogenesis ,LIFE-STYLE ,medicine.symptom ,business ,Dietary Carbohydrates - Abstract
This Review discusses the role of dietary fats and carbohydrates in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Studies on the dietary habits of patients with NAFLD, and the effect on liver fat accumulation of altering dietary macronutrients, are also reviewed. The global prevalence of nonalcoholic fatty liver disease (NAFLD) has dramatically increased in parallel with the epidemic of obesity. Controversy has emerged around dietary guidelines recommending low-fat-high-carbohydrate diets and the roles of dietary macronutrients in the pathogenesis of metabolic disease. In this Review, the topical questions of whether and how dietary fats and carbohydrates, including free sugars, differentially influence the accumulation of liver fat (specifically, intrahepatic triglyceride (IHTG) content) are addressed. Focusing on evidence from humans, we examine data from stable isotope studies elucidating how macronutrients regulate IHTG synthesis and disposal, alter pools of bioactive lipids and influence insulin sensitivity. In addition, we review cross-sectional studies on dietary habits of patients with NAFLD and randomized controlled trials on the effects of altering dietary macronutrients on IHTG. Perhaps surprisingly, evidence to date shows no differential effects between free sugars, with both glucose and fructose increasing IHTG in the context of excess energy. Moreover, saturated fat raises IHTG more than polyunsaturated or monounsaturated fats, with adverse effects on insulin sensitivity, which are probably mediated in part by increased ceramide synthesis. Taken together, the data support the use of diets that have a reduced content of free sugars, refined carbohydrates and saturated fat in the treatment of NAFLD.
- Published
- 2021
9. Natural Course of Nonalcoholic Fatty Liver Disease and Type 2 Diabetes in Patients With Human Immunodeficiency Virus With and Without Combination Antiretroviral Therapy–associated Lipodystrophy: A 16-Year Follow-up Study
- Author
-
Panu K. Luukkonen, Hannele Yki-Järvinen, Susanna Lallukka-Brück, Elina Isokuortti, Jussi Sutinen, Nina Lundbom, Antti Hakkarainen, HUS Internal Medicine and Rehabilitation, Department of Medicine, University of Helsinki, Helsinki University Hospital Area, HUS Medical Imaging Center, Department of Diagnostics and Therapeutics, HUS Inflammation Center, Clinicum, and Infektiosairauksien yksikkö
- Subjects
nonalcoholic fatty liver disease ,Microbiology (medical) ,medicine.medical_specialty ,glucose metabolism ,HIV-INFECTED PATIENTS ,Type 2 diabetes ,Gastroenterology ,GLUCOSE ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Fibrosis ,Internal medicine ,Nonalcoholic fatty liver disease ,medicine ,030212 general & internal medicine ,liver fibrosis ,11832 Microbiology and virology ,human immunodeficiency virus ,business.industry ,Type 2 Diabetes Mellitus ,medicine.disease ,magnetic resonance elastography ,PREVALENCE ,3. Good health ,Infectious Diseases ,3121 General medicine, internal medicine and other clinical medicine ,RISK-FACTORS ,030211 gastroenterology & hepatology ,Lipodystrophy ,Hepatic fibrosis ,Transient elastography ,business - Abstract
BackgroundAbnormal glucose metabolism and nonalcoholic fatty liver disease (NAFLD) are common in patients with human immunodeficiency virus (HIV+ patients), but longitudinal data are lacking. We determined the natural course of NAFLD (liver fat [LFAT]) and type 2 diabetes mellitus (T2DM) in HIV+ patients with and without lipodystrophy (LD+ and LD–, respectively) during a 16-year longitudinal study.MethodsLFAT (by proton magnetic resonance spectroscopy) and clinical characteristics were measured in 41 HIV+ patients at baseline and after 16 years. Liver fibrosis was estimated by measuring liver stiffness using transient elastography (TE) and magnetic resonance elastography (MRE) at 16 years. We also longitudinally studied 28 healthy subjects.ResultsDuring follow-up, the HIV+ patients gained more body fat (8.6% ± 0.7%) than the control patients (4.5% ± 0.6%, P < .001). Features of insulin resistance increased significantly in the HIV+ patients but not the control patients. A significant proportion (20%, P < .01 vs 0% at baseline) of the HIV+ but none of the control patients developed T2DM. LFAT was significantly higher at baseline in the LD+ (4.3 [1.9–11.8]) than the LD– (1.0 [0.5–1.5]; P < .001) HIV+ patients. LFAT remained stable during follow-up in all groups. At follow-up, liver stiffness measured with TE was similar among all HIV, LD+, LD–, and control patients and between the LD+ and LD– patients measured with MRE. Advanced fibrosis by MRE was observed in 3 of LD+ and none of LD– patients.ConclusionsDuring 16 years of follow-up, progression of NAFLD is rare compared to development of T2DM in HIV+ patients.
- Published
- 2019
- Full Text
- View/download PDF
10. 501-P: Lower Plasma Membrane Sn-1,2-Diacylglycerol Content and PKCepsilon/theta Activity Explain the Athlete’s Paradox
- Author
-
Panu K. Luukkonen, Mario Kahn, Rafael Calais Gaspar, José Rodrigo Pauli, Dongyan Zhang, Sandro M. Hirabara, Gary W. Cline, Kun Lyu, Brooks P. Leitner, Brandon T. Hubbard, Rachel J. Perry, Ikki Sakuma, Gerald I. Shulman, Ali Nasiri, and Kitt Falk Petersen
- Subjects
medicine.medical_specialty ,Triglyceride ,business.industry ,Endocrinology, Diabetes and Metabolism ,Theta activity ,Skeletal muscle ,Insulin sensitivity ,medicine.disease ,PKCepsilon ,chemistry.chemical_compound ,medicine.anatomical_structure ,Endocrinology ,Insulin resistance ,chemistry ,Internal medicine ,Internal Medicine ,Medicine ,Intramyocellular lipids ,business ,Diacylglycerol kinase - Abstract
Increases in intramyocellular lipids (IMCL) are strongly associated with muscle insulin resistance. An important exception to this observation occurs in athletes, who manifest increased IMCL content despite increased muscle insulin sensitivity. This phenomenon has been coined the “Athlete’s Paradox” and is not well understood. Here, we examined the hypothesis that the Athlete’s Paradox can be explained by lower plasma membrane (PM) associated sn-1,2-diacylglycerol (DAG) content leading to lower PKCε and PKCθ translocation in skeletal muscle despite increased muscle triglyceride (TAG) content. To address this hypothesis we studied 3 groups of male C57BL/6J mice following 6 weeks of: 1) Regular chow feeding (RC), 2) High-fat diet feeding (HFD); 3) RC-feeding and running wheel exercise (EX). DAG stereoisomers were assessed in five subcellular compartments [PM, endoplasmic reticulum (ER), mitochondria (Mito), and lipid droplet (LD)] using a novel liquid chromatography-tandem mass spectrometry/cellular fractionation method. Consistent with the Athlete’s Paradox we found that EX mice manifested improved glucose tolerance and muscle insulin sensitivity compared to HFD mice, as assessed by glucose tolerance and hyperinsulinemic-euglycemic clamp studies, despite similar increases in muscle TAG content (RC=5.7 ± 3.2 µg/mg; HFD=17.8 ± 6.0 µg/mg; EX=13.7 ± 5.2 µg/mg) compared to RC mice (RC vs. HFD P Conclusion: Lower PM sn-1,2-DAG content and PKCε/PKCθ activity provides an explanation for the preserved insulin sensitivity in EX mice and thus the Athlete’s Paradox. Disclosure R. C. Gaspar: None. M. Kahn: None. G. Cline: None. J. R. Pauli: None. R. J. Perry: None. K. Petersen: Advisory Panel; Spouse/Partner; AstraZeneca, iMetabolic Biopharma Corporation, Janssen Research & Development, LLC, Merck & Co., Inc., Consultant; Spouse/Partner; Aegerion Pharmaceuticals Inc., Novo Nordisk, Research Support; Self; Gilead Sciences, Inc., Merck & Co., Inc. G. I. Shulman: Consultant; Self; 89bio, Inc., BridgeBio, Ionis Pharmaceuticals, Maze Therapeutics, Novo Nordisk, Other Relationship; Self; AstraZeneca, Esperion Therapeutics, Inc, Generian Pharmaceuticals, Inc., Gilead Sciences, Inc., iMetabolic Biopharma Corporation, Janssen Research & Development, LLC, Merck & Co., Inc., The Liver Company. K. Lyu: None. B. T. Hubbard: None. B. Leitner: None. P. Luukkonen: Consultant; Self; Boehringer Ingelheim Pharmaceuticals, Inc., Research Support; Self; Novo Nordisk Foundation. S. Hirabara: None. I. Sakuma: None. A. Nasiri: Employee; Spouse/Partner; Medtronic. D. Zhang: None. Funding São Paulo Research Foundation (2019/11338-9, 2017/20542-3); National Institutes of Health (R01DK116774, P30DK045735)
- Published
- 2021
- Full Text
- View/download PDF
11. The PNPLA3-I148M Variant Confers an Antiatherogenic Lipid Profile in Insulin-resistant Patients
- Author
-
Panu K. Luukkonen, Anne Juuti, Marju Orho-Melander, Anne K. Penttilä, Sami Qadri, Henna Sammalkorpi, Tiina Lehtimäki, Johanna Arola, Hannele Yki-Järvinen, Antti Hakkarainen, HUS Internal Medicine and Rehabilitation, Department of Medicine, Clinicum, Doctoral Programme in Clinical Research, Department of Diagnostics and Therapeutics, HUS Medical Imaging Center, Helsinki University Hospital Area, HUS Abdominal Center, Department of Surgery, Department of Pathology, and Hannele Yki-Järvinen Research Group
- Subjects
Male ,Very low-density lipoprotein ,Steatosis ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Biochemistry ,Lipoprotein particle ,Cohort Studies ,0302 clinical medicine ,Endocrinology ,Methionine ,insulin resistance ,rasvamaksatauti ,genetics ,metabolinen oireyhtymä ,aineenvaihdunta ,Finland ,Disease Resistance ,2. Zero hunger ,0303 health sciences ,medicine.diagnostic_test ,rasvamaksa ,Middle Aged ,3. Good health ,Liver biopsy ,030211 gastroenterology & hepatology ,Female ,non-alcoholic fatty liver ,Adult ,medicine.medical_specialty ,ateroskleroosi ,genetiikka ,ei-alkoholiperäinen rasvamaksatauti ,lipoproteiinit ,Polymorphism, Single Nucleotide ,metabolic syndrome ,TM6SF2 ,steatoosi ,03 medical and health sciences ,Insulin resistance ,Internal medicine ,NAFLD ,metabolia ,medicine ,Humans ,Isoleucine ,Genetic Association Studies ,PNPLA3 ,030304 developmental biology ,fatty liver ,business.industry ,Biochemistry (medical) ,dyslipidemia ,Membrane Proteins ,non-alcoholic fatty liver disease ,Lipase ,medicine.disease ,Lipid Metabolism ,insuliiniresistenssi ,patatin-like phospholipase domain containing 3 ,lipoproteins ,Cross-Sectional Studies ,Amino Acid Substitution ,3121 General medicine, internal medicine and other clinical medicine ,Lipidomics ,fatty liver disease ,Metabolic syndrome ,atherosclerosis ,business ,Lipid profile ,Body mass index ,metabolism ,Lipoprotein - Abstract
Context The I148M (rs738409-G) variant in PNPLA3 increases liver fat content but may be protective against cardiovascular disease. Insulin resistance (IR) amplifies the effect of PNPLA3-I148M on liver fat. Objective To study whether PNPLA3-I148M confers an antihyperlipidemic effect in insulin-resistant patients. Design Cross-sectional study comparing the impact of PNPLA3-I148M on plasma lipids and lipoproteins in 2 cohorts, both divided into groups based on rs738409-G allele carrier status and median HOMA-IR. Setting Tertiary referral center. Patients A total of 298 obese patients who underwent a liver biopsy during bariatric surgery (bariatric cohort: age 49 ± 9 years, body mass index [BMI] 43.2 ± 6.8 kg/m2), and 345 less obese volunteers in whom liver fat was measured by proton magnetic resonance spectroscopy (nonbariatric cohort: age 45 ± 14 years, BMI 29.7 ± 5.7 kg/m2). Main Outcome Measures Nuclear magnetic resonance profiling of plasma lipids, lipoprotein particle subclasses and their composition. Results In both cohorts, individuals carrying the PNPLA3-I148M variant had significantly higher liver fat content than noncarriers. In insulin-resistant and homozygous carriers, PNPLA3-I148M exerted a distinct antihyperlipidemic effect with decreased very-low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) particles and their constituents, and increased high-density lipoprotein particles and their constituents, compared with noncarriers. VLDL particles were smaller and LDL particles larger in PNPLA3-I148M carriers. These changes were geometrically opposite to those due to IR. PNPLA3-I148M did not have a measurable effect in patients with lower IR, and its effect was smaller albeit still significant in the less obese than in the obese cohort. Conclusions PNPLA3-I148M confers an antiatherogenic plasma lipid profile particularly in insulin-resistant individuals.
- Published
- 2021
12. The PNPLA3-I148M variant increases polyunsaturated triglycerides in human adipose tissue
- Author
-
Henna Sammalkorpi, Matej Orešič, Vesa M. Olkkonen, Julia Perttilä, Panu K. Luukkonen, Marju Orho-Melander, Leanne Hodson, Sami Qadri, Antti Hakkarainen, Anne K. Penttilä, Hannele Yki-Järvinen, Tuulia Hyötyläinen, Tiina Lehtimäki, Susanna Lallukka-Brück, Amalia Gastaldelli, Anne Juuti, You Zhou, Department of Medicine, Clinicum, Doctoral Programme in Clinical Research, HUS Internal Medicine and Rehabilitation, University of Helsinki, Helsinki University Hospital Area, HUS Abdominal Center, Department of Surgery, HUS Medical Imaging Center, Department of Diagnostics and Therapeutics, and Hannele Yki-Järvinen Research Group
- Subjects
lipolyysi ,rasvahapot ,Adipose tissue ,0302 clinical medicine ,rasvamaksatauti ,genetics ,triglycerides ,variantti ,2. Zero hunger ,chemistry.chemical_classification ,education.field_of_study ,Fatty liver ,rasvamaksa ,adipose tissue ,030220 oncology & carcinogenesis ,OBESITY ,030211 gastroenterology & hepatology ,INSULIN RESISTANCE ,medicine.medical_specialty ,genetiikka ,ei-alkoholiperäinen rasvamaksatauti ,fatty acids ,steatoosi ,03 medical and health sciences ,Insulin resistance ,NEFA ,Internal medicine ,NAFLD ,medicine ,Lipolysis ,Humans ,STEATOSIS ,Adiponutrin ,Genetic Predisposition to Disease ,lipidomiikka ,adiponutriini ,education ,PNPLA3 ,fatty liver ,ADIPONUTRIN ,ihonalaisrasva ,Hepatology ,Fatty acid ,Membrane Proteins ,non-alcoholic fatty liver disease ,triglyseridit ,Lipase ,medicine.disease ,insuliiniresistenssi ,Endocrinology ,chemistry ,variant ,3121 General medicine, internal medicine and other clinical medicine ,mutaatio ,lipolysis ,lipidomics ,fatty liver disease ,lihavuus ,Steatosis ,mutation ,rasvakudos - Abstract
Background & Aims\ud \ud The I148M variant in PNPLA3 is the major genetic risk factor for non‐alcoholic fatty liver disease (NAFLD). The liver is enriched with polyunsaturated triglycerides (PUFA‐TGs) in PNPLA3‐I148M carriers. Gene expression data indicate that PNPLA3 is liver‐specific in humans, but whether it functions in adipose tissue (AT) is unknown. We investigated whether PNPLA3‐I148M modifies AT metabolism in human NAFLD.\ud Methods\ud \ud Profiling of the AT lipidome and fasting serum non‐esterified fatty acid (NEFA) composition was conducted in 125 volunteers (PNPLA3 148MM/MI, n = 63; PNPLA3 148II, n = 62). AT fatty acid composition was determined in 50 volunteers homozygous for the variant (PNPLA3 148MM, n = 25) or lacking the variant (PNPLA3 148II, n = 25). Whole‐body insulin sensitivity of lipolysis was determined using [2H5]glycerol, and PNPLA3 mRNA and protein levels were measured in subcutaneous AT and liver biopsies in a subset of the volunteers.\ud Results\ud \ud PUFA‐TGs were significantly increased in AT in carriers versus non‐carriers of PNPLA3‐I148M. The variant did not alter the rate of lipolysis or the composition of fasting serum NEFAs. PNPLA3 mRNA was 33‐fold higher in the liver than in AT (P < .0001). In contrast, PNPLA3 protein levels per tissue protein were three‐fold higher in AT than the liver (P < .0001) and nine‐fold higher when related to whole‐body AT and liver tissue masses (P < .0001).\ud Conclusions\ud \ud Contrary to previous assumptions, PNPLA3 is highly abundant in AT. PNPLA3‐I148M locally remodels AT TGs to become polyunsaturated as it does in the liver, without affecting lipolysis or composition of serum NEFAs. Changes in AT metabolism do not contribute to NAFLD in PNPLA3‐I148M carriers.
- Published
- 2020
13. 205-OR: Hepatic Protein Kinase C-e Is Necessary and Sufficient in Mediating Lipid-Induced Hepatic Insulin Resistance
- Author
-
Gary W. Cline, Gerald I. Shulman, Marcos Ricardo da Silva Rodrigues, Panu K. Luukkonen, Jesse Rinehart, Kun Lyu, Seohyuk Lee, Jonathan S. Bogan, Varman T. Samuel, Mario Kahn, Sanjay Bhanot, Morten Grønbech Rasch, Sandro M. Hirabara, Niels Blume, Mireille J. Serlie, and Dongyan Zhang
- Subjects
medicine.medical_specialty ,Ceramide ,biology ,Kinase ,business.industry ,Endocrinology, Diabetes and Metabolism ,medicine.disease ,chemistry.chemical_compound ,Endocrinology ,Insulin resistance ,chemistry ,Internal medicine ,Nonalcoholic fatty liver disease ,Internal Medicine ,medicine ,biology.protein ,Phosphorylation ,Glycogen synthase ,business ,Protein kinase B ,Diacylglycerol kinase - Abstract
Nonalcoholic fatty liver disease (NAFLD) is strongly associated with hepatic insulin resistance (HIR), however the key lipid species and molecular mechanisms linking these conditions are widely debated. In order to address these questions, we examined the metabolic impact of a liver-specific antisense oligonucleotide against PKCε and found that liver-specific PKCε knockdown ameliorated high-fat diet-induced HIR in rats, reflected by ∼2x increase in insulin-stimulated hepatic glycogen synthesis rate and improved insulin-mediated suppression of endogenous glucose production during a hyperinsulinemic-hyperglycemic (HH) clamp. This was associated with increased IRK Y1162 and Akt S473 phosphorylation and could be attributed to decreased PKCε-mediated IRK T1160 phosphorylation. Overexpressing a constitutively active PKCε (A159E) in the liver induced HIR in regular chow-fed rats, reflected by reduced insulin-stimulated glycogen synthesis rate during an HH clamp. This was associated with impaired IRK Y1162 and Akt S473 phosphorylation and could be attributed to increased PKCε-mediated IRK T1160 phosphorylation. We also developed a subcellular fractionation method to quantify diacylglycerol (DAG) stereoisomers and ceramides in the endoplasmic reticulum, mitochondria, plasma membrane (PM), lipid droplet and cytosol and applied this method to a cohort of obese human subjects with and without NAFLD and HIR. Liver PM sn-1,2 DAG content was ∼5x higher in HIR individuals with NAFLD compared to insulin-sensitive individuals without NAFLD, associated with ∼3x higher IRK T1160 phosphorylation, supporting the importance of PM sn-1,2 DAG-PKCε-IRK pT1160 pathway to mediate HIR in humans with NAFLD. In contrast there were no significant differences in ceramide content in any of the subcellular compartments. Conclusion: These data identify PM sn-1,2 DAGs as the key lipids that activate PKCε, and that hepatic PKCε is both necessary and sufficient in mediating HIR. Disclosure K. Lyu: None. D. Zhang: None. M. Kahn: None. M.R.S. Rodrigues: None. S. Hirabara: None. P. Luukkonen: None. S. Lee: None. S. Bhanot: None. J. Rinehart: None. N. Blume: Employee; Self; Novo Nordisk A/S. Stock/Shareholder; Self; Novo Nordisk A/S. M. Rasch: Employee; Self; Novo Nordisk A/S. Stock/Shareholder; Self; Novo Nordisk A/S. M.J. Serlie: None. J. Bogan: None. G. Cline: None. V. Samuel: None. G.I. Shulman: Advisory Panel; Self; AstraZeneca, Janssen Research & Development, LLC, Merck & Co., Inc. Advisory Panel; Spouse/Partner; Merck & Co., Inc. Consultant; Self; Novo Nordisk A/S. Consultant; Spouse/Partner; Novo Nordisk A/S. Other Relationship; Self; Gilead Sciences, Inc., iMetabolic Biopharma Corporation. Other Relationship; Spouse/Partner; iMetabolic Biopharma Corporation. Other Relationship; Self; Maze Therapeutics. Funding U.S. Public Health Service (R01DK116774, R01DK119968, R01DK113984, P30DK045735, R01 DK092661); U.S. Department of Veternas Affairs (I01BX000901)
- Published
- 2020
- Full Text
- View/download PDF
14. MARC1 variant rs2642438 increases hepatic phosphatidylcholines and decreases severity of non-alcoholic fatty liver disease in humans
- Author
-
Marju Orho-Melander, Panu K. Luukkonen, Anne K. Penttilä, Anne Juuti, Matej Orešič, Tuulia Hyötyläinen, Johanna Arola, Hannele Yki-Järvinen, and Henna Sammalkorpi
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Hepatology ,business.industry ,Fatty liver ,Non alcoholic ,macromolecular substances ,Disease ,medicine.disease ,Severity of Illness Index ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Liver ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,medicine ,Phosphatidylcholines ,Humans ,030211 gastroenterology & hepatology ,business - Abstract
MARC1 variant rs2642438 increases hepatic phosphatidylcholines and decreases severity of non-alcoholic fatty liver disease in humans
- Published
- 2020
15. The Pi∗MZ Allele in Alpha-1 Antitrypsin Increases Liver-Related Outcomes in a Population-Based Study
- Author
-
Fredrik Åberg, Panu K. Luukkonen, and Veikko Salomaa
- Subjects
0303 health sciences ,medicine.medical_specialty ,Hepatology ,business.industry ,Gastroenterology ,Alpha (ethology) ,Population based study ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Pi ,030211 gastroenterology & hepatology ,Genetic risk ,Allele ,business ,030304 developmental biology - Published
- 2021
- Full Text
- View/download PDF
16. Impaired hepatic lipid synthesis from polyunsaturated fatty acids in TM6SF2 E167K variant carriers with NAFLD
- Author
-
Panu K. Luukkonen, Jeremy M. Palmer, Taru Tukiainen, Marja Leivonen, Marju Orho-Melander, Hannele Yki-Järvinen, Adnan Ali, Johanna Arola, Petter Vikman, Leif Groop, Matej Orešič, Tuulia Hyötyläinen, Emma Scott, P.A. Nidhina Haridas, Vesa M. Olkkonen, Quentin M. Anstee, You Zhou, Anne Juuti, Linda Ahonen, Om Prakash Dwivedi, Department of Medicine, Clinicum, Institute for Molecular Medicine Finland, II kirurgian klinikka, Department of Surgery, Medicum, Department of Pathology, Leif Groop Research Group, Hannele Yki-Järvinen Research Group, HUS Internal Medicine and Rehabilitation, and HUS Abdominal Center
- Subjects
Male ,0301 basic medicine ,Apolipoprotein B ,Lipoproteins, VLDL ,chemistry.chemical_compound ,ta318 ,chemistry.chemical_classification ,INSULIN-RESISTANCE ,biology ,NONALCOHOLIC STEATOHEPATITIS ,Middle Aged ,Lipids ,Transmembrane protein ,3. Good health ,Liver ,Arachidonic acid ,LOW-DENSITY LIPOPROTEINS ,CARDIOVASCULAR-DISEASE ,Phosphatidylethanolamine N-methyltransferase ,Lipogenesis ,Fatty Acids, Unsaturated ,Phosphatidylcholines ,Female ,lipids (amino acids, peptides, and proteins) ,Polyunsaturated fatty acid ,Adult ,Heterozygote ,medicine.medical_specialty ,Genotype ,APOLIPOPROTEIN-B ,Cholesterol esters ,digestive system ,03 medical and health sciences ,Insulin resistance ,LIVER-DISEASE ,Internal medicine ,medicine ,Humans ,Fatty acids ,Triglycerides ,Hepatology ,PHOSPHATIDYLETHANOLAMINE N-METHYLTRANSFERASE ,Membrane Proteins ,nutritional and metabolic diseases ,ARACHIDONIC-ACID ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Endocrinology ,chemistry ,3121 General medicine, internal medicine and other clinical medicine ,Hepatocytes ,FIBROSIS PROGRESSION ,biology.protein ,Transmembrane 6 superfamily member 2 ,SUPERFAMILY MEMBER 2 ,Non-alcoholic fatty liver disease ,TM6SF2 - Abstract
Background: Carriers of the transmembrane 6 superfamily member 2 E167K gene variant (TM6SF2(EK/KK)) have decreased expression of the TM6SF2 gene and increased risk of NAFLD and NASH. Unlike common 'obese/metabolic' NAFLD, these subjects lack hypertriglyceridemia and have lower risk of cardiovascular disease. In animals, phosphatidylcholine (PC) deficiency results in a similar phenotype. PCs surround the core of VLDL consisting of triglycerides (TGs) and cholesteryl-esters (CEs). We determined the effect of the TM6SF2 E167K on these lipids in the human liver and serum and on hepatic gene expression and studied the effect of TM6SF2 knockdown on hepatocyte handling of these lipids. Methods: Liver biopsies were taken from subjects characterized with respect to the TM6SF2 genotype, serum and liver lipidome, gene expression and histology. In vitro, after TM6SF2 knockdown in HuH-7 cells, we compared incorporation of different fatty acids into TGs, CEs, and PCs. Results: The TM6SF2(EK/KK) and TM6SF2EE groups had similar age, gender, BMI and HOMA-IR. Liver TGs and CEs were higher and liver PCs lower in the TM6SF2(EK/KK) than the TM6SF2EE group (p Conclusions: Hepatic lipid synthesis from PUFAs is impaired and could contribute to deficiency in PCs and increased intrahepatic TG in TM6SF2 E167K variant carriers. (C) 2017 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.
- Published
- 2017
- Full Text
- View/download PDF
17. Diabetes, Liver Cancer, and Cirrhosis: What Next?
- Author
-
Hannele Yki-Järvinen and Panu K. Luukkonen
- Subjects
medicine.medical_specialty ,Cirrhosis ,Hepatology ,business.industry ,Incidence (epidemiology) ,Fatty liver ,medicine.disease ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Internal medicine ,Diabetes mellitus ,medicine ,030211 gastroenterology & hepatology ,Liver cancer ,Prospective cohort study ,business ,Body mass index - Published
- 2018
- Full Text
- View/download PDF
18. rs641738C>T near MBOAT7 is positively associated with liver fat, ALT, and histological severity of NAFLD: a meta-analysis
- Author
-
Jesus M. Banales, Jerome I. Rotter, Connor A. Emdin, Leon A. Adams, Sreekumar G. Pillai, Stephan Buch, Yu Ri Im, Thomas Berg, Claudia Langenberg, Kendra A. Young, Felix Stickel, Elizabeth K. Speliotes, Julia Kozlitina, Sonia Caprio, Madison Fairey, Kevin Teo, Hanieh Yaghootkar, Abhijit Chowdhury, Eu-Pnafld Investigators, Yii-Der Ida Chen, Harriet Hunter, Faiza Qayyum, Pavel Strnad, Matthias C. Reichert, Elmar Aigner, Marcin Krawczyk, Dana de Gracia Hahn, Lin Li, Matthew Hickman, Nicholette D. Palmer, Panu K. Luukkonen, Jiawen Dong, Christian Datz, Ali Canbay, Kent D. Taylor, Joel E. Lavine, Philip E. Melton, Nicola Santoro, Richard M. Watanabe, Xiuqing Guo, Giuseppina Rosaria Umano, Lynne E. Wagenknecht, Amedine Duret, Pallav Bhatnagar, Trevor A. Mori, Johanna K. DiStefano, Anne Tybjærg-Hansen, Stefano Romeo, Julia Wattacheril, Jake P. Mann, Luca Valenti, Ankita Chatterjee, Stefan Stender, Jochen Hampe, Rajarshi Banerjee, Glenn S. Gerhard, Frank Lammert, Rohit Loomba, Christian A. Hudert, Kushala W M Abeysekera, L Heintz, Constantinos A. Parisinos, Hong Kai Lim, Christopher D. Still, Matt Kelly, Mrudula Utukuri, Priyadarshi Basu, Nicholas J. Wareham, Hannele Yki-Järvinen, and Clemens Schafmayer
- Subjects
0303 health sciences ,medicine.medical_specialty ,business.industry ,Insulin ,medicine.medical_treatment ,Fatty liver ,Genome-wide association study ,medicine.disease ,Gastroenterology ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Internal medicine ,Genetic model ,medicine ,Steatosis ,Risk factor ,business ,030304 developmental biology - Abstract
Background & AimsA common genetic variant near MBOAT7 (rs641738C>T) has been previously associated with hepatic fat and advanced histology in non-alcoholic fatty liver disease (NAFLD), however, these findings have not been consistently replicated in the literature. We aimed to establish whether rs641738C>T is a risk factor across the spectrum of NAFLD and characterize its role in the regulation of related metabolic phenotypes through meta-analysis.MethodsWe performed meta-analysis of studies with data on the association between rs641738C>T genotype and: liver fat, NAFLD histology, and serum ALT, lipids, or insulin. These included directly genotyped studies and population-level data from genome-wide association studies (GWAS). We performed random effects meta-analysis using recessive, additive, and dominant genetic models.ResultsData from 1,047,265 participants (8,303 with liver biopsies) across 42 studies was included in the meta-analysis. rs641738C>T was associated with higher liver fat on CT/MRI (+0.03 standard deviations [95% CI: 0.02 - 0.05]) and diagnosis of NAFLD (OR 1.22 [95% CI 1.08 - 1.39]) in Caucasian adults. The variant was also positively associated with presence of severe steatosis, NASH, and advanced fibrosis (OR: 1.32 [95% CI: 1.06 - 1.63]) in Caucasian adults using a recessive model of inheritance (CC+CT vs. TT). Meta-analysis of data from previous GWAS found the variant to be associated with higher ALT (Pz=0.002) and lower serum triglycerides (Pz=1.5×10−4). rs641738C>T was not associated with fasting insulin and no effect was observed in children with NAFLD.ConclusionOur study validates rs641738C>T near MBOAT7 as a risk factor for the presence and severity of NAFLD in individuals of European descent.
- Published
- 2019
- Full Text
- View/download PDF
19. Human PNPLA3-I148M variant increases hepatic retention of polyunsaturated fatty acids
- Author
-
Matej Orešič, Maarit Hölttä-Vuori, Hannele Yki-Järvinen, Marju Orho-Melander, Markku Peltonen, Auli Nick, Leanne Hodson, Elina Ikonen, Antti Hakkarainen, Christoph Thiele, Tuulia Hyötyläinen, Susanna Lallukka-Brück, Panu K. Luukkonen, Elina Isokuortti, Nina Lundbom, You Zhou, Minerva Foundation Institute for Medical Research Helsinki, University of Bonn, Department of Neuroscience and Biomedical Engineering, University of Helsinki, National Institute for Health and Welfare, Lund University, University of Turku, Örebro University, University of Oxford, Aalto-yliopisto, Aalto University, HUS Internal Medicine and Rehabilitation, Department of Medicine, Director and Common Matters, Department of Anatomy, Medicum, University Management, HUS Medical Imaging Center, Department of Diagnostics and Therapeutics, STEMM - Stem Cells and Metabolism Research Program, and Lipid Trafficking Lab
- Subjects
0301 basic medicine ,Male ,Very low-density lipoprotein ,Cholesterol, VLDL ,Phospholipase ,OXIDATION ,DISEASE ,I148M ,chemistry.chemical_compound ,0302 clinical medicine ,Transacylation ,Loss of Function Mutation ,chemistry.chemical_classification ,Fatty Acids ,food and beverages ,General Medicine ,Middle Aged ,Postprandial ,Liver ,030220 oncology & carcinogenesis ,Fatty Acids, Unsaturated ,TRIGLYCERIDE ,lipids (amino acids, peptides, and proteins) ,Female ,Polyunsaturated fatty acid ,Research Article ,EXPRESSION ,Adult ,medicine.medical_specialty ,Heterozygote ,Adolescent ,WEIGHT-LOSS ,METABOLISM ,Cell Line ,03 medical and health sciences ,Young Adult ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Triglycerides ,Triglyceride ,nutritional and metabolic diseases ,Membrane Proteins ,Lipase ,LIPOPROTEIN ,030104 developmental biology ,Endocrinology ,chemistry ,GREATER ,LIVER FAT ,3111 Biomedicine ,Lipoprotein ,Chylomicron - Abstract
openaire: EC/H2020/634413/EU//EPoS | openaire: EC/H2020/777377/EU//LITMUS The common patatin-like phospholipase domain-containing protein 3 (PNPLA3) variant I148M predisposes to nonalcoholic liver disease but not its metabolic sequelae. We compared the handling of labeled polyunsaturated fatty acids (PUFAs) and saturated fatty acids (SFA) in vivo in humans and in cells harboring different PNPLA3 genotypes. In 148M homozygous individuals, triglycerides (TGs) in very low-density lipoproteins (VLDL) were depleted of PUFAs both under fasting and postprandial conditions compared with 148I homozygotes, and the PUFA/SFA ratio in VLDL-TGs was lower relative to the chylomicron precursor pool. In human PNPLA3-148M and PNPLA3-KO cells, PUFA but not SFA incorporation into TGs was increased at the expense of phosphatidylcholines, and under lipolytic conditions, PUFA-containing diacylglycerols (DAGs) accumulated compared with PNPLA3-148I cells. Polyunsaturated TGs were increased, while phosphatidylcholines (PCs) were decreased in the human liver in 148M homozygous individuals as compared with 148I homozygotes. We conclude that human PNPLA3-I148M is a loss-of-function allele that remodels liver TGs in a polyunsaturated direction by impairing hydrolysis/transacylation of PUFAs from DAGs to feed phosphatidylcholine synthesis.
- Published
- 2019
20. Hepatic ceramides dissociate steatosis and insulin resistance in patients with non-alcoholic fatty liver disease
- Author
-
Sanja Sädevirta, Matej Orešič, Panu K. Luukkonen, Tuulia Hyötyläinen, Marja Leivonen, Hannele Yki-Järvinen, Johanna Arola, You Zhou, Department of Medicine, Clinicum, II kirurgian klinikka, Department of Surgery, Medicum, Department of Pathology, and Hannele Yki-Järvinen Research Group
- Subjects
Male ,0301 basic medicine ,Biopsy ,VARIANT ,Adipose tissue ,GLUCOSE ,I148M ,chemistry.chemical_compound ,Patatin-like phospholipase domain containing protein 3 ,0302 clinical medicine ,High-density lipoprotein ,Non-alcoholic Fatty Liver Disease ,Risk Factors ,Nonalcoholic fatty liver disease ,Non-alcoholic steatohepatitis ,Fatty liver ,GENETIC-VARIATION ,Middle Aged ,3. Good health ,ADIPOSE-TISSUE ,HISTOLOGICAL SEVERITY ,Liver ,Lipogenesis ,Female ,030211 gastroenterology & hepatology ,OBESE SUBJECTS ,Adult ,medicine.medical_specialty ,Adolescent ,Biology ,Free fatty acids ,Ceramides ,Young Adult ,03 medical and health sciences ,Insulin resistance ,Diabetes mellitus ,Internal medicine ,medicine ,Humans ,PNPLA3 ,Aged ,Hepatology ,nutritional and metabolic diseases ,DIABETES-MELLITUS ,ACIDS ,medicine.disease ,R1 ,030104 developmental biology ,Endocrinology ,chemistry ,3121 General medicine, internal medicine and other clinical medicine ,Dihydroceramides ,Insulin Resistance ,Steatosis - Abstract
Background & Aims: Recent data in mice have identified de novo ceramide synthesis as the key mediator of hepatic insulin resistance (IR) that in humans characterizes increases in liver fat due to IR ('Metabolic NAFLD' but not that due to the I148M gene variant in PNPLA3 ('PNPLA3 NAFLD'). We determined which bioactive lipids co-segregate with IR in the human liver. Methods: Liver lipidome was profiled in liver biopsies from 125 subjects that were divided into equally sized groups based on median HOMA-IR ('High and Low HOMA-IR', n = 62 and n = 63) or PNPLA3 genotype (PNPIA3(148MM/MI), n = 61 vs. PNPLA3(148II), n = 64). The subjects were also divided into 4 groups who had either IR, the I148M gene variant, both of the risk factors or neither. Results: Steatosis and NASH prevalence were similarly increased in 'High HOMA-IR' and PNPLA3(148MM/MI) groups compared to their respective control groups. The 'High HOMA-IR' but not the PNPLA3(148MM/MI) group had features of IR. The liver in 'High HOMA-IR' vs. low HOMA-IR' was markedly enriched in saturated and monounsaturated triacylglycerols and free fatty acids, dihydroceramides (markers of de novo ceramide synthesis) and ceramides. Markers of other ceramide synthetic pathways were unchanged. In PNPLA3(148MM/MI) vs. PNPLA3(148II), the increase in liver fat was due to polyunsaturated triacylglycerols while other lipids were unchanged. Similar changes were observed when data were analyzed using the 4 subgroups. Conclusions: Similar increases in liver fat and NASH are associated with a metabolically harmful saturated, ceramide-enriched liver lipidome in 'Metabolic NAFLD' but not in 'PNPLA3 NAFLD'. This difference may explain why metabolic but not PNPLA3 NAFLD increases the risk of type 2 diabetes and cardiovascular disease. (C) 2016 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.
- Published
- 2016
- Full Text
- View/download PDF
21. Predictors of Liver Fat and Stiffness in Non-Alcoholic Fatty Liver Disease (NAFLD) – an 11-Year Prospective Study
- Author
-
Panu K. Luukkonen, Antti Hakkarainen, Markus T. Kallio, Marju Orho-Melander, Sanja Sädevirta, Susanna Lallukka, Nina Lundbom, You Zhou, Hannele Yki-Järvinen, Clinicum, University of Helsinki, Department of Medicine, HUS Internal Medicine and Rehabilitation, Department of Diagnostics and Therapeutics, and HUS Medical Imaging Center
- Subjects
Male ,0301 basic medicine ,VARIANT ,lcsh:Medicine ,Adipose tissue ,Disease ,Gastroenterology ,GLUCOSE ,0302 clinical medicine ,Non-alcoholic Fatty Liver Disease ,Risk Factors ,Fibrosis ,Prospective Studies ,lcsh:Science ,Prospective cohort study ,CONFERS SUSCEPTIBILITY ,METABOLIC SYNDROME ,GENERAL-POPULATION ,Multidisciplinary ,Fatty liver ,Middle Aged ,MAGNETIC-RESONANCE-SPECTROSCOPY ,ADIPOSE-TISSUE ,Liver ,Female ,030211 gastroenterology & hepatology ,Adult ,medicine.medical_specialty ,Article ,03 medical and health sciences ,Insulin resistance ,Internal medicine ,medicine ,Humans ,HEPATIC STEATOSIS ,business.industry ,lcsh:R ,medicine.disease ,R1 ,Fatty Liver ,030104 developmental biology ,Endocrinology ,3121 General medicine, internal medicine and other clinical medicine ,INSULIN THERAPY ,lcsh:Q ,Insulin Resistance ,Metabolic syndrome ,FOLLOW-UP ,Transient elastography ,business - Abstract
Liver fat can be non-invasively measured by proton magnetic resonance spectroscopy (1H-MRS) and fibrosis estimated as stiffness using transient elastography (FibroScan). There are no longitudinal data on changes in liver fat in Europids or on predictors of liver stiffness using these methods. We determined liver fat (1H-MRS) and clinical characteristics including features of insulin resistance at baseline and after a median follow-up period of 11.3 (range 7.3–13.4) years in 97 Finnish subjects. Liver stiffness was measured at 11.3 years. Liver fat content decreased by 5% (p r = 0.81, p
- Published
- 2017
- Full Text
- View/download PDF
22. The MBOAT7 variant rs641738 alters hepatic phosphatidylinositols and increases severity of non-alcoholic fatty liver disease in humans
- Author
-
Panu K. Luukkonen, Matej Orešič, Tuulia Hyötyläinen, Hannele Yki-Järvinen, Marja Leivonen, You Zhou, Marju Orho-Melander, Johanna Arola, Department of Medicine, Clinicum, II kirurgian klinikka, Department of Surgery, Medicum, and Department of Pathology
- Subjects
0301 basic medicine ,medicine.medical_specialty ,education ,macromolecular substances ,Disease ,Phosphatidylinositols ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Fibrosis ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,medicine ,FIBROSIS ,Humans ,Genetic Predisposition to Disease ,RISK ,Hepatology ,business.industry ,Fatty liver ,Membrane Proteins ,Non alcoholic ,medicine.disease ,3126 Surgery, anesthesiology, intensive care, radiology ,R1 ,3. Good health ,030104 developmental biology ,Liver ,3121 General medicine, internal medicine and other clinical medicine ,030211 gastroenterology & hepatology ,3111 Biomedicine ,business ,Acyltransferases - Abstract
The MBOAT7 variant rs641738 alters hepatic phosphatidylinositols and increases severity of non-alcoholic fatty liver disease in humans
- Published
- 2016
23. Heterogeneity of non-alcoholic fatty liver disease
- Author
-
Hannele Yki-Järvinen and Panu K. Luukkonen
- Subjects
0303 health sciences ,medicine.medical_specialty ,Hepatology ,business.industry ,Fatty liver ,Non alcoholic ,Disease ,medicine.disease ,Obesity ,Fatty Liver ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Endocrinology ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,medicine ,Humans ,030211 gastroenterology & hepatology ,Steatosis ,Insulin Resistance ,business ,030304 developmental biology - Published
- 2015
- Full Text
- View/download PDF
24. Continuous grading of early fibrosis in nafld using label-free imaging
- Author
-
Antti Isomäki, Sanja Sädevirta, Hannele Yki-Järvinen, Taina Pihlajaniemi, Juho Pirhonen, Panu K. Luukkonen, Sanna-Maria Karppinen, Elina Ikonen, Mika Hukkanen, and Johanna Arola
- Subjects
0301 basic medicine ,Nonalcoholic steatohepatitis ,medicine.medical_specialty ,Pathology ,030102 biochemistry & molecular biology ,business.industry ,medicine.disease ,Gastroenterology ,03 medical and health sciences ,Fibrosis ,Internal medicine ,medicine ,Cardiology and Cardiovascular Medicine ,business ,Grading (tumors) ,Label free - Published
- 2016
- Full Text
- View/download PDF
25. Saturated Fat Is More Metabolically Harmful for the Human Liver Than Unsaturated Fat or Simple Sugars
- Author
-
Anne Salonen, Matej Orešič, Antti Hakkarainen, Linda Ahonen, Véronique Pelloux, Leanne Hodson, Brandon D. Kayser, Hannele Yki-Järvinen, Marju Orho-Melander, Susanna Lallukka, Melania Gaggini, Panu K. Luukkonen, Sanja Sädevirta, Ching Jian, Helena Gylling, Adnan Ali, Nina Lundbom, Aila Rissanen, Amalia Gastaldelli, Tuulia Hyötyläinen, You Zhou, and Karine Clément
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Saturated fat ,Adipose tissue ,Weight Gain ,03 medical and health sciences ,chemistry.chemical_compound ,Insulin resistance ,Dietary Fats, Unsaturated ,Non-alcoholic Fatty Liver Disease ,Internal medicine ,Internal Medicine ,medicine ,Humans ,Insulin ,Lipolysis ,Pathophysiology/Complications ,Triglycerides ,Unsaturated fatty acid ,2. Zero hunger ,Advanced and Specialized Nursing ,Triglyceride ,business.industry ,Fatty Acids ,Monosaccharides ,Unsaturated fat ,Feeding Behavior ,Middle Aged ,Overweight ,Lipid Metabolism ,medicine.disease ,3. Good health ,030104 developmental biology ,Endocrinology ,Adipose Tissue ,Liver ,chemistry ,Saturated fatty acid ,Carbohydrate Metabolism ,Female ,Insulin Resistance ,business - Abstract
OBJECTIVE Nonalcoholic fatty liver disease (i.e., increased intrahepatic triglyceride [IHTG] content), predisposes to type 2 diabetes and cardiovascular disease. Adipose tissue lipolysis and hepatic de novo lipogenesis (DNL) are the main pathways contributing to IHTG. We hypothesized that dietary macronutrient composition influences the pathways, mediators, and magnitude of weight gain-induced changes in IHTG. RESEARCH DESIGN AND METHODS We overfed 38 overweight subjects (age 48 ± 2 years, BMI 31 ± 1 kg/m2, liver fat 4.7 ± 0.9%) 1,000 extra kcal/day of saturated (SAT) or unsaturated (UNSAT) fat or simple sugars (CARB) for 3 weeks. We measured IHTG (1H-MRS), pathways contributing to IHTG (lipolysis ([2H5]glycerol) and DNL (2H2O) basally and during euglycemic hyperinsulinemia), insulin resistance, endotoxemia, plasma ceramides, and adipose tissue gene expression at 0 and 3 weeks. RESULTS Overfeeding SAT increased IHTG more (+55%) than UNSAT (+15%, P < 0.05). CARB increased IHTG (+33%) by stimulating DNL (+98%). SAT significantly increased while UNSAT decreased lipolysis. SAT induced insulin resistance and endotoxemia and significantly increased multiple plasma ceramides. The diets had distinct effects on adipose tissue gene expression. CONCLUSIONS Macronutrient composition of excess energy influences pathways of IHTG: CARB increases DNL, while SAT increases and UNSAT decreases lipolysis. SAT induced the greatest increase in IHTG, insulin resistance, and harmful ceramides. Decreased intakes of SAT could be beneficial in reducing IHTG and the associated risk of diabetes.
- Full Text
- View/download PDF
26. Hydroxysteroid 17-β dehydrogenase 13 variant increases phospholipids and protects against fibrosis in nonalcoholic fatty liver disease
- Author
-
Matej Orešič, Johanna Arola, Amalia Gastaldelli, Henna Sammalkorpi, Marju Orho-Melander, Leanne Hodson, Tuulia Hyötyläinen, Antti Hakkarainen, Anne Juuti, Taru Tukiainen, Onni Niemelä, Petri T. Kovanen, Om Prakash Dwivedi, Hannele Yki-Järvinen, Leif Groop, P.A. Nidhina Haridas, Panu K. Luukkonen, HUS Internal Medicine and Rehabilitation, Department of Medicine, University of Helsinki, Genomics of Sex Differences, Institute for Molecular Medicine Finland, HUS Abdominal Center, II kirurgian klinikka, HUSLAB, Department of Pathology, HUS Medical Imaging Center, Department of Diagnostics and Therapeutics, Centre of Excellence in Complex Disease Genetics, Minerva Foundation Institute for Medical Research Helsinki, Tampere University, Lund University, Department of Neuroscience and Biomedical Engineering, Wihuri Research Institute Finland, University of Oxford, National Research Council of Italy, Örebro University, University of Turku, Aalto-yliopisto, and Aalto University
- Subjects
Liver Cirrhosis ,Male ,0301 basic medicine ,medicine.medical_specialty ,HOMEOSTASIS ,Cirrhosis ,17-Hydroxysteroid Dehydrogenases ,Lipolysis ,NUTRIENT ,GLUCOSE ,STEATOHEPATITIS ,03 medical and health sciences ,Liver disease ,0302 clinical medicine ,Insulin resistance ,Non-alcoholic Fatty Liver Disease ,Fibrosis ,Internal medicine ,PHOSPHATIDYLCHOLINE ,Nonalcoholic fatty liver disease ,medicine ,Humans ,Phospholipids ,DE-NOVO LIPOGENESIS ,INSULIN-RESISTANCE ,Chemistry ,General Medicine ,Middle Aged ,medicine.disease ,3. Good health ,INDIVIDUALS ,030104 developmental biology ,Endocrinology ,030220 oncology & carcinogenesis ,3121 General medicine, internal medicine and other clinical medicine ,Lipidomics ,Lipogenesis ,CHOLINE ,Female ,Insulin Resistance ,Steatosis ,Steatohepatitis ,Transcriptome ,Research Article ,PACKAGE - Abstract
Carriers of the hydroxysteroid 17-beta dehydrogenase 13 (HSD17B13) gene variant (rs72613567:TA) have a reduced risk of NASH and cirrhosis but not steatosis. We determined its effect on liver histology, lipidome, and transcriptome using ultra performance liquid chromatography-mass spectrometry and RNA-seq. In carriers and noncarriers of the gene variant, we also measured pathways of hepatic fatty acids (de novo lipogenesis [ONLI and adipose tissue lipolysis [ATL] using (H2O)-H-2 and H-2-glycerol) and insulin sensitivity using H-3-glucose and euglycemic-hyperinsulinemic clamp) and plasma cytokines. Carriers and noncarriers had similar age, sex and BMI. Fibrosis was significantly less frequent while phospholipids, but not other lipids, were enriched in the liver in carriers compared with noncarriers. Expression of 274 genes was altered in carriers compared with noncarriers, consisting predominantly of downregulated inflammation-related gene sets. Plasma IL-6 concentrations were lower, but DNL, ATL and hepatic insulin sensitivity were similar between the groups. In conclusion, carriers of the HSD17B13 variant have decreased fibrosis and expression of inflammation-related genes but increased phospholipids in the liver. These changes are not secondary to steatosis, ONL, ATL, or hepatic insulin sensitivity. The increase in phospholipids and decrease in fibrosis are opposite to features of choline-deficient models of liver disease and suggest HSD17B13 as an attractive therapeutic target.
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.