1
|
Rabehl M, Wei Z, Leineweber CG, Enssle J, Rothe M, Jung A, Schmöcker C, Elbelt U, Weylandt KH, Pietzner A. Effect of FADS1 SNPs rs174546, rs174547 and rs174550 on blood fatty acid profiles and plasma free oxylipins. Front Nutr 2024; 11:1356986. [PMID: 39021601 PMCID: PMC11253720 DOI: 10.3389/fnut.2024.1356986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/06/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Previous studies have indicated that activity of fatty acid desaturase 1 (FADS1), is involved in cardiometabolic risk. Recent experimental data have shown that FADS1 knockdown can promote lipid accumulation and lipid droplet formation in liver cells. In this study, we aimed to characterize whether different FADS1 genotypes affect liver fat content, essential fatty acid content and free oxylipin mediators in the blood. Methods We analyzed the impact of FADS1 single-nucleotide polymorphisms (SNPs) rs174546, rs174547, and rs174550 on blood fatty acids and free oxylipins in a cohort of 85 patients from an academic metabolic medicine outpatient center. Patients were grouped based on their genotype into the homozygous major (derived) allele group, the heterozygous allele group, and the homozygous minor (ancestral) allele group. Omega-3 polyunsaturated fatty acids (n-3 PUFA) and omega-6 polyunsaturated fatty acids (n-6 PUFA) in the blood cell and plasma samples were analyzed by gas chromatography. Free Oxylipins in plasma samples were analyzed using HPLC-MS/MS. Liver fat content and fibrosis were evaluated using Fibroscan technology. Results Patients with the homozygous ancestral (minor) FADS1 genotype exhibited significantly lower blood levels of the n-6 PUFA arachidonic acid (AA), but no significant differences in the n-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). There were no significant differences in liver fat content or arachidonic acid-derived lipid mediators, such as thromboxane B2 (TXB2), although there was a trend toward lower levels in the homozygous ancestral genotype group. Discussion Our findings suggest that FADS1 genotypes influence the blood levels of n-6 PUFAs, while not significantly affecting the n-3 PUFAs EPA and DHA. The lack of significant differences in liver fat content and arachidonic acid-derived lipid mediators suggests that the genotype-related variations in fatty acid levels may not directly translate to differences in liver fat or inflammatory lipid mediators in this cohort. However, the trend towards lower levels of certain lipid mediators in the homozygous ancestral genotype group warrants further investigation to elucidate the underlying mechanisms of different FADS1 genotypes and potential implications for cardiometabolic risk.
Collapse
Affiliation(s)
- Miriam Rabehl
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Zeren Wei
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Can G. Leineweber
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Jörg Enssle
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | | | - Adelheid Jung
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
| | - Christoph Schmöcker
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Ulf Elbelt
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Karsten H. Weylandt
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Brandenburg Institute for Clinical Ultrasound, Brandenburg Medical School, Neuruppin, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| | - Anne Pietzner
- Medical Department B, Division of Hepatology, Gastroenterology, Oncology, Hematology, Palliative Care, Endocrinology and Diabetes, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, Neuruppin, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, Potsdam, Germany
- Experimental Lipidology, Brandenburg Medical School, Neuruppin, Germany
| |
Collapse
|
2
|
Pavlova NI, Krylov AV, Bochurov AA, Alekseev VA, Kurtanov KA. High Frequency of Ancestral Haplotype A of Fatty Acid Desaturase Genes in the Yakut Population. Genet Test Mol Biomarkers 2024. [PMID: 38860387 DOI: 10.1089/gtmb.2024.0085] [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: 06/12/2024] Open
Abstract
Aims: The purpose of this study was to study the correlation of the body weight of Yakuts with the variability of polymorphisms rs174537, rs174546 and rs3834458 of the FADS1 - FADS2 region to identify the connection of certain genotypes with obesity. Materials and Methods: For genotyping, classical methods of PCR-RFLP analysis were used. A sample of 446 DNA samples from Yakut volunteers without chronic diseases (143 women and 303 men) was studied. Results: The predominance of the ancestral alleles of SNPs rs174537, rs174546 and rs3834458 was established in all of our studied groups. Analysis of the odds ratio of allele and genotype frequencies in patients with normal BMI, high BMI and obesity did not show statistically significant values. We did not find an association between rs174537, rs174546 and rs3834458 with obesity, but we did not take into account the diet of the subjects, which may have had a stronger effect on BMI. Analysis of pairwise linkage disequilibrium and assessment of haplotypes for 3 SNPs in the FADS1 and FADS2 genes showed strong linkage of all three SNPs to each other (r2 = 0.93-0.96). Conclusions: According to the result of genotyping of SNP rs174537, the frequency of haplotype A in the Yakut population was 0.76 and, in comparison with other world data, is quite high. Which in turn is associated with lower conversion of short-chain polyunsaturated fatty acid to long-chain polyunsaturated fatty acid. Accordingly, a shift in nutrition towards more plant foods can negatively impact the health of the Yakuts. At the moment, the exact dosage of polyunsaturated fatty acids (PUFAs) for humans has not yet been established, but judging by the fact that all recommendations are mainly made on the basis of European populations, in connection with the results of the study, the Yakuts have a particularly high need for PUFAs.
Collapse
Affiliation(s)
- Nadezhda I Pavlova
- Federal State Budgetary Scientific Institution, Yakut Scientific Center for Medical Problems, Yakutsk, Russian Federation
| | - Alexey V Krylov
- Federal State Budgetary Scientific Institution, Yakut Scientific Center for Medical Problems, Yakutsk, Russian Federation
| | - Alexey A Bochurov
- Federal State Budgetary Scientific Institution, Yakut Scientific Center for Medical Problems, Yakutsk, Russian Federation
| | - Vladislav A Alekseev
- Federal State Budgetary Scientific Institution, Yakut Scientific Center for Medical Problems, Yakutsk, Russian Federation
| | - Khariton A Kurtanov
- Federal State Autonomous Educational Institution of Higher Education, M K Ammosov North-Eastern Federal University, Yakutsk, Russian Federation
| |
Collapse
|
3
|
Abou Assi A, Heude B, Plancoulaine S, Sarté C, Tafflet M, Yuan WL, Charles MA, Armand M, Bernard JY. Patterns of perinatal polyunsaturated fatty acid status and associated dietary or candidate-genetic factors. J Lipid Res 2024; 65:100562. [PMID: 38762122 PMCID: PMC11231547 DOI: 10.1016/j.jlr.2024.100562] [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: 02/20/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/20/2024] Open
Abstract
Perinatal exposure to omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) can be characterized through biomarkers in maternal or cord blood or breast milk. Objectives were to describe perinatal PUFA status combining multiple biofluids and to investigate how it was influenced by dietary intake during pregnancy and maternal FADS and ELOVL gene polymorphisms. This study involved 1,901 mother-child pairs from the EDEN cohort, with PUFA levels measured in maternal and cord erythrocytes, and colostrum. Maternal dietary PUFA intake during the last trimester was derived from a food frequency questionnaire. Twelve single-nucleotide polymorphisms in FADS and ELOVL genes were genotyped from maternal DNA. Principal component analysis incorporating PUFA levels from the three biofluids identified patterns of perinatal PUFA status. Spearman's correlations explored associations between patterns and PUFA dietary intake, and linear regression models examined pattern associations with FADS or ELOVL haplotypes. Five patterns were retained: "High omega-3 LC-PUFAs, low omega-6 LC-PUFAs"; "Omega-6 LC-PUFAs"; "Colostrum LC-PUFAs"; "Omega-6 precursor (LA) and DGLA"; "Omega-6 precursor and colostrum ALA". Maternal omega-3 LC-PUFA intakes were correlated with "High omega-3 LC-PUFAs, low omega-6 LC-PUFAs" (r(DHA) = 0.33) and "Omega-6 LC-PUFAs" (r(DHA) = -0.19) patterns. Strong associations were found between FADS haplotypes and PUFA patterns except for "High omega-3 LC-PUFAs, low omega-6 LC-PUFAs". Lack of genetic association with the "High omega-3 LC-PUFAs, low omega-6 LC-PUFAs" pattern, highly correlated with maternal omega-3 LC-PUFA intake, emphasizes the importance of adequate omega-3 LC-PUFA intake during pregnancy and lactation. This study offers a more comprehensive assessment of perinatal PUFA status and its determinants.
Collapse
Affiliation(s)
- Aline Abou Assi
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France
| | - Barbara Heude
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France.
| | - Sabine Plancoulaine
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France; Université Claude Bernard Lyon 1, INSERM, CNRS, Centre de Recherche en Neurosciences de Lyon CRNL U1028 UMR5292, WAKING, Bron, France
| | | | - Muriel Tafflet
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France
| | - Wen Lun Yuan
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France; Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research (A∗STAR), Singapore, Singapore
| | - Marie-Aline Charles
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France
| | - Martine Armand
- Aix Marseille Université, CNRS, CRMBM, Marseille, France
| | - Jonathan Y Bernard
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, INRAE, Centre for Research in Epidemiology and StatisticS (CRESS), Paris, France
| |
Collapse
|
4
|
Rasaei N, Daneshzad E, Khadem A, Gholami F, Samadi M, Mirzaei K. Investigation of the interaction between genetic risk score (GRS) and fatty acid quality indices on metabolic syndrome among overweight and obese women. BMC Med Genomics 2024; 17:113. [PMID: 38685080 PMCID: PMC11057091 DOI: 10.1186/s12920-024-01838-2] [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: 02/26/2023] [Accepted: 02/27/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND AND AIM Metabolic syndrome is one of the major public-health challenges, affecting one-quarter of the world population. Fatty acid quality indices are novel determinants of this disease and their interactions with genetic factors may have an impact on metabolic syndrome risk. Therefore, we aimed to investigate the interaction between genetic risk score (GRS) and fatty acid quality indices with metabolic syndrome (MetS) among overweight and obese women. METHODS In the present cross-sectional study, 279 overweight and obese women (18-48 years old) were included. Several anthropometric measurements such as weight, height, body mass index (BMI), waist circumference (WC), and body fat percent (BF%) were measured. Also, systolic and diastolic blood pressure (SBP and DBP) were measured. Biochemical determination was performed for fasting blood glucose (FBS), triglyceride (TG), and high-density lipoprotein (HDL). MetS was determined according to National Cholesterol Education Program (NCEP ATP III) criteria. Dietary intake was evaluated by a validated and reliable 147-item semi-quantitative food frequency questionnaire. Cholesterol-saturated fat index (CSI) and the ratio of omega-6/omega-3 (ω-6/ω-3) essential fatty acids were considered as fat quality indices. The salting-out method was used to extract the total DNA. The unweighted GRS was calculated using the risk alleles of the three single nucleotide polymorphisms. The total average GRS value was 2 and the sum of the risk alleles of the 3 polymorphisms was 6. RESULT The results of our analysis showed that after controlling for age, energy intake, BMI, and physical activity, there was a positive interaction between T2 of GRS and T2 of N6/N3 ratio on WC (β = 7.95, 95%CI = 0.83,15.08, P = 0.029), T3 of GRS and T2 of N6/N3 ratio on DBP (β = 5.93, 95%CI= -0.76,12.63, P = 0.083), and FBS (β = 6.47, 95%CI = 0.59,13.53, P = 0.073), T3 of GRS and T3 of N6/N3 ratio on TG (β = 54.42, 95%CI = 1.76,107.08, P = 0.043), and T3 of GRS and T3 of CSI on BF% (β = 3.55, 95%CI= -0.35,7.45, P = 0.075). Also T2 of GRS in the interaction with T3 of CSI leads to an decrease - 8.35 mg/dl in HDL level after adjustment in (β= -8.35, 95%CI= -17.34,0.62, P = 0.068). CONCLUSION It seems the interaction of GRS and fatty acid quality indices is positively associated with several components of metabolic syndrome such as WC, TG and BF%. Our findings are of importance to public health, considering the high consumption of foods that are high on fatty acids. Conflicting evidence of many previous studies regarding the effect of fat intake and obesity and cardiovascular diseases could be because of the gene-diet interactions and genetic heterogeneity across various ethnic groups. Hence, the synergism effect of genetic and dietay intakes should be considered in future studies.
Collapse
Affiliation(s)
- Niloufar Rasaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, P. O. Box: 14155-6117, Iran
| | - Elnaz Daneshzad
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Alireza Khadem
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Gholami
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, P. O. Box: 14155-6117, Iran
| | - Mahsa Samadi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, P. O. Box: 14155-6117, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, P. O. Box: 14155-6117, Iran.
| |
Collapse
|
5
|
Zhang Y, Zhang XY, Shi SR, Ma CN, Lin YP, Song WG, Guo SD. Natural products in atherosclerosis therapy by targeting PPARs: a review focusing on lipid metabolism and inflammation. Front Cardiovasc Med 2024; 11:1372055. [PMID: 38699583 PMCID: PMC11064802 DOI: 10.3389/fcvm.2024.1372055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Inflammation and dyslipidemia are critical inducing factors of atherosclerosis. Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors and control the expression of multiple genes that are involved in lipid metabolism and inflammatory responses. However, synthesized PPAR agonists exhibit contrary therapeutic effects and various side effects in atherosclerosis therapy. Natural products are structural diversity and have a good safety. Recent studies find that natural herbs and compounds exhibit attractive therapeutic effects on atherosclerosis by alleviating hyperlipidemia and inflammation through modulation of PPARs. Importantly, the preparation of natural products generally causes significantly lower environmental pollution compared to that of synthesized chemical compounds. Therefore, it is interesting to discover novel PPAR modulator and develop alternative strategies for atherosclerosis therapy based on natural herbs and compounds. This article reviews recent findings, mainly from the year of 2020 to present, about the roles of natural herbs and compounds in regulation of PPARs and their therapeutic effects on atherosclerosis. This article provides alternative strategies and theoretical basis for atherosclerosis therapy using natural herbs and compounds by targeting PPARs, and offers valuable information for researchers that are interested in developing novel PPAR modulators.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Endocrinology and Metabolism, Guiqian International General Hospital, Guiyang, China
| | - Xue-Ying Zhang
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Shan-Rui Shi
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Chao-Nan Ma
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| | - Yun-Peng Lin
- Department of General Surgery, Qixia Traditional Chinese Medicine Hospital in Shandong Province, Yantai, China
| | - Wen-Gang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, School of Pharmacy, Shandong Second Medical University, Weifang, China
| |
Collapse
|
6
|
Bernhard B, Heydari B, Abdullah S, Francis SA, Lumish H, Wang W, Jerosch-Herold M, Harris WS, Kwong RY. Effect of six month's treatment with omega-3 acid ethyl esters on long-term outcomes after acute myocardial infarction: The OMEGA-REMODEL randomized clinical trial. Int J Cardiol 2024; 399:131698. [PMID: 38184150 DOI: 10.1016/j.ijcard.2023.131698] [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: 09/29/2023] [Revised: 12/15/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND Omega-3 polyunsaturated fatty acids (O3-FA) have been shown to reduce inflammation and adverse cardiac remodeling after acute myocardial infarction (AMI). However, the impact of O3-FA on long-term clinical outcomes remains uncertain. AIMS To investigate the impact of O3-FA on adverse cardiac events in long-term follow up post AMI in a pilot-study. METHODS Consecutive patients with AMI were randomized 1:1 to receive 6 months of O3-FA (4 g/daily) or placebo in the prospective, multicenter OMEGA-REMODEL trial. Primary endpoint was a composite of major adverse cardiovascular events (MACE) encompassing all-cause death, heart failure hospitalizations, recurrent acute coronary syndrome, and late coronary artery bypass graft (CABG). RESULTS A total of 358 patients (62.8% male; 48.1 ± 16.1 years) were followed for a median of 6.6 (IQR: 5.0-9.1) years. Among those receiving O3-FA (n = 180), MACE occurred in 65 (36.1%) compared to 62 (34.8%) of 178 assigned to placebo. By intention-to-treat analysis, O3-FA treatment assignment did not reduce MACE (HR = 1.014; 95%CI = 0.716-1.436; p = 0.938), or its individual components. However, patients with a positive response to O3-FA treatment (n = 43), defined as an increase in the red blood cell omega-3 index (O3I) ≥5% after 6 months of treatment, had lower annualized MACE rates compared to those without (2.9% (95%CI = 1.2-5.1) vs 7.1% (95%CI = 5.7-8.9); p = 0.001). This treatment benefit persisted after adjustment for baseline characteristics (HRadjusted = 0.460; 95%CI = 0.218-0.970; p = 0.041). CONCLUSION In long-term follow-up of the OMEGA-REMODEL randomized trial, O3-FA did not reduce MACE after AMI by intention to treat principle, however, patients who achieved a ≥ 5% increase of O3I subsequent to treatment had favorable outcomes.
Collapse
Affiliation(s)
- Benedikt Bernhard
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Bobak Heydari
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Stephenson Cardiac Imaging Center, University of Calgary, Calgary, Alberta, Canada
| | - Shuaib Abdullah
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; VA North Texas Medical Center and University of Texas-Southwestern Medical School, Dallas, TX, USA
| | - Sanjeev A Francis
- Department of Cardiovascular Medicine, Maine Medical Center, Portland, USA
| | - Heidi Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Wei Wang
- Division of Sleep Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Jerosch-Herold
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - William S Harris
- Fatty Acid Research Institute, Sioux Falls, SD 57106, USA; Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA
| | - Raymond Y Kwong
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
7
|
Qi L, Ye Z, Lin H. Identification of Differential Metabolites Between
Type 2 Diabetes and Postchronic Pancreatitis Diabetes (Type 3c) Based on an Untargeted Metabolomics Approach. Lab Med 2023; 54:562-573. [PMID: 36864551 DOI: 10.1093/labmed/lmad004] [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] [Indexed: 03/04/2023] Open
Abstract
OBJECTIVE A nontargeted metabolomics approach was established to characterize serum metabolic profile in type 3c diabetes mellitus (T3cDM) secondary to chronic pancreatitis and compare with T2DM. METHODS Forty patients were recruited for metabolite analysis based on liquid chromatography-mass spectrometry. Cluster heatmap and KEGG metabolic pathway enrichment analysis were used to analyze the specific and differential metabolites. The receiver operating characteristics (ROCs) were generated and correlation analysis with clinical data was conducted. RESULTS Metabolites including sphingosine, lipids, carnitine, bile acid, and hippuric acid were found to be different between T2DM and T3cDM, mainly enriched in bile acid biosynthesis, fatty acid biosynthesis, and sphingolipid metabolic pathways. The ROCs were generated with an area under the curve of 0.907 (95% confidence interval, 0.726-1) for the model with 15 metabolites. CONCLUSION T3cDM is characterized by increased sphingosine, carnitine, bile acid, and most lipids, providing novel biomarkers for clinical diagnosis and a future direction in research on pathophysiological mechanisms.
Collapse
Affiliation(s)
- Liang Qi
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zheng Ye
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hao Lin
- Department of Clinical Science and Research, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
8
|
Kim DJ, Lim JE, Jung HU, Chung JY, Baek EJ, Jung H, Kwon SY, Kim HK, Kang JO, Park K, Won S, Kim TB, Oh B. Identification of asthma-related genes using asthmatic blood eQTLs of Korean patients. BMC Med Genomics 2023; 16:259. [PMID: 37875944 PMCID: PMC10599017 DOI: 10.1186/s12920-023-01677-7] [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: 02/03/2023] [Accepted: 09/29/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND More than 200 asthma-associated genetic variants have been identified in genome-wide association studies (GWASs). Expression quantitative trait loci (eQTL) data resources can help identify causal genes of the GWAS signals, but it can be difficult to find an eQTL that reflects the disease state because most eQTL data are obtained from normal healthy subjects. METHODS We performed a blood eQTL analysis using transcriptomic and genotypic data from 433 Korean asthma patients. To identify asthma-related genes, we carried out colocalization, Summary-based Mendelian Randomization (SMR) analysis, and Transcriptome-Wide Association Study (TWAS) using the results of asthma GWASs and eQTL data. In addition, we compared the results of disease eQTL data and asthma-related genes with two normal blood eQTL data from Genotype-Tissue Expression (GTEx) project and a Japanese study. RESULTS We identified 340,274 cis-eQTL and 2,875 eGenes from asthmatic eQTL analysis. We compared the disease eQTL results with GTEx and a Japanese study and found that 64.1% of the 2,875 eGenes overlapped with the GTEx eGenes and 39.0% with the Japanese eGenes. Following the integrated analysis of the asthmatic eQTL data with asthma GWASs, using colocalization and SMR methods, we identified 15 asthma-related genes specific to the Korean asthmatic eQTL data. CONCLUSIONS We provided Korean asthmatic cis-eQTL data and identified asthma-related genes by integrating them with GWAS data. In addition, we suggested these asthma-related genes as therapeutic targets for asthma. We envisage that our findings will contribute to understanding the etiological mechanisms of asthma and provide novel therapeutic targets.
Collapse
Affiliation(s)
- Dong Jun Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Eun Lim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hae-Un Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Ju Yeon Chung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | | | - Hyein Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Shin Young Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Han Kyul Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ji-One Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kyungtaek Park
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Sungho Won
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Republic of Korea
- Department of Public Health Sciences, School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Tae-Bum Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Bermseok Oh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea.
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea.
- Mendel Inc, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Perrone B, Ruffo P, Augimeri G, Sisci D, Sinicropi MS, Tripepi G, Mammì C, Bonofiglio D, Conforti FL. Association between HMGCR, CRP, and CETP gene polymorphisms and metabolic/inflammatory serum profile in healthy adolescents. J Transl Med 2023; 21:718. [PMID: 37833739 PMCID: PMC10576320 DOI: 10.1186/s12967-023-04571-z] [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: 08/17/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND The complex interplay between health, lifestyle and genetics represents a critical area of research for understanding and promoting human well-being. Importantly, genetics plays a key role in determining individual susceptibility to disease and response to lifestyle. The aim of the present study was to identify genetic factors related to the metabolic/inflammatory profile of adolescents providing new insights into the individual predisposition to the different effects of the substances from the environment. METHODS Association analysis of genetic variants and biochemical parameters was performed in a total of 77 healthy adolescents recruited in the context of the DIMENU study. RESULTS Polymorphisms of 3-hydroxy-3-methylglutaril coenzyme A reductase (HMGCR; rs142563098), C-reactive protein gene (CRP; rs1417938, rs1130864), cholesteryl ester transfer protein (CETP; rs5030708), interleukin (IL)-10 (IL-10; rs3024509) genes were significantly associated (p < 0.05) with various serum metabolic parameters. Of particular interest were also the correlations between the HMGCRpolymorphism (rs3846663) and tumor necrosis factor (TNF)-α levels, as well Fatty-acid desaturase (FADS) polymorphism (rs7481842) and IL-10 level opening a new link between lipidic metabolism genes and inflammation. CONCLUSION In this study, we highlighted associations between single nucleotide polymorphisms (SNPs) and serum levels of metabolic and inflammatory parameters in healthy young individuals, suggesting the importance of genetic profiling in the prevention and management of chronic disease.
Collapse
Affiliation(s)
- Benedetta Perrone
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
| | - Paola Ruffo
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
| | - Giuseppina Augimeri
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
| | - Diego Sisci
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
- Centro Sanitario, University of Calabria, Via P Bucci, Rende, CS, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
| | - Giovanni Tripepi
- Institute of Clinical Physiology of Reggio Calabria, IFC-CNR, Reggio Calabria, Italy
| | - Corrado Mammì
- Great Metropolitan Hospital BMM, Reggio Calabria, Italy.
| | - Daniela Bonofiglio
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy
- Centro Sanitario, University of Calabria, Via P Bucci, Rende, CS, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, Rende, CS, Italy.
- Centro Sanitario, University of Calabria, Via P Bucci, Rende, CS, Italy.
| |
Collapse
|
10
|
Mattinzoli D, Turolo S, Ikehata M, Vettoretti S, Montini G, Agostoni C, Conti C, Benedetti M, Messa P, Alfieri CM, Castellano G. MCP1 Inverts the Correlation between FGF23 and Omega 6/3 Ratio: Is It Also True in Renal Transplantation? J Clin Med 2023; 12:5928. [PMID: 37762869 PMCID: PMC10532002 DOI: 10.3390/jcm12185928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
During chronic kidney disease (CKD) progression, an increase in fibroblast growth factor (FGF23) is present. In stage 5, a positive correlation between FGF23 and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) emerges. Hypothesizing that the rising positive correlation between monocyte chemoattractant protein 1 (MCP1) and n-6 in stage 4 could be the cause, we previously explored FGF23 and MCP1's roles in dyslipidemia and cardiovascular risk in CKD. In the present paper, we retraced the study evaluating 40 kidney transplant patients (KTx), a cohort where several factors might modify the previous relationships found. An ELISA and gas chromatography assessed the MCP1, FGF23, and PUFA levels. Despite the FGF23 increase (p < 0.0001), low MCP1 levels were found. A decrease in the n-6/n-3 ratio (p = 0.042 CKD stage 4 vs. 5) lowered by the increase in both n-3 αlinolenic (p = 0.012) and docosapentaenoic acid (p = 0.049) was observed. A negative correlation between FGF23 and the n-6/n-3 ratio in CKD stage 4 (r2 -0.3 p = 0.043) and none with MCP1 appeared. According to our findings, different mechanisms in the relationship between FGF23, PUFAs, and MCP1 in CKD and KTx patients might be present, which is possibly related to the immunosuppressive status of the last. Future research will further clarify our hypothesis.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefano Turolo
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Masami Ikehata
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Simone Vettoretti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Pediatric-Immunorheumatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Costanza Conti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, 20157 Milan, Italy
| | - Matteo Benedetti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, 20157 Milan, Italy
| | - Piergiorgio Messa
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo Maria Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
11
|
Šarac I, Debeljak-Martačić J, Takić M, Stevanović V, Milešević J, Zeković M, Popović T, Jovanović J, Vidović NK. Associations of fatty acids composition and estimated desaturase activities in erythrocyte phospholipids with biochemical and clinical indicators of cardiometabolic risk in non-diabetic Serbian women: the role of level of adiposity. Front Nutr 2023; 10:1065578. [PMID: 37545582 PMCID: PMC10397414 DOI: 10.3389/fnut.2023.1065578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Fatty acids (FAs) composition and desaturase activities can be altered in different metabolic conditions, but the adiposity-independent associations with clinical and biochemical indicators of cardiometabolic risk are still unclear. This study aimed to analyze the associations of FAs composition and estimated desaturase activities with anthropometric, clinical, and biochemical cardiometabolic risk indicators in non-diabetic Serbian women, and to investigate if these associations were independent of the level of adiposity and other confounders. Methods In 76 non-diabetic, otherwise healthy Serbian women, aged 24-68 years, with or without metabolic syndrome or obesity (BMI=23.6±5.6 kg/m2), FA composition in erythrocyte phospholipids was measured by gas-liquid chromatography. Desaturase activities were estimated from product/precursor FAs ratios (D9D:16:1n-7/16:0; D6D:20:3n-6/18:2n-6; D5D:20:4n-6/20:3n-6). Correlations were made with anthropometric, biochemical (serum glucose, triacylglycerols, LDL-C, HDL-C, ALT, AST, and their ratios) and clinical (blood pressure) indicators of cardiometabolic risk. Linear regression models were performed to test the independence of these associations. Results Estimated desaturase activities and certain FAs were associated with anthropometric, clinical and biochemical indicators of cardiometabolic risk: D9D, D6D, 16:1n-7 and 20:3n-6 were directly associated, while D5D and 18:0 were inversely associated. However, the associations with clinical and biochemical indicators were not independent of the associations with the level of adiposity, since they were lost after controlling for anthropometric indices. After controlling for multiple confounders (age, postmenopausal status, education, smoking, physical activity, dietary macronutrient intakes, use of supplements, alcohol consumption), the level of adiposity was the most significant predictor of desaturase activities and aforementioned FAs levels, and mediated their association with biochemical/clinical indicators. Vice versa, desaturase activities predicted the level of adiposity, but not other components of cardiometabolic risk (if the level of adiposity was accounted). While the associations of anthropometric indices with 16:1n-7, 20:3n-6, 18:0 and D9D and D6D activities were linear, the associations with D5D activity were the inverse U-shaped. The only adiposity-independent association of FAs profiles with the indicators of cardiometabolic risk was a positive association of 20:5n-3 with ALT/AST ratio, which requires further exploration. Discussion Additional studies are needed to explore the mechanisms of the observed associations.
Collapse
Affiliation(s)
- Ivana Šarac
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Debeljak-Martačić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marija Takić
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vuk Stevanović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jelena Milešević
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Zeković
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Popović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jovica Jovanović
- Department of Occupational Health, Faculty of Medicine, University of Niš, Niš, Serbia
| | - Nevena Kardum Vidović
- Centre of Research Excellence in Nutrition and Metabolism, Group for Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
12
|
Huang L, Zhang F, Xu P, Zhou Y, Liu Y, Zhang H, Tan X, Ge X, Xu Y, Guo M, Long Y. Effect of Omega-3 Polyunsaturated Fatty Acids on Cardiovascular Outcomes in Patients with Diabetes: A Meta-analysis of Randomized Controlled Trials. Adv Nutr 2023; 14:629-636. [PMID: 37121469 PMCID: PMC10334152 DOI: 10.1016/j.advnut.2023.04.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023] Open
Abstract
The current guidelines recommend that people consume 2 or more servings of fat-rich fish per week to obtain enough omega-3 (ω-3) polyunsaturated fatty acids to prevent cardiovascular events. However, the cardiovascular benefits of ω-3 polyunsaturated fatty acids in patients with diabetes are unclear, and related large-scale trials have produced conflicting results. We aimed to perform a meta-analysis of all randomized controlled trials that attempted to assess the effects of ω-3 fatty acid supplementation on cardiovascular outcomes in patients with diabetes. In PubMed, EMBASE, and the Cochrane Library, we searched for data from all randomized controlled trials on ω-3 fatty acids and cardiovascular outcomes in patients with diabetes published before July 2022. Eight eligible studies involving 57,754 participants were ultimately included. Meta-analysis showed that ω-3 fatty acid supplementation reduces cardiovascular disease (CVD) risk in patients with diabetes (rate ration [RR] = 0.93; 95% confidence interval [CI]: 0.90, 0.97; P = 0.0009). Among them, eicosapentaenoic acid (EPA), but not EPA plus docosahexaenoic acid (DHA), significantly reduced the risk of CVD in patients with diabetes (EPA [RR = 0.81; 95% CI: 0.73, 0.90; P=0.0001]). This meta-analysis suggests that ω-3 fatty acid supplementation is an effective strategy to prevent CVD in patients with diabetes, but further well-designed, large-scale randomized controlled trials are necessary to evaluate the safety of ω-3 fatty acid supplementation, and its effect on atrial fibrillation. This study was registered with PROSPERO as CRD42022346302.
Collapse
Affiliation(s)
- Linlin Huang
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Fanjie Zhang
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Ping Xu
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yijie Zhou
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yijun Liu
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hongdie Zhang
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaozhen Tan
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Xinxu Ge
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China
| | - Man Guo
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| | - Yang Long
- Experimental Medicine Center, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China; Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, Sichuan, China; Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
13
|
Zhao J, Yang Q, Liu Z, Xu P, Tian L, Yan J, Li K, Lin B, Bian L, Xi Z, Liu X. The impact of subchronic ozone exposure on serum metabolome and the mechanisms of abnormal bile acid and arachidonic acid metabolisms in the liver. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114573. [PMID: 36701875 DOI: 10.1016/j.ecoenv.2023.114573] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 06/17/2023]
Abstract
Ambient ozone (O3) pollution can induce respiratory and cardiovascular toxicity. However, its impact on the metabolome and the underlying mechanisms remain unclear. This study first investigated the serum metabolite changes in rats exposed to 0.5 ppm O3 for 3 months using untargeted metabolomic approach. Results showed chronic ozone exposure significantly altered the serum levels of 34 metabolites with potential increased risk of digestive, respiratory and cardiovascular disease. Moreover, bile acid synthesis and secretion, and arachidonic acid (AA) metabolism became the most prominent affected metabolic pathways after O3 exposure. Further studies on the mechanisms found that the elevated serum toxic bile acid was not due to the increased biosynthesis in the liver, but the reduced reuptake from the portal vein to hepatocytes owing to repressed Ntcp and Oatp1a1, and the decreased bile acid efflux in hepatocytes as a results of inhibited Bsep, Ostalpha and Ostbeta. Meanwhile, decreased expressions of detoxification enzyme of SULT2A1 and the important regulators of FXR, PXR and HNF4α also contributed to the abnormal bile acids. In addition, O3 promoted the conversion of AA into thromboxane A2 (TXA2) and 20-hydroxyarachidonic acid (20-HETE) in the liver by up-regulation of Fads2, Cyp4a and Tbxas1 which resulting in decreased AA and linoleic acid (LA), and increased thromboxane B2 (TXB2) and 20-HETE in the serum. Furthermore, apparent hepatic chronic inflammation, fibrosis and abnormal function were found in ozone-exposed rats. These results indicated chronic ozone exposure could alter serum metabolites by interfering their metabolism in the liver, and inducing liver injury to aggravate metabolic disorders.
Collapse
Affiliation(s)
- Jiao Zhao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Qingcheng Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Zhiyuan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Pengfei Xu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Liping Bian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Exercise Physiology and Sports Medicine, Tianjin Sport University, Tianjin 301617, China.
| |
Collapse
|
14
|
Dong Q, Sidra S, Gieger C, Wang-Sattler R, Rathmann W, Prehn C, Adamski J, Koenig W, Peters A, Grallert H, Sharma S. Metabolic Signatures Elucidate the Effect of Body Mass Index on Type 2 Diabetes. Metabolites 2023; 13:metabo13020227. [PMID: 36837846 PMCID: PMC9965667 DOI: 10.3390/metabo13020227] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Obesity plays an important role in the development of insulin resistance and diabetes, but the molecular mechanism that links obesity and diabetes is still not completely understood. Here, we used 146 targeted metabolomic profiles from the German KORA FF4 cohort consisting of 1715 participants and associated them with obesity and type 2 diabetes. In the basic model, 83 and 51 metabolites were significantly associated with body mass index (BMI) and T2D, respectively. Those metabolites are branched-chain amino acids, acylcarnitines, lysophospholipids, or phosphatidylcholines. In the full model, 42 and 3 metabolites were significantly associated with BMI and T2D, respectively, and replicate findings in the previous studies. Sobel mediation testing suggests that the effect of BMI on T2D might be mediated via lipids such as sphingomyelin (SM) C16:1, SM C18:1 and diacylphosphatidylcholine (PC aa) C38:3. Moreover, mendelian randomization suggests a causal relationship that BMI causes the change of SM C16:1 and PC aa C38:3, and the change of SM C16:1, SM C18:1, and PC aa C38:3 contribute to T2D incident. Biological pathway analysis in combination with genetics and mice experiments indicate that downregulation of sphingolipid or upregulation of phosphatidylcholine metabolism is a causal factor in early-stage T2D pathophysiology. Our findings indicate that metabolites like SM C16:1, SM C18:1, and PC aa C38:3 mediate the effect of BMI on T2D and elucidate their role in obesity related T2D pathologies.
Collapse
Affiliation(s)
- Qiuling Dong
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Faculty of Medicine, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Sidra Sidra
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Cornelia Prehn
- Metabolomics and Proteomics Core Facility, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Wolfgang Koenig
- German Research Center for Cardiovascular Disease (DZHK), Partner site Munich Heart Alliance, 81377 Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, 81377 Munich, Germany
- Institute of Epidemiology and Medical Biometry, University of Ulm, 89069 Ulm, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Chair of Epidemiology, Faculty of Medicine, Ludwig-Maximilians-University München, 81377 Munich, Germany
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
- Correspondence: (H.G.); (S.S.)
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Chair of Food Chemistry and Molecular Sensory Science, Technical University of Munich, 85354 Freising-Weihenstephan, Germany
- Correspondence: (H.G.); (S.S.)
| |
Collapse
|
15
|
Wuni R, Ventura EF, Curi-Quinto K, Murray C, Nunes R, Lovegrove JA, Penny M, Favara M, Sanchez A, Vimaleswaran KS. Interactions between genetic and lifestyle factors on cardiometabolic disease-related outcomes in Latin American and Caribbean populations: A systematic review. Front Nutr 2023; 10:1067033. [PMID: 36776603 PMCID: PMC9909204 DOI: 10.3389/fnut.2023.1067033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction The prevalence of cardiometabolic diseases has increased in Latin American and the Caribbean populations (LACP). To identify gene-lifestyle interactions that modify the risk of cardiometabolic diseases in LACP, a systematic search using 11 search engines was conducted up to May 2022. Methods Eligible studies were observational and interventional studies in either English, Spanish, or Portuguese. A total of 26,171 publications were screened for title and abstract; of these, 101 potential studies were evaluated for eligibility, and 74 articles were included in this study following full-text screening and risk of bias assessment. The Appraisal tool for Cross-Sectional Studies (AXIS) and the Risk Of Bias In Non-Randomized Studies-of Interventions (ROBINS-I) assessment tool were used to assess the methodological quality and risk of bias of the included studies. Results We identified 122 significant interactions between genetic and lifestyle factors on cardiometabolic traits and the vast majority of studies come from Brazil (29), Mexico (15) and Costa Rica (12) with FTO, APOE, and TCF7L2 being the most studied genes. The results of the gene-lifestyle interactions suggest effects which are population-, gender-, and ethnic-specific. Most of the gene-lifestyle interactions were conducted once, necessitating replication to reinforce these results. Discussion The findings of this review indicate that 27 out of 33 LACP have not conducted gene-lifestyle interaction studies and only five studies have been undertaken in low-socioeconomic settings. Most of the studies were cross-sectional, indicating a need for longitudinal/prospective studies. Future gene-lifestyle interaction studies will need to replicate primary research of already studied genetic variants to enable comparison, and to explore the interactions between genetic and other lifestyle factors such as those conditioned by socioeconomic factors and the built environment. The protocol has been registered on PROSPERO, number CRD42022308488. Systematic review registration https://clinicaltrials.gov, identifier CRD420223 08488.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | - Eduard F. Ventura
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | | | - Claudia Murray
- Department of Real Estate and Planning, University of Reading, Reading, United Kingdom
| | - Richard Nunes
- Department of Real Estate and Planning, University of Reading, Reading, United Kingdom
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | - Mary Penny
- Instituto de Investigación Nutricional, Lima, Peru
| | - Marta Favara
- Oxford Department of International Development, University of Oxford, Oxford, United Kingdom
| | - Alan Sanchez
- Grupo de Análisis para el Desarrollo (GRADE), Lima, Peru
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading, United Kingdom
| |
Collapse
|
16
|
Shi H, Prough RA, McClain CJ, Song M. Different Types of Dietary Fat and Fructose Interactions Result in Distinct Metabolic Phenotypes in Male Mice. J Nutr Biochem 2023; 111:109189. [PMID: 36272691 DOI: 10.1016/j.jnutbio.2022.109189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/19/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Fat and fructose are the two major components over-represented in the Western diet. The aim of this study was to determine the combined effects of different types of dietary fat and fructose on the development of nonalcoholic fatty liver disease (NAFLD) in a murine model. Eight-week-old male C57BL/6J mice were fed with high-fat diet enriched with saturated fat (HSF), or omega-6 polyunsaturated fat (n6HUSF), or omega-3 polyunsaturated fat (n3HUSF) with 42% of calories derived from the fat. Fructose supplementation was given via 10% fructose (w/v) in the drinking water ad libitum for 20 weeks. While both HSF and n6HUSF fed mice developed obesity, HSF fed mice exhibited severe hepatic steatosis associated with hepatomegaly and liver injury. Fructose feeding promotes the development of liver fibrosis in HSF fed mice. n6HUSF fed mice were characterized with moderate hepatic steatosis, accompanied with hypertriglyceridemia and hyperlipidemia. Notably, fructose supplementation led to remarkable glucose intolerance in n6HUSF fed mice compared to controls. Hepatic lipidomic analysis revealed that the total saturated fatty acids and total monounsaturated fatty acids were significantly increased by fructose in the free fatty acid pool in HSF fed mice. Moreover, fructose supplementation increased hepatic and plasma cholesterol levels in the HSF fed mice. Our data suggest that excess energy from HSF intake results in fat storage in the liver, likely due to impaired triglyceride secretion; whereas excess energy from n6HUSF diet is stored in the periphery. Both effects are exacerbated by fructose supplementation. n3HUSF is beneficial, even consumed with fructose.
Collapse
Affiliation(s)
- Hongxue Shi
- Department of Pharmacology and Toxicology; Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Russell A Prough
- Hepatobiology and Toxicology Center; Department of Biochemistry and Molecular Genetics
| | - Craig J McClain
- Department of Pharmacology and Toxicology; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition; Hepatobiology and Toxicology Center; University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, Kentucky, USA; Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| | - Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition; Hepatobiology and Toxicology Center.
| |
Collapse
|
17
|
Ali S, Aiello A, Zotti T, Accardi G, Cardinale G, Vito P, Calabrò A, Ligotti ME, Intrieri M, Corbi G, Caruso C, Candore G, Scapagnini G, Davinelli S. Age-associated changes in circulatory fatty acids: new insights on adults and long-lived individuals. GeroScience 2022; 45:781-796. [PMID: 36449220 PMCID: PMC9886696 DOI: 10.1007/s11357-022-00696-z] [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: 06/14/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Long-lived individuals (LLIs) are considered an ideal model to study healthy human aging. Blood fatty acid (FA) profile of a cohort of LLIs (90-111 years old, n = 49) from Sicily was compared to adults (18-64 years old, n = 69) and older adults (65-89 years old, n = 54) from the same area. Genetic variants in key enzymes related to FA biosynthesis and metabolism were also genotyped to investigate a potential genetic predisposition in determining the FA profile. Gas chromatography was employed to determine the FA profile, and genotyping was performed using high-resolution melt (HRM) analysis. Blood levels of total polyunsaturated FA (PUFA) and total trans-FA decreased with age, while the levels of saturated FA (SFA) remained unchanged. Interestingly, distinctively higher circulatory levels of monounsaturated FA (MUFA) in LLIs compared to adults and older adults were observed. In addition, among LLIs, rs174537 in the FA desaturase 1/2 (FADS1/2) gene was associated with linoleic acid (LA, 18:2n-6) and docosatetraenoic acid (DTA, 22:4n-6) levels, and the rs953413 in the elongase of very long FA 2 (ELOVL2) was associated with DTA levels. We further observed that rs174579 and rs174626 genotypes in FADS1/2 significantly affect delta-6 desaturase (D6D) activity. In conclusion, our results suggest that the LLIs have a different FA profile characterized by high MUFA content, which indicates reduced peroxidation while maintaining membrane fluidity.
Collapse
Affiliation(s)
- Sawan Ali
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via De Sanctis 1, Campobasso, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Tiziana Zotti
- Dipartimento Di Scienze E Tecnologie, Università Degli Studi del Sannio, Benevento, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | - Pasquale Vito
- Dipartimento Di Scienze E Tecnologie, Università Degli Studi del Sannio, Benevento, Italy
| | - Anna Calabrò
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Mariano Intrieri
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via De Sanctis 1, Campobasso, Italy
| | - Graziamaria Corbi
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via De Sanctis 1, Campobasso, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via De Sanctis 1, Campobasso, Italy.
| | - Sergio Davinelli
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Via De Sanctis 1, Campobasso, Italy
| |
Collapse
|
18
|
In Vitro Gene Expression Responses of Bovine Rumen Epithelial Cells to Different pH Stresses. Animals (Basel) 2022; 12:ani12192621. [PMID: 36230362 PMCID: PMC9559271 DOI: 10.3390/ani12192621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/17/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Ruminal acidosis often occurs in production, which greatly affects animal health and production efficiency. Subacute rumen acidosis (SARA) occurs when rumen pH drops rapidly to 5.5−5.8, and acute rumen acidosis (ARA) occurs when rumen pH drops below 5.0, but the molecular regulation mechanism of the rumen epithelium after the rapid decrease in pH is still unclear. Bovine rumen epithelial cells (BRECs) were cultured at pH = 7.4 (control), 5.5 (SARA), and 4.5 (ARA). Transcriptome and metabolomic methods were used to obtain the molecular-based response of BRECs to different pH treatments; pH = 4.5 can significantly induce apoptosis of BRECs. The RNA-seq experiments revealed 1381 differently expressed genes (DEGs) in the control vs. SARA groups (p < 0.05). Fibroblast growth factor (FGF) and tumor necrosis factor (TNF) were upregulated 4.25 and 6.86 fold, respectively, and TLR4 was downregulated 0.58 fold. In addition, 283 DEGs were identified in the control vs. ARA comparison (p < 0.05), and prostaglandin-endoperoxide synthase 2 (PSTG2) was downregulated 0.54 fold. Our research reveals that the MAPK/TNF signaling pathway regulates the inflammatory response of BRECs. Metabolomics identified 35 biochemical compounds that were significantly affected (p < 0.05) in control vs. SARA and 51 in control vs. ARA. Bioinformatics analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database revealed that drug metabolism-cytochrome P450 metabolic and alpha-linolenic acid metabolism changes occurred. These transcriptional and metabolic changes are related to the adaptation of BRECs to low-pH stresses. In conclusion, the combined data analyses presented a worthy strategy to characterize the cellular, transcriptomic, and metabonomic adaptation of BRECs to pH in vitro. We demonstrated transcriptional expression changes in BRECs under pH stress and activation of the molecular mechanisms controlling inflammation.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW HIV and antiretroviral therapy (ART) use are linked to an increased incidence of atherosclerotic cardiovascular disease (ASCVD). Immune activation persists in ART-treated people with HIV (PWH), and markers of inflammation (i.e. IL-6, C-reactive protein) predict mortality in this population. This review discusses underlying mechanisms that likely contribute to inflammation and the development of ASCVD in PWH. RECENT FINDINGS Persistent inflammation contributes to accelerated ASCVD in HIV and several new insights into the underlying immunologic mechanisms of chronic inflammation in PWH have been made (e.g. clonal haematopoiesis, trained immunity, lipidomics). We will also highlight potential pro-inflammatory mechanisms that may differ in vulnerable populations, including women, minorities and children. SUMMARY Mechanistic studies into the drivers of chronic inflammation in PWH are ongoing and may aid in tailoring effective therapeutic strategies that can reduce ASCVD risk in this population. Focus should also include factors that lead to persistent disparities in HIV care and comorbidities, including sex as a biological factor and social determinants of health. It remains unclear whether ASCVD progression in HIV is driven by unique mediators (HIV itself, ART, immunodeficiency), or if it is an accelerated version of disease progression seen in the general population.
Collapse
Affiliation(s)
- Sahera Dirajlal-Fargo
- Rainbow Babies and Children’s Hospital, Cleveland, OH
- Case Western Reserve University, Cleveland, OH
| | - Nicholas Funderburg
- School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH
| |
Collapse
|
20
|
Zhou C, Zhang W, Lin H, Zhang L, Wu F, Wang Y, Yu S, Peng X, Cheng W, Li M, Pan X, Huang Z, Zhang W. Effect of theaflavin-3,3′-digallate on leptin-deficient induced nonalcoholic fatty liver disease might be related to lipid metabolism regulated by the Fads1/PPARδ/Fabp4 axis and gut microbiota. Front Pharmacol 2022; 13:925264. [PMID: 36105184 PMCID: PMC9464872 DOI: 10.3389/fphar.2022.925264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), one of the risk factors for hepatitis, cirrhosis, and even hepatic carcinoma, has been a global public health problem. The polyphenol compound theaflavin-3,3′-digallate (TF3), mainly extracted from black tea, has been reported to produce an effect on hypoglycemic and antilipid deposition in vitro. In our study, we further investigated the function and novel mechanisms of TF3 in protecting NAFLD in vivo. By using leptin-deficient obese (ob/ob) mice with NAFLD symptoms, TF3 treatment prevented body weight and waistline gain, reduced lipid accumulation, and alleviated liver function injury, as well as decreased serum lipid levels and TG levels in livers in ob/ob mice, observing no side effects. Furthermore, the transcriptome sequencing of liver tissue showed that TF3 treatment corrected the expression profiles of livers in ob/ob mice compared with that of the model group. It is interesting to note that TF3 might regulate lipid metabolism via the Fads1/PPARδ/Fabp4 axis. In addition, 16S rRNA sequencing demonstrated that TF3 increased the abundance of Prevotellaceae_UCG-001, norank_f_Ruminococcaceae, and GCA-900066575 and significantly decreased that of Parvibacter. Taken together, the effect of TF3 on NAFLD might be related to lipid metabolism regulated by the Fads1/PPARδ/Fabp4 axis and gut microbiota. TF3 might be a promising candidate for NAFLD therapy.
Collapse
Affiliation(s)
- Cheng Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenji Zhang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hui Lin
- Department of Radiation Oncology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Luyun Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Fan Wu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Yan Wang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Susu Yu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xinyue Peng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenli Cheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Min Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xiaoying Pan
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zhenrui Huang
- Guangdong Provincial Engineering & Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Key Laboratory of Crop Genetic Improvement of Guangdong Province, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- *Correspondence: Zhenrui Huang, ; Wenjuan Zhang,
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Zhenrui Huang, ; Wenjuan Zhang,
| |
Collapse
|
21
|
Heravi G, Jang H, Wang X, Long Z, Peng Z, Kim S, Liu W. Fatty acid desaturase 1 (FADS1) is a cancer marker for patient survival and a potential novel target for precision cancer treatment. Front Oncol 2022; 12:942798. [PMID: 36046053 PMCID: PMC9423679 DOI: 10.3389/fonc.2022.942798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Fatty Acid Desaturase-1 (FADS1) or delta 5 desaturase (D5D) is a rate-limiting enzyme involved in the biosynthesis of long-chain polyunsaturated fatty acids (LC-PUFAs), i.e., arachidonic acid (ARA) and eicosapentaenoic (EPA). These LC-PUFAs and their metabolites play essential and broad roles in cancer cell proliferation, metastasis, and tumor microenvironment. However, the role of FADS1 in cancers remains incompletely understood. Utilizing The Cancer Genome Atlas (TCGA) database, we explored the role of FADS1 across different cancer types using multiple bioinformatics and statistical tools. Moreover, we studied the impact of a FADS1 inhibitor (D5D-IN-326) on proliferation of multiple cancer cell lines. We identified that FADS1 gene is a predictor for cancer survival in multiple cancer types. Compared to normal tissue, the mRNA expression of FADS1 is significantly increased in primary tumors while even higher in metastatic and recurrent tumors. Mechanistically, pathway analysis demonstrated that FADS1 is associated with cholesterol biosynthesis and cell cycle control genes. Interestingly, FADS1 expression is higher when TP53 is mutated. Tumors with increased FADS1 expression also demonstrated an increased signatures of fibroblasts and macrophages infiltration among most cancer types. Our in vitro assays showed that D5D-IN-326 significantly inhibited cell proliferation of kidney, colon, breast, and lung cancer cell lines in a dose-dependent manner. Lastly, single nucleotide polymorphisms (SNPs) which are well-established expression quantitative trait loci (eQTLs) for FADS1 in normal human tissues are also significantly correlated with FADS1 expression in tumors of multiple tissue types, potentially serving as a marker to stratify cancer patients with high/low FADS1 expression in their tumor tissue. Our study suggests that FADS1 plays multiple roles in cancer biology and is potentially a novel target for precision cancer treatment.
Collapse
Affiliation(s)
- Gioia Heravi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, United States
| | - Hyejeong Jang
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Xiaokun Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, United States
| | - Ze Long
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, United States
| | - Zheyun Peng
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, United States
| | - Seongho Kim
- Biostatistics and Bioinformatics Core, Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, United States
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
22
|
Žák A, Jáchymová M, Burda M, Staňková B, Zeman M, Slabý A, Vecka M, Šeda O. FADS Polymorphisms Affect the Clinical and Biochemical Phenotypes of Metabolic Syndrome. Metabolites 2022; 12:metabo12060568. [PMID: 35736500 PMCID: PMC9228863 DOI: 10.3390/metabo12060568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 12/04/2022] Open
Abstract
Long-chain polyunsaturated fatty acids (LC-PUFAs) play important roles in human health, from controlling inflammation to lipid and glucose homeostasis. In our previous study, which employed a cluster analysis of a plasma fatty acid (FA) pattern, we identified two clusters of metabolic syndrome (MetS) independent of clinical and biochemical parameters within the whole study group (controls together with metabolic syndrome (MetS) patients). FA desaturase (FADS) genes are the key regulators of LC-PUFA metabolism. The aim of this study was to analyze associations between FADS polymorphisms and clusters of MetS. The study group consisted of 188 controls and 166 patients with MetS. The first cluster contained 71 controls (CON1) and 109 MetS patients (MetS1). The second cluster consisted of 117 controls (CON2) and 57 MetS patients (MetS2). In comparison with MetS2, cluster MetS1 displayed a more adverse risk profile. Cluster CON1 had, in comparison with CON2, higher body weight and increased triacylglycerol levels (p < 0.05). We found that the FADS rs174537 (p < 0.001), rs174570 (p < 0.01), and rs174602 (p < 0.05) polymorphisms along with two inferred haplotypes had statistically significant genotype associations with the splitting of MetS into MetS1 and MetS2. Conversely, we observed no significant differences in the distribution of FADS polymorphisms between MetS and CON subjects, or between CON1 and CON2. These associations between FADS polymorphisms and two clusters of MetS (differing in waist circumference, HOMA-IR, lipolysis, and oxidative stress) implicate the important influence of genetic factors on the phenotypic manifestation of MetS.
Collapse
Affiliation(s)
- Aleš Žák
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Marie Jáchymová
- Institute of Clinical Chemistry and Laboratory Diagnostics, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic;
| | - Michal Burda
- Institute for Research and Applications of Fuzzy Modeling, University of Ostrava, 701 03 Ostrava, Czech Republic;
| | - Barbora Staňková
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Miroslav Zeman
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Adolf Slabý
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
| | - Marek Vecka
- 4th Department of Medicine, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic; (A.Ž.); (B.S.); (M.Z.); (A.S.)
- Institute of Clinical Chemistry and Laboratory Diagnostics, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 08 Prague, Czech Republic;
- Correspondence:
| | - Ondřej Šeda
- Institute of Biology and Medical Genetics, 1st Medical Faculty, Charles University and the General University Hospital in Prague, 128 00 Prague, Czech Republic;
| |
Collapse
|
23
|
Niwa S, Kawabata T, Shoji K, Ogata H, Kagawa Y, Nakayama K, Yanagisawa Y, Iwamoto S, Tatsuta N, Asato K, Arima T, Yaegashi N, Nakai K. Investigation of Maternal Diet and FADS1 Polymorphism Associated with Long-Chain Polyunsaturated Fatty Acid Compositions in Human Milk. Nutrients 2022; 14:nu14102160. [PMID: 35631303 PMCID: PMC9143760 DOI: 10.3390/nu14102160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
Increasing the amount of long-chain polyunsaturated fatty acids (LCPUFA) in human milk is an important strategy for infant growth and development. We investigated the associations of LCPUFA compositions in human milk with maternal diet (especially fish and shellfish intake), with fatty acid Δ5 desaturase gene (FADS1) polymorphisms, and with gene-diet interactions. The present study was performed as part of an adjunct study of the Japan Environment and Children’s Study. The participants were 304 lactating females, who provided human milk 6−7 months after delivery. Fatty acids in human milk were analyzed by gas chromatography, and dietary surveys were conducted using a brief self-administered diet history questionnaire. We also analyzed a single nucleotide polymorphism of FADS1 (rs174547, T/C). There was a significant difference in arachidonic acid (ARA) composition in human milk among the genotype groups, and the values were decreasing in the order of TT > TC > CC. The concentrations of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) were also different between TT and CC genotype, indicating a tendency for decreasing values in the same order. The composition of ARA showed significant gene−dietary interactions in multiple regression analysis, and the positive correlation between fish and shellfish intake and ARA composition in human milk was significant only in the CC genotype. Moreover, the factor most strongly associated with EPA and DHA composition in human milk was fish and shellfish intake. Therefore, it was suggested that increasing fish and shellfish intake in mothers may increase EPA and DHA composition in human milk, while increasing fish and shellfish intake in CC genotype mothers may lead to increased ARA composition in human milk.
Collapse
Affiliation(s)
- Sakurako Niwa
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
- Faculty of Home Economics, Gifu Women’s University, 80 Taromaru, Gifu City 501-2592, Gifu, Japan
- Correspondence: ; Tel.: +81-49-282-3705
| | - Terue Kawabata
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Kumiko Shoji
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Hiromitsu Ogata
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Yasuo Kagawa
- Faculty of Nutrition, Kagawa Nutrition University, 3-9-21 Chiyoda, Sakado 350-0288, Saitama, Japan; (T.K.); (K.S.); (H.O.); (Y.K.)
| | - Kazuhiro Nakayama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8562, Chiba, Japan;
| | - Yoshiko Yanagisawa
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan; (Y.Y.); (S.I.)
| | - Sadahiko Iwamoto
- Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke 329-0498, Tochigi, Japan; (Y.Y.); (S.I.)
| | - Nozomi Tatsuta
- Department of Development and Environmental Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (N.T.); (K.A.); (K.N.)
| | - Kaname Asato
- Department of Development and Environmental Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (N.T.); (K.A.); (K.N.)
| | - Takahiro Arima
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (T.A.); (N.Y.)
| | - Nobuo Yaegashi
- Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (T.A.); (N.Y.)
| | - Kunihiko Nakai
- Department of Development and Environmental Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Miyagi, Japan; (N.T.); (K.A.); (K.N.)
- School of Sport and Health Science, Tokai Gakuen University, Nishinohora 21-233, Miyoshi 470-0207, Aichi, Japan
| |
Collapse
|
24
|
Suh JH, Zyba SJ, Shigenaga M, McDonald CM, King JC. Marginal Zinc Deficiency Alters Essential Fatty Acid Metabolism in Healthy Men. J Nutr 2022; 152:671-679. [PMID: 34919682 PMCID: PMC9076996 DOI: 10.1093/jn/nxab425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/29/2021] [Accepted: 12/09/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Rice biofortification with Zinc (Zn) can improve the Zn status of rice-consuming populations. However, the metabolic impact in humans consuming Zn-biofortified rice is unknown. OBJECTIVES To determine the effects of Zn-biofortified rice on lipid metabolism in normolipidemic men. METHODS The men consumed a rice-based diet containing 6 mg Zn/d and 1.5 g phytate (phytate/Zn ratio = 44) for 2 wk followed by a 10-mg Zn/d diet without phytate for 4 wk. An ad libitum diet supplemented with 25 mg Zn/d was then fed for 3 wk. Fasting blood samples were taken at baseline and at the end of each metabolic period for measuring plasma zinc, glucose, insulin, triglyceride (TG), LDL and HDL cholesterol, fatty acids, oxylipins, and fatty acid desaturase activities. Statistical differences were assessed by linear mixed model. RESULTS Fatty acid desaturase (FADS) 1 activity decreased by 29.1% (P = 0.007) when the 6-mg Zn/d diet was consumed for 2 wk. This change was associated with significant decreases in HDL and LDL cholesterol. The alterations in FADS1, HDL cholesterol, and TG remained unchanged when Zn intakes were increased to 10 mg/d for 4 wk. Supplementation with 25 mg Zn/d for 3 wk normalized these metabolic changes and significantly increased LDL cholesterol at the end of this metabolic period compared with baseline. FADS1 activity was inversely correlated with FADS2 (rmcorr = -0.52; P = 0.001) and TG (rmcorr = -0.55; P = 0.001) at all time points. CONCLUSIONS A low-zinc, high-phytate rice-based diet reduced plasma HDL cholesterol concentrations and altered fatty acid profiles in healthy men within 2 wk. Consuming 10 mg Zn/d without phytate for 4 wk did not improve the lipid profiles, but a 25-mg Zn/d supplement corrects these alterations in lipid metabolism within 3 wk.
Collapse
Affiliation(s)
- Jung H Suh
- UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | - Sarah J Zyba
- UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, CA, USA
- Institute for Global Nutrition Department of Nutrition University of California, Davis, CA, USA
| | - Mark Shigenaga
- UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, CA, USA
| | | | - Janet C King
- UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| |
Collapse
|
25
|
FADS1 and FADS2 Gene Polymorphisms Modulate the Relationship of Omega-3 and Omega-6 Fatty Acid Plasma Concentrations in Gestational Weight Gain: A NISAMI Cohort Study. Nutrients 2022; 14:nu14051056. [PMID: 35268031 PMCID: PMC8912382 DOI: 10.3390/nu14051056] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
The polymorphisms of fatty acid desaturase genes FADS1 and FADS2 have been associated with an increase in weight gain. We investigated FADS1 and FADS2 gene polymorphisms and the relation between ω-3 and ω-6 fatty acid plasma concentrations and gestational weight gain. A prospective cohort study of 199 pregnant women was followed in Santo Antônio de Jesus, Brazil. Plasma levels of polyunsaturated fatty acids (PUFAs) were measured at baseline and gestational weight gain during the first, second, and third trimesters. Fatty acid recognition was carried out with the aid of gas chromatography. Single nucleotide polymorphisms (SNPs) were genotyped using real-time PCR. Statistical analyses included Structural Equation Modelling. A direct effect of FADS1 and FADS2 gene polymorphisms on gestational weight was observed; however, only the SNP rs174575 (FADS2) showed a significant positive direct effect on weight over the course of the pregnancy (0.106; p = 0.016). In terms of the influence of SNPs on plasma levels of PUFAs, it was found that SNP rs174561 (FADS1) and SNP rs174575 (FADS2) showed direct adverse effects on plasma concentrations of ω-3 (eicosapentaenoic acid and alpha-linoleic acid), and only SNP rs174575 had positive direct effects on plasma levels of ARA and the ARA/LA (arachidonic acid/linoleic acid) ratio, ω-6 products, while the SNP rs3834458 (FADS2) had an adverse effect on plasma concentrations of EPA, leading to its increase. Pregnant women who were heterozygous and homozygous for the minor allele of the SNP rs3834458 (FADS2), on the other hand, showed larger concentrations of series ω-3 substrates, which indicates a protective factor for women’s health.
Collapse
|
26
|
Chilton FH, Manichaikul A, Yang C, O'Connor TD, Johnstone LM, Blomquist S, Schembre SM, Sergeant S, Zec M, Tsai MY, Rich SS, Bridgewater SJ, Mathias RA, Hallmark B. Interpreting Clinical Trials With Omega-3 Supplements in the Context of Ancestry and FADS Genetic Variation. Front Nutr 2022; 8:808054. [PMID: 35211495 PMCID: PMC8861490 DOI: 10.3389/fnut.2021.808054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022] Open
Abstract
Human diets in developed countries such as the US have changed dramatically over the past 75 years, leading to increased obesity, inflammation, and cardiometabolic dysfunction. Evidence over the past decade indicates that the interaction of genetic variation with changes in the intake of 18-carbon essential dietary omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA), linoleic acid (LA) and α-linolenic acid (ALA), respectively, has impacted numerous molecular and clinical phenotypes. Interactions are particularly relevant with the FADS1 and FADS2 genes, which encode key fatty acid desaturases in the pathway that converts LA and ALA to their long chain (≥20 carbons), highly unsaturated fatty acid (HUFA) counterparts. These gene by nutrient interactions affect the levels and balance of n-6 and n-3 HUFA that in turn are converted to a wide array of lipids with signaling roles, including eicosanoids, docosanoids, other oxylipins and endocannabinoids. With few exceptions, n-6 HUFA are precursors of pro-inflammatory/pro-thrombotic signaling lipids, and n-3 HUFA are generally anti-inflammatory/anti-thrombotic. We and others have demonstrated that African ancestry populations have much higher frequencies (vs. European-, Asian- or indigenous Americas-ancestry populations) of a FADS “derived” haplotype that is associated with the efficient conversion of high levels of dietary n-6 PUFA to pro-inflammatory n-6 HUFA. By contrast, an “ancestral” haplotype, carrying alleles associated with a limited capacity to synthesize HUFA, which can lead to n-3 HUFA deficiency, is found at high frequency in certain Hispanic populations and is nearly fixed in several indigenous populations from the Americas. Based on these observations, a focused secondary subgroup analysis of the VITAL n-3 HUFA supplementation trial stratifying the data based on self-reported ancestry revealed that African Americans may benefit from n-3 HUFA supplementation, and both ancestry and FADS variability should be factored into future clinical trials design.
Collapse
Affiliation(s)
- Floyd H. Chilton
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States
- BIO5 Institute, University of Arizona, Tucson, AZ, United States
- *Correspondence: Floyd H. Chilton
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
| | - Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Timothy D. O'Connor
- Program in Personalized and Genomic Medicine, Department of Medicine, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Laurel M. Johnstone
- University of Arizona Genetics Core, University of Arizona, Tucson, AZ, United States
| | - Sarah Blomquist
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States
| | - Susan M. Schembre
- Department of Family and Community Medicine, College of Medicine-Tucson, University of Arizona, Tucson, AZ, United States
| | - Susan Sergeant
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Manja Zec
- School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
| | | | - Rasika A. Mathias
- Department of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brian Hallmark
- Center for Biomedical Informatics and Biostatistics, BIO5 Institute, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
27
|
Wang C, Enssle J, Pietzner A, Schmöcker C, Weiland L, Ritter O, Jaensch M, Elbelt U, Pagonas N, Weylandt KH. Essential Polyunsaturated Fatty Acids in Blood from Patients with and without Catheter-Proven Coronary Artery Disease. Int J Mol Sci 2022; 23:ijms23020766. [PMID: 35054948 PMCID: PMC8775772 DOI: 10.3390/ijms23020766] [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] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/30/2021] [Accepted: 01/07/2022] [Indexed: 01/21/2023] Open
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Statins reduce morbidity and mortality of CAD. Intake of n-3 polyunsaturated fatty acid (n-3 PUFAs), particularly eicosapentaenoic acid (EPA), is associated with reduced morbidity and mortality in patients with CAD. Previous data indicate that a higher conversion of precursor fatty acids (FAs) to arachidonic acid (AA) is associated with increased CAD prevalence. Our study explored the FA composition in blood to assess n-3 PUFA levels from patients with and without CAD. We analyzed blood samples from 273 patients undergoing cardiac catheterization. Patients were stratified according to clinically relevant CAD (n = 192) and those without (n = 81). FA analysis in full blood was performed by gas chromatography. Indicating increased formation of AA from precursors, the ratio of dihomo-gamma-linolenic acid (DGLA) to AA, the delta-5 desaturase index (D5D index) was higher in CAD patients. CAD patients had significantly lower levels of omega-6 polyunsaturated FAs (n-6 PUFA) and n-3 PUFA, particularly EPA, in the blood. Thus, our study supports a role of increased EPA levels for cardioprotection.
Collapse
Affiliation(s)
- Chaoxuan Wang
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Jörg Enssle
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, 14469 Potsdam, Germany
| | - Anne Pietzner
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
| | - Christoph Schmöcker
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
| | - Linda Weiland
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Oliver Ritter
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Monique Jaensch
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Ulf Elbelt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Medical Department, Division of Psychosomatic Medicine, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Nikolaos Pagonas
- Division of Medicine, Department of Cardiology, University Hospital Brandenburg an der Havel, Brandenburg Medical School, 14770 Brandenburg an der Havel, Germany; (L.W.); (O.R.); (M.J.); (N.P.)
| | - Karsten H. Weylandt
- Division of Medicine, Department of Gastroenterology, Metabolism and Oncology, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany; (C.W.); (J.E.); (A.P.); (C.S.); (U.E.)
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology, Brandenburg Medical School and University of Potsdam, 14469 Potsdam, Germany
- Correspondence: ; Tel.: +49-(0)3391-39-3210
| |
Collapse
|
28
|
Malyarchuk BA, Derenko MV, Denisova GA. Adaptive Changes in Fatty Acid Desaturation Genes in Indigenous Populations of Northeast Siberia. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421120103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Palavicini JP, Chavez-Velazquez A, Fourcaudot M, Tripathy D, Pan M, Norton L, DeFronzo RA, Shannon CE. The Insulin-Sensitizer Pioglitazone Remodels Adipose Tissue Phospholipids in Humans. Front Physiol 2021; 12:784391. [PMID: 34925073 PMCID: PMC8674727 DOI: 10.3389/fphys.2021.784391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/08/2021] [Indexed: 12/11/2022] Open
Abstract
The insulin-sensitizer pioglitazone exerts its cardiometabolic benefits in type 2 diabetes (T2D) through a redistribution of body fat, from ectopic and visceral areas to subcutaneous adipose depots. Whereas excessive weight gain and lipid storage in obesity promotes insulin resistance and chronic inflammation, the expansion of subcutaneous adipose by pioglitazone is associated with a reversal of these immunometabolic deficits. The precise events driving this beneficial remodeling of adipose tissue with pioglitazone remain unclear, and whether insulin-sensitizers alter the lipidomic composition of human adipose has not previously been investigated. Using shotgun lipidomics, we explored the molecular lipid responses in subcutaneous adipose tissue following 6months of pioglitazone treatment (45mg/day) in obese humans with T2D. Despite an expected increase in body weight following pioglitazone treatment, no robust effects were observed on the composition of storage lipids (i.e., triglycerides) or the content of lipotoxic lipid species (e.g., ceramides and diacylglycerides) in adipose tissue. Instead, pioglitazone caused a selective remodeling of the glycerophospholipid pool, characterized by a decrease in lipids enriched for arachidonic acid, such as plasmanylethanolamines and phosphatidylinositols. This contributed to a greater overall saturation and shortened chain length of fatty acyl groups within cell membrane lipids, changes that are consistent with the purported induction of adipogenesis by pioglitazone. The mechanism through which pioglitazone lowered adipose tissue arachidonic acid, a major modulator of inflammatory pathways, did not involve alterations in phospholipase gene expression but was associated with a reduction in its precursor linoleic acid, an effect that was also observed in skeletal muscle samples from the same subjects. These findings offer important insights into the biological mechanisms through which pioglitazone protects the immunometabolic health of adipocytes in the face of increased lipid storage.
Collapse
Affiliation(s)
- Juan P. Palavicini
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Alberto Chavez-Velazquez
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Marcel Fourcaudot
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Devjit Tripathy
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Meixia Pan
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Luke Norton
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ralph A. DeFronzo
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Christopher E. Shannon
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
30
|
Wang C, Murphy J, Delaney KZ, Khor N, Morais JA, Tsoukas MA, Lowry DE, Mutch DM, Santosa S. Association between rs174537 FADS1 polymorphism and immune cell profiles in abdominal and femoral subcutaneous adipose tissue: an exploratory study in adults with obesity. Adipocyte 2021; 10:124-130. [PMID: 33595419 PMCID: PMC7894460 DOI: 10.1080/21623945.2021.1888470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Fatty acid desaturase 1 (FADS1) polymorphisms alter fatty acid content in subcutaneous adipose tissue (SAT); however, existing evidence is limited and conflicting regarding the association between FADS1 variants and SAT inflammatory status. To advance this area, we conducted an exploratory study to investigate whether the common rs174537 polymorphism in FADS1 was associated with immune cell profiles in abdominal and femoral SAT in individuals with obesity. FADS1 gene expression and immune cell profiles in SAT depots were assessed by qPCR and flow cytometry, respectively. Although FADS1 gene expression was associated with genotype, no associations were observed with immune cell profiles in either depot. Our study provides additional evidence that rs174537 in FADS1 has minimal impact on inflammatory status in obese SAT.
Collapse
Affiliation(s)
- Chenxuan Wang
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Canada
| | - Jessica Murphy
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Montreal, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
| | - Kerri Z. Delaney
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Montreal, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
| | - Natalie Khor
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Montreal, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
| | - José A. Morais
- Division of Geriatric Medicine, McGill University Health Centre, Montreal, Canada
| | - Michael A. Tsoukas
- Division of Endocrinology, Department of Medicine, McGill University, Montréal, Canada
| | - Dana E. Lowry
- 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
| | - Sylvia Santosa
- Department of Health, Kinesiology and Applied Physiology, Concordia University, Montreal, Canada
- Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Montreal, Canada
- Centre de recherche - Axe maladies chroniques, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Ile-de-Montréal, Hôpital du Sacré-Coeur de Montréal, Montreal, Canada
| |
Collapse
|
31
|
Fatty Acid Metabolism and Derived-Mediators Distinctive of PPAR-α Activation in Obese Subjects Post Bariatric Surgery. Nutrients 2021; 13:nu13124340. [PMID: 34959892 PMCID: PMC8705357 DOI: 10.3390/nu13124340] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022] Open
Abstract
Bariatric surger (BS) is characterized by lipid metabolic changes as a response to the massive release of non-esterified fatty acids (NEFA) from adipose depots. The study aimed at evaluating changes in polyunsaturated fatty acids (PUFA) metabolism and biosynthesis of the lipid mediators N-acylethanolamines (NAE), as indices of nuclear peroxisome proliferator-activated receptor (PPAR)-α activation. The observational study was performed on 35 subjects (27 female, 8 male) with obesity, undergoing bariatric surgery. We assessed plasma FA and NAE profiles by LC-MS/MS, clinical parameters and anthropometric measures before and 1 and 6 months after bariatric surgery. One month after bariatric surgery, as body weight and clinical parameters improved significantly, we found higher plasma levels of N-oleoylethanolamine, arachidonic and a 22:6-n3/20:5-n3 ratio as evidence of PPAR-α activation. These changes corresponded to higher circulating levels of NEFA and a steep reduction of the fat mass. After 6 months 22:6-n3/20:5-n3 remained elevated and fat mass was further reduced. Our data suggest that the massive release of NEFA from adipose tissue at 1-Post, possibly by inducing PPAR-α, may enhance FA metabolism contributing to fat depot reduction and improved metabolic parameters in the early stage. However, PUFA metabolic changes favor n6 PUFA biosynthesis, requiring a nutritional strategy aimed at reducing the n6/n3 PUFA ratio.
Collapse
|
32
|
Miladipour A, Gholipour M, Honarmand Tamizkar K, Abak A, Kholghi Oskooei V, Taheri M, Ghafouri-Fard S. Investigation of FADS Gene Cluster Single Nucleotide Polymorphisms in End-Stage Renal Disease Compared With Normal Controls. Front Genet 2021; 12:716151. [PMID: 34603380 PMCID: PMC8481823 DOI: 10.3389/fgene.2021.716151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/12/2021] [Indexed: 12/03/2022] Open
Abstract
End-stage renal disease (ESRD) is a public health problem with a high burden. The condition is associated with abnormalities in lipid metabolism. The fatty acid desaturase (FADS) gene cluster includes three genes that are significantly correlated with a number of pathologic conditions related to abnormal lipid levels. In the current study, we genotyped rs174556, rs99780, and rs7115739 single nucleotide polymorphisms within the FADS cluster in a population of ESRD patients and healthy controls. The rs174556 of the FADS1 gene and rs99780 of the FADS2 gene were not associated with the risk of ESRD in any inheritance model. However, the rs7115739 of FADS3 was associated with the risk of ESRD in all models except for the recessive model. The T allele of this SNP was significantly less prevalent among cases compared with controls [odds ratio (OR) (95% CI) = 0.44 (0.25-0.77), P value = 0.004]. GT and TT genotypes has been shown to decrease the risk of ESRD in a codominant model [OR (95% CI) = 0.49 (0.26-0.92) and OR (95% CI) = 0.18 (0.02-1.6), respectively; P value = 0.019]. In the dominant model, GT + TT status was associated with lower risk of ESRD [OR (95% CI) = 0.45 (0.24-0.82), P value = 0.0078]. Assessment of association between this SNP and risk of ESRD in an overdominant model revealed that GT genotype decreases the risk of this condition [OR (95% CI) = 0.5 (0.27-0.94), P value = 0.029]. Taken together, the rs7115739 of FADS3 is suggested as a putative modulator of the risk of ESRD in the Iranian population.
Collapse
Affiliation(s)
- Amirhossein Miladipour
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholipour
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Honarmand Tamizkar
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Kholghi Oskooei
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
- Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Dirajlal-Fargo S, Sattar A, Yu J, Albar Z, Chaves FC, Riedl K, Kityo C, Bowman E, McComsey GA, Funderburg N. Lipidome association with vascular disease and inflammation in HIV+ Ugandan children. AIDS 2021; 35:1615-1623. [PMID: 33878042 PMCID: PMC8286331 DOI: 10.1097/qad.0000000000002923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE HIV infection and antiretroviral therapy (ART) have both been linked to dyslipidemia and increased cardiovascular disease (CVD). The relationships among the lipidome, immune activation, and subclinical vascular disease in children with perinatally acquired HIV (PHIV) have not been investigated. METHODS Serum lipid composition, including 13 lipid classes constituting 850 different lipid species were measured by direct infusion-tandem mass spectrometry in samples from 20 ART-treated PHIV and 20 age-matched and sex-matched HIV- Ugandan children. All participants were between 10 and 18 years of age with no other known active infections. PHIVs had HIV-1 RNA level 50 copies/ml or less. In addition, common carotid artery intima--media thickness (IMT), as well as plasma marker of systemic inflammation (hsCRP, IL6, sTNFRa I), monocyte activation (soluble CD14 and CD163), and T-cell activation (expression of CD38 and HLA-DR on CD4+ and CD8+) were evaluated. RESULTS Median age (Q1, Q3) of study participants was 13 years (11, 15), 37% were boys, 75% were on an NNRTI-based ART regimen. The concentrations of cholesterol ester, LCER, phosphatidylcholines, and sphingomyelin lipid classes were significantly increased in serum of PHIV compared with HIV (P≤0.04). Biomarkers associated with CVD risk including hsCRP, sCD163, and T-cell activation were directly correlated with lipid species in PHIV (P ≤ 0.04). Contents of free fatty acids including palmitic (16 : 0), stearic (18 : 0), and arachidic acid (20 : 0) were positively correlated with IMT in PHIV. CONCLUSION Serum lipidome is altered in young virally suppressed PHIV on ART. A direct association between inflammation and lipid species known to be associated with CVD was observed.
Collapse
Affiliation(s)
- Sahera Dirajlal-Fargo
- University Hospitals Cleveland Medical Center
- Rainbow Babies and Children's Hospital
- Case Western Reserve University, Cleveland
| | | | - Jiao Yu
- Case Western Reserve University, Cleveland
| | | | - Fabio C Chaves
- Department of Food Science and Technology, and the OSU Comprehensive Cancer Center, The Ohio State University, OH, USA
| | - Ken Riedl
- Department of Food Science and Technology, and the OSU Comprehensive Cancer Center, The Ohio State University, OH, USA
| | - Cissy Kityo
- Joint Clinical Research Centre, Kampala, Uganda
| | - Emily Bowman
- Ohio State University School of Health and Rehabilitation Sciences, Columbus, OH, USA
| | - Grace A McComsey
- University Hospitals Cleveland Medical Center
- Rainbow Babies and Children's Hospital
- Case Western Reserve University, Cleveland
| | - Nicholas Funderburg
- Ohio State University School of Health and Rehabilitation Sciences, Columbus, OH, USA
| |
Collapse
|
34
|
Liput KP, Lepczyński A, Ogłuszka M, Nawrocka A, Poławska E, Grzesiak A, Ślaska B, Pareek CS, Czarnik U, Pierzchała M. Effects of Dietary n-3 and n-6 Polyunsaturated Fatty Acids in Inflammation and Cancerogenesis. Int J Mol Sci 2021; 22:6965. [PMID: 34203461 PMCID: PMC8268933 DOI: 10.3390/ijms22136965] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/30/2022] Open
Abstract
The dietary recommendation encourages reducing saturated fatty acids (SFA) in diet and replacing them with polyunsaturated fatty acids (PUFAs) n-3 (omega-3) and n-6 (omega-6) to decrease the risk of metabolic disturbances. Consequently, excessive n-6 PUFAs content and high n-6/n-3 ratio are found in Western-type diet. The importance of a dietary n-6/n-3 ratio to prevent chronic diseases is linked with anti-inflammatory functions of linolenic acid (ALA, 18:3n-3) and longer-chain n-3 PUFAs. Thus, this review provides an overview of the role of oxylipins derived from n-3 PUFAs and oxylipins formed from n-6 PUFAs on inflammation. Evidence of PUFAs' role in carcinogenesis was also discussed. In vitro studies, animal cancer models and epidemiological studies demonstrate that these two PUFA groups have different effects on the cell growth, proliferation and progression of neoplastic lesions.
Collapse
Affiliation(s)
- Kamila P. Liput
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland; (K.P.L.); (M.O.); (A.N.); (E.P.)
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Adam Lepczyński
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology, ul. K. Janickiego 29, 71-270 Szczecin, Poland; (A.L.); (A.G.)
| | - Magdalena Ogłuszka
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland; (K.P.L.); (M.O.); (A.N.); (E.P.)
| | - Agata Nawrocka
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland; (K.P.L.); (M.O.); (A.N.); (E.P.)
- Department of Experimental Genomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland
| | - Ewa Poławska
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland; (K.P.L.); (M.O.); (A.N.); (E.P.)
| | - Agata Grzesiak
- Department of Physiology, Cytobiology and Proteomics, West Pomeranian University of Technology, ul. K. Janickiego 29, 71-270 Szczecin, Poland; (A.L.); (A.G.)
| | - Brygida Ślaska
- Institute of Biological Bases of Animal Production, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland;
| | - Chandra S. Pareek
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, ul. J. Gagarina 7, 87-100 Toruń, Poland;
- Division of Functional Genomics in Biological and Biomedical Research, Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, ul. Wilenska 4, 87-100 Torun, Poland
| | - Urszula Czarnik
- Department of Pig Breeding, Faculty of Animal Bio-Engineering, University of Warmia and Mazury in Olsztyn, ul. M. Oczapowskiego 5, 10-719 Olsztyn, Poland;
| | - Mariusz Pierzchała
- Department of Genomics and Biodiversity, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzebiec, 05-552 Magdalenka, Poland; (K.P.L.); (M.O.); (A.N.); (E.P.)
| |
Collapse
|
35
|
Gene Expression Profile in Different Age Groups and Its Association with Cognitive Function in Healthy Malay Adults in Malaysia. Cells 2021; 10:cells10071611. [PMID: 34199148 PMCID: PMC8304476 DOI: 10.3390/cells10071611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/13/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
The mechanism of cognitive aging at the molecular level is complex and not well understood. Growing evidence suggests that cognitive differences might also be caused by ethnicity. Thus, this study aims to determine the gene expression changes associated with age-related cognitive decline among Malay adults in Malaysia. A cross-sectional study was conducted on 160 healthy Malay subjects, aged between 28 and 79, and recruited around Selangor and Klang Valley, Malaysia. Gene expression analysis was performed using a HumanHT-12v4.0 Expression BeadChip microarray kit. The top 20 differentially expressed genes at p < 0.05 and fold change (FC) = 1.2 showed that PAFAH1B3, HIST1H1E, KCNA3, TM7SF2, RGS1, and TGFBRAP1 were regulated with increased age. The gene set analysis suggests that the Malay adult's susceptibility to developing age-related cognitive decline might be due to the changes in gene expression patterns associated with inflammation, signal transduction, and metabolic pathway in the genetic network. It may, perhaps, have important implications for finding a biomarker for cognitive decline and offer molecular targets to achieve successful aging, mainly in the Malay population in Malaysia.
Collapse
|
36
|
Khodarahmi M, Nikniaz L, Abbasalizad Farhangi M. The Interaction Between Fatty Acid Desaturase-2 (FADS2) rs174583 Genetic Variant and Dietary Quality Indices (DASH and MDS) Constructs Different Metabolic Phenotypes Among Obese Individuals. Front Nutr 2021; 8:669207. [PMID: 34164423 PMCID: PMC8215104 DOI: 10.3389/fnut.2021.669207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/22/2021] [Indexed: 01/02/2023] Open
Abstract
Background and Aim: Genetic variation in fatty acid desaturases (FADS) has previously been linked to several diet-related diseases. We aimed to determine whether the FADS2 rs174583 variant interacts with the Dietary Approach to Stop Hypertension (DASH) score and Mediterranean dietary score (MDS) to influence cardio-metabolic risk factors among obese adults. Methods: This cross-sectional study was performed among 347 apparently healthy obese adults (aged 20-50 years). Dietary quality indicator scores (DASH and MDS) were generated using a validated 147-item Food Frequency Questionnaire (FFQ). The FADS2 rs174583 variant was genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The gene-diet interaction was analyzed by the ANCOVA multivariate interaction model. Results: A significant interaction was observed between rs174583 and adherence to the DASH score in relation to serum triglyceride (TG) concentration among the female group (P Interaction = 0.046); CT-genotype carriers who were assigned to the second tertile of DASH compared with those in the first tertile had a lower TG level (P < 0.05). Another significant interaction was revealed between adherence to MDS score and rs174583 polymorphism on serum glucose levels (P Interaction = 0.044); the lowest mean of glucose level was observed in homozygous minor subjects (TT) in the third tertile of MDS, in comparison with other tertiles of this dietary index (P < 0.05). There was a similar significant interaction between DASH and rs174583 in relation to diastolic blood pressure (P Interaction = 0.038) among the male group. Additionally, a significant positive association was found between TT genotype and odds of having high TG both in the crude (OR, 3.21; 95% CI, 1.02-10.14) and adjusted (OR, 3.58; 95% CI, 1.07-11.97) models, taking into account different confounders. Conclusion: Adherence to the dietary quality indicators (DASH and MDS) modified the relationship between FADS2 rs174583 polymorphism and cardio-metabolic risk factors in obese subjects. Prospective cohort studies are needed to confirm the results of our study.
Collapse
Affiliation(s)
- Mahdieh Khodarahmi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Nikniaz
- Tabriz Health Services Management Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Abbasalizad Farhangi
- Department of Community Nutrition, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
37
|
Hardy DS, Racette SB, Garvin JT, Gebrekristos HT, Mersha TB. Ancestry specific associations of a genetic risk score, dietary patterns and metabolic syndrome: a longitudinal ARIC study. BMC Med Genomics 2021; 14:118. [PMID: 33933074 PMCID: PMC8088631 DOI: 10.1186/s12920-021-00961-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/15/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Associations have been observed among genetic variants, dietary patterns, and metabolic syndrome (MetS). A gap in knowledge is whether a genetic risk score (GRS) and dietary patterns interact to increase MetS risk among African Americans. We investigated whether MetS risk was influenced by interaction between a GRS and dietary patterns among Whites and African Americans. A secondary aim examined if molecular genetic clusterings differed by racial ancestry. METHODS We used longitudinal data over 4-visits (1987-1998) that included 10,681 participants aged 45-64y at baseline from the Atherosclerosis Risk in Communities study (8451 Whites and 2230 African Americans). We constructed a simple-count GRS as the linear weighted sum of high-risk alleles (0, 1, 2) from cardiovascular disease polymorphisms from the genome-wide association studies catalog associated with MetS risk. Three dietary patterns were determined by factor analysis of food frequency questionnaire data: Western, healthy, and high-fat dairy. MetS was defined according to the 2016 National Cholesterol Education Program Adult Treatment Panel III criteria but used 2017 American Heart Association/American College of Cardiology criteria for elevated blood pressure. Analyses included generalized linear model risk ratios (RR), 95% confidence intervals (CI), and Bonferroni correction for multiple testing. RESULTS The Western dietary pattern was associated with higher risk for MetS across increasing GRS tertiles among Whites (p < 0.017). The high-fat dairy pattern was protective against MetS, but its impact was most effective in the lowest GRS tertile in Whites (RR = 0.62; CI: 0.52-0.74) and African Americans (RR = 0.67; CI: 0.49-0.91). Among each racial group within GRS tertiles, the Western dietary pattern was associated with development and cycling of MetS status between visits, and the high-fat dairy pattern with being free from MetS (p < 0.017). The healthy dietary pattern was associated with higher risk of MetS among African Americans which may be explained by higher sucrose intake (p < 0.0001). Fewer genes, but more metabolic pathways for obesity, body fat distribution, and lipid and carbohydrate metabolism were identified in African Americans than Whites. Some polymorphisms were linked to the Western and high-fat dairy patterns. CONCLUSION The influence of dietary patterns on MetS risk appears to differ by genetic predisposition and racial ancestry.
Collapse
Affiliation(s)
- Dale S. Hardy
- Department of Internal Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310 USA
| | - Susan B. Racette
- Program in Physical Therapy and Department of Medicine, Washington University School of Medicine, St. Louis, MO 63108 USA
| | - Jane T. Garvin
- College of Nursing, Augusta University, Augusta, GA 30912 USA
| | - Hirut T. Gebrekristos
- Department of Internal Medicine, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310 USA
| | - Tesfaye B. Mersha
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, 3333 Burnet Ave, Cincinnati, OH 45229 USA
| |
Collapse
|
38
|
Nosrati-Oskouie M, Aghili-Moghaddam NS, Sathyapalan T, Sahebkar A. Impact of curcumin on fatty acid metabolism. Phytother Res 2021; 35:4748-4762. [PMID: 33825246 DOI: 10.1002/ptr.7105] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/21/2021] [Accepted: 03/19/2021] [Indexed: 01/12/2023]
Abstract
Free fatty acids (FFAs) and fatty acid synthesis (FAS) activity have significantly contributed to disease states such as insulin resistance, obesity, type 2 diabetes, myocardial infarction, blood pressure, and several types of cancer. Currently, several treatment options are available for patients with these conditions. Due to safety concerns, adverse effects, limited efficacy, and low tolerability associated with many medications, the identification of novel agents with less toxicity and a more favorable outcome is warranted. Curcumin is a phenolic compound derived from the turmeric plant with various biological activities, including anticarcinogenic, antioxidant, antiinflammatory, and hypolipidemic properties. PubMed, Scopus, and Web of Science were searched up to February 2020 for studies that demonstrated the efficacy and mechanisms of curcumin action on FFAs, FAS, and β-oxidation activity, as well as the desaturation system. Most of the evidence is in-vivo and in-vitro studies that demonstrate that curcumin possesses regulatory properties on FFAs levels through its effects on FAS and β-oxidation activity as well as desaturation system, which could improve insulin resistance, obesity, and other FFAs-related disorders. The present study provides a review of the existing in-vitro, in-vivo, and clinical evidence on the effect of curcumin on FFAs and FAS activity, β-oxidation, and desaturation system.
Collapse
Affiliation(s)
- Mohammad Nosrati-Oskouie
- Student Research Committee, Department of Clinical Nutrition, Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
39
|
Lankinen MA, de Mello VD, Meuronen T, Sallinen T, Ågren J, Virtanen KA, Laakso M, Pihlajamäki J, Schwab U. The FADS1 Genotype Modifies Metabolic Responses to the Linoleic Acid and Alpha-linolenic Acid Containing Plant Oils-Genotype Based Randomized Trial FADSDIET2. Mol Nutr Food Res 2021; 65:e2001004. [PMID: 33548080 DOI: 10.1002/mnfr.202001004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/23/2020] [Indexed: 12/19/2022]
Abstract
SCOPE The article investigates the FADS1 rs174550 genotype interaction with dietary intakes of high linoleic acid (LA) and high alpha-linolenic acid (ALA) on the response of fatty acid composition of plasma phospholipids (PLs), and of markers of low-grade inflammation and glucose-insulin homeostasis. METHODS AND RESULTS One-hundred thirty homozygotes men for FADS1 rs174550 SNP (TT and CC genotypes) were randomized to an 8-week intervention with either LA- or ALA-enriched diet (13 E% PUFA). The source of LA and ALA are 30-50 mL of sunflower oil (SFO, 62-63% LA) and Camelina sativa oil (CSO, 30- are randomized to an 35% ALA), respectively. In the SFO arm, there is a significant genotype x diet interaction for the proportion of arachidonic acid in plasma phospholipids (p < 0.001), disposition index (DI30 ) (p = 0.039), and for serum high-sensitive c-reactive protein (hs-CRP, p = 0.029) after excluding the participants with hs-CRP concentration of >10 mg L-1 and users of statins or anti-inflammatory therapy. In the CSO arm, there are significant genotype x diet interactions for n-3 polyunsaturated fatty acids, but not for the clinical characteristics. CONCLUSIONS The FADS1 genotype modifies the response to high PUFA diets, especially to high-LA diet. These findings suggest that approaches considering FADS variation may be useful in personalized dietary counseling.
Collapse
Affiliation(s)
- Maria A Lankinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Vanessa D de Mello
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Topi Meuronen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Taisa Sallinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jyrki Ågren
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Kirsi A Virtanen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Ursula Schwab
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
40
|
Francis M, Li C, Sun Y, Zhou J, Li X, Brenna JT, Ye K. Genome-wide association study of fish oil supplementation on lipid traits in 81,246 individuals reveals new gene-diet interaction loci. PLoS Genet 2021; 17:e1009431. [PMID: 33760818 PMCID: PMC8021161 DOI: 10.1371/journal.pgen.1009431] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 04/05/2021] [Accepted: 02/16/2021] [Indexed: 01/23/2023] Open
Abstract
Fish oil supplementation is widely used for reducing serum triglycerides (TAGs) but has mixed effects on other circulating cardiovascular biomarkers. Many genetic polymorphisms have been associated with blood lipids, including high- and low-density-lipoprotein cholesterol (HDL-C, LDL-C), total cholesterol, and TAGs. Here, the gene-diet interaction effects of fish oil supplementation on these lipids were analyzed in a discovery cohort of up to 73,962 UK Biobank participants, using a 1-degree-of-freedom (1df) test for interaction effects and a 2-degrees-of-freedom (2df) test to jointly analyze interaction and main effects. Associations with P < 1×10-6 in either test (26,157; 18,300 unique variants) were advanced to replication in up to 7,284 participants from the Atherosclerosis Risk in Communities (ARIC) Study. Replicated associations reaching 1df P < 0.05 (2,175; 1,763 unique variants) were used in meta-analyses. We found 13 replicated and 159 non-replicated (UK Biobank only) loci with significant 2df joint tests that were predominantly driven by main effects and have been previously reported. Four novel interaction loci were identified with 1df P < 5×10-8 in meta-analysis. The lead variant in the GJB6-GJB2-GJA3 gene cluster, rs112803755 (A>G; minor allele frequency = 0.041), shows exclusively interaction effects. The minor allele is significantly associated with decreased TAGs in individuals with fish oil supplementation, but with increased TAGs in those without supplementation. This locus is significantly associated with higher GJB2 expression of connexin 26 in adipose tissue; connexin activity is known to change upon exposure to omega-3 fatty acids. Significant interaction effects were also found in three other loci in the genes SLC12A3 (HDL-C), ABCA6 (LDL-C), and MLXIPL (LDL-C), but highly significant main effects are also present. Our study identifies novel gene-diet interaction effects for four genetic loci, whose effects on blood lipids are modified by fish oil supplementation. These findings highlight the need and possibility for personalized nutrition.
Collapse
Affiliation(s)
- Michael Francis
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Yitang Sun
- Department of Genetics, University of Georgia, Athens, Georgia, United States of America
| | - Jingqi Zhou
- Department of Genetics, University of Georgia, Athens, Georgia, United States of America
| | - Xiang Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - J. Thomas Brenna
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States of America
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin, Austin, Texas, United States of America
- Departments of Nutrition and Chemistry, University of Texas at Austin, Austin, Texas, United States of America
| | - Kaixiong Ye
- Institute of Bioinformatics, University of Georgia, Athens, Georgia, United States of America
- Department of Genetics, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
41
|
Athinarayanan S, Fan YY, Wang X, Callaway E, Cai D, Chalasani N, Chapkin RS, Liu W. Fatty Acid Desaturase 1 Influences Hepatic Lipid Homeostasis by Modulating the PPARα-FGF21 Axis. Hepatol Commun 2021; 5:461-477. [PMID: 33681679 PMCID: PMC7917273 DOI: 10.1002/hep4.1629] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022] Open
Abstract
The fatty acid desaturase 1 (FADS1), also known as delta-5 desaturase (D5D), is one of the rate-limiting enzymes involved in the desaturation and elongation cascade of polyunsaturated fatty acids (PUFAs) to generate long-chain PUFAs (LC-PUFAs). Reduced function of D5D and decreased hepatic FADS1 expression, as well as low levels of LC-PUFAs, were associated with nonalcoholic fatty liver disease. However, the causal role of D5D in hepatic lipid homeostasis remains unclear. In this study, we hypothesized that down-regulation of FADS1 increases susceptibility to hepatic lipid accumulation. We used in vitro and in vivo models to test this hypothesis and to delineate the molecular mechanisms mediating the effect of reduced FADS1 function. Our study demonstrated that FADS1 knockdown significantly reduced cellular levels of LC-PUFAs and increased lipid accumulation and lipid droplet formation in HepG2 cells. The lipid accumulation was associated with significant alterations in multiple pathways involved in lipid homeostasis, especially fatty acid oxidation. These effects were demonstrated to be mediated by the reduced function of the peroxisome proliferator-activated receptor alpha (PPARα)-fibroblast growth factor 21 (FGF21) axis, which can be reversed by treatment with docosahexaenoic acid, PPARα agonist, or FGF21. In vivo, FADS1-knockout mice fed with high-fat diet developed increased hepatic steatosis as compared with their wild-type littermates. Molecular analyses of the mouse liver tissue largely corroborated the observations in vitro, especially along with reduced protein expression of PPARα and FGF21. Conclusion: Collectively, these results suggest that dysregulation in FADS1 alters liver lipid homeostasis in the liver by down-regulating the PPARα-FGF21 signaling axis.
Collapse
Affiliation(s)
- Shaminie Athinarayanan
- Department of Medicinal Chemistry and Molecular PharmacologyCollege of PharmacyPurdue UniversityWest LafayetteINUSA
| | - Yang-Yi Fan
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
| | - Xiaokun Wang
- Department of Pharmaceutical SciencesWayne State UniversityDetroitMIUSA
| | - Evelyn Callaway
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA
| | - Defeng Cai
- Department of Pharmaceutical SciencesWayne State UniversityDetroitMIUSA
| | - Naga Chalasani
- Division of Gastroenterology and HepatologyIndiana University School of MedicineIndianapolisINUSA
| | - Robert S Chapkin
- Program in Integrative Nutrition and Complex DiseasesTexas A&M UniversityCollege StationTXUSA.,Texas A&M Center for Environmental Health ResearchTexas A&M UniversityCollege StationTXUSA
| | - Wanqing Liu
- Department of Medicinal Chemistry and Molecular PharmacologyCollege of PharmacyPurdue UniversityWest LafayetteINUSA.,Department of Pharmaceutical SciencesWayne State UniversityDetroitMIUSA.,Department of PharmacologyWayne State UniversityDetroitMIUSA
| |
Collapse
|
42
|
Liebisch G, Plagge J, Höring M, Seeliger C, Ecker J. The effect of gut microbiota on the intestinal lipidome of mice. Int J Med Microbiol 2021; 311:151488. [PMID: 33662870 DOI: 10.1016/j.ijmm.2021.151488] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/01/2021] [Accepted: 02/23/2021] [Indexed: 02/08/2023] Open
Abstract
Gut microbiota significantly influence the plasma and liver lipidome. An interconnecting metabolite is acetate generated after degradation and fermentation of dietary fiber by the gut microbiota, which is metabolized in the liver into longer chain fatty acids and complex lipids reaching the circulation. Whether these systemic changes are accompanied by alternations of the intestinal lipidome is unclear. Therefore, we quantified glycerophospholipids, sphingolipids and sterols in ileum and colon, the two segments containing the highest densities of microbes in the gastrointestinal tract, of germfree and specific pathogen free mice using mass spectrometry-based lipidomics. We found that the presence of gut microbes lowers the free cholesterol content in colon while elevating phosphatidylcholine levels. Further, PUFA-containing phosphatidylcholine and -ethanolamine fractions are increased in ileum and colon of germfree compared to SPF mice. A total fatty acid analysis by GC-MS revealed higher levels of arachidonic and docosahexaenoic acid in the ileum of germfree mice indicating that the gut microbiota inhibits PUFA metabolism in the small intestine.
Collapse
Affiliation(s)
- Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany.
| | - Johannes Plagge
- ZIEL - Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Claudine Seeliger
- ZIEL - Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany
| | - Josef Ecker
- ZIEL - Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Freising, Germany.
| |
Collapse
|
43
|
Rifkin SB, Shrubsole MJ, Cai Q, Smalley WE, Ness RM, Swift LL, Milne G, Zheng W, Murff HJ. Differences in erythrocyte phospholipid membrane long-chain polyunsaturated fatty acids and the prevalence of fatty acid desaturase genotype among African Americans and European Americans. Prostaglandins Leukot Essent Fatty Acids 2021; 164:102216. [PMID: 33310680 DOI: 10.1016/j.plefa.2020.102216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/31/2020] [Accepted: 11/12/2020] [Indexed: 01/02/2023]
Abstract
Numerous studies have reported an association between genetic variants in fatty acid desaturases (FADS1 and FADS2) and plasma or erythrocyte long chain polyunsaturated fatty acid (PUFA) levels. Increased levels of n-6 PUFAs have been associated with inflammation and several chronic diseases, including diabetes and cancer. We hypothesized that genetic variants of FADS that more efficiently convert precursor n-6 PUFA to arachidonic acid (AA) may explain the higher burden of chronic diseases observed in African Americans. To test this hypothesis, we measured the level of n-6 and n-3 PUFAs in erythrocyte membrane phospholipids and genotyped the rs174537 FADS variants associated with higher AA conversion among African American and European American populations. We included data from 1,733 individuals who participated in the Tennessee Colorectal Polyp Study, a large colonoscopy-based case-control study. Erythrocyte membrane PUFA percentages were measured using gas chromatography. Generalized linear models were used to estimate association of race and genotype on erythrocyte phospholipid membrane PUFA levels while controlling for self-reported dietary intake. We found that African Americans have higher levels of AA and a higher prevalence of GG allele compared to whites, 81% vs 43%, respectively. Homozygous GG genotype was negatively associated with precursor PUFAs (linoleic [LA], di-homo-γ-linolenic [DGLA]), positively associated with both product PUFA (AA, docosahexaenoic acid [DHA]), product to precursor ratio (AA to DGLA), an indirect measure of FADs efficiency and increased urinary isoprostane F2 (F2-IsoP) and isoprostane F3 (F3-IsoP), markers of oxidative stress. Increased consumption of n-6 PUFA and LA resulting in increased AA and subsequent inflammation may be fueling increased prevalence of chronic diseases especially in African descent.
Collapse
Affiliation(s)
- S B Rifkin
- Division of Gastroenterology and Hepatology, University of Michigan School of Medicine, 1150 W. Medical Center Drive, 6520 MSRB1, Ann Arbor, Michigan, United States.
| | - M J Shrubsole
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Division of Epidemiology, Vanderbilt University School of Medicine, United States; Geriatrics Research, Education and Clinical Center (GRECC), Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, United States
| | - Q Cai
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Division of Epidemiology, Vanderbilt University School of Medicine, United States
| | - W E Smalley
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Division of Gastroenterology, Vanderbilt University School of Medicine, United States
| | - R M Ness
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Division of Gastroenterology, Vanderbilt University School of Medicine, United States
| | - L L Swift
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, United States
| | - G Milne
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Division of Clinical Pharmacology, Vanderbilt University School of Medicine, United States
| | - W Zheng
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Division of Epidemiology, Vanderbilt University School of Medicine, United States; Geriatrics Research, Education and Clinical Center (GRECC), Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, United States
| | - H J Murff
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States; Geriatrics Research, Education and Clinical Center (GRECC), Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, United States; Division of General Internal Medicine and Public Health, Vanderbilt University Medical Center, United States
| |
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: 0.8] [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
|
Ye S, Matthan NR, Lamon-Fava S, Aguilar GS, Turner JR, Walker ME, Chai Z, Lakshman S, Urban JF, Lichtenstein AH. Western and heart healthy dietary patterns differentially affect the expression of genes associated with lipid metabolism, interferon signaling and inflammation in the jejunum of Ossabaw pigs. J Nutr Biochem 2020; 90:108577. [PMID: 33388349 PMCID: PMC8982565 DOI: 10.1016/j.jnutbio.2020.108577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
Diet quality and statin therapy are established modulators of coronary artery disease (CAD) progression, but their effect on the gastrointestinal tract and subsequent sequelae that could affect CAD progression are relatively unexplored. To address this gap, Ossabaw pigs (N = 32) were randomly assigned to receive isocaloric amounts of a Western-type diet (WD; high in saturated fat, refined carbohydrate, and cholesterol, and low in fiber) or a heart healthy-type diet (HHD; high in unsaturated fat, whole grains, fruits and vegetables, supplemented with fish oil, and low in cholesterol), with or without atorvastatin, for 6 months. At the end of the study, RNA sequencing with 100 base pair single end reads on NextSeq 500 platform was conducted in isolated pig jejunal mucosa. A two-factor edgeR analysis revealed that the dietary patterns resulted in three differentially expressed genes related to lipid metabolism (SCD, FADS1, and SQLE). The expression of these genes was associated with cardiometabolic risk factors and atherosclerotic lesion severity. Subsequent gene enrichment analysis indicated the WD, compared to the HHD, resulted in higher interferon signaling and inflammation, with some of these genes being significantly associated with serum TNF-α and/or hsCRP concentrations, but not atherosclerotic lesion severity. No significant effect of atorvastatin therapy on gene expression, nor its interaction with dietary patterns, was identified. In conclusion, Western and heart healthy-type dietary patterns differentially affect the expression of genes associated with lipid metabolism, interferon signaling, and inflammation in the jejunum of Ossabaw pigs.
Collapse
Affiliation(s)
- Shumao Ye
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Gloria Solano Aguilar
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Woman's Hospital and Harvard Medical School, Boston, MA, USA
| | - Maura E Walker
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - Zhi Chai
- Intercollege Graduate Degree Program in Physiology, Department of Nutritional Science, Pennsylvania State University, University Park, PA, USA
| | - Sukla Lakshman
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Joseph F Urban
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA.
| |
Collapse
|
46
|
Buoli M, Caldiroli L, Guenzani D, Carnevali GS, Cesari M, Turolo S, Barkin JL, Messa P, Agostoni C, Vettoretti S. Associations Between Cholesterol and Fatty Acid Profile on the Severity of Depression in Older Persons With Nondialysis Chronic Kidney Disease. J Ren Nutr 2020; 31:537-540. [PMID: 33386204 DOI: 10.1053/j.jrn.2020.08.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/22/2020] [Accepted: 08/30/2020] [Indexed: 11/11/2022] Open
Abstract
Depressive symptoms worsen the outcomes of patients affected by chronic kidney disease (CKD). The purpose of the present article is to study the association between serum lipid profile and the severity of depression in patients with CKD. We evaluated 132 older subjects with advanced CKD (stage 3-5, not receiving dialysis) in regular follow-up in a nephrology clinic. Blood samples were collected after an overnight fast. All patients were evaluated with the Geriatric Depression Scale which is comprised of 30 items that assess the severity of depressive symptoms. A backward multivariate regression analysis was performed to study the association between lipid profile and severity of depression. Low-density lipoprotein levels (β = 2.77, P = .008) and arachidonic acid/linoleic acid ratio (β = 2.51, P = .015) were found to be significantly associated with severity of depressive symptoms. Change in dietary habits or the use of hypocholesterolemic drugs could potentially prevent depressive symptoms and ameliorate outcome of patients affected by CKD. Data from prospective studies are needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Massimiliano Buoli
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| | - Lara Caldiroli
- Unit of Nephrology Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dalila Guenzani
- Department of Neurosciences and Mental Health, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Matteo Cesari
- Geriatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Stefano Turolo
- Pediatric Department of Nephrology Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Piergiorgio Messa
- Unit of Nephrology Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simone Vettoretti
- Unit of Nephrology Dialysis and Renal Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | |
Collapse
|
47
|
Kothapalli KSD, Park HG, Brenna JT. Polyunsaturated fatty acid biosynthesis pathway and genetics. implications for interindividual variability in prothrombotic, inflammatory conditions such as COVID-19 ✰,✰✰,★,★★. Prostaglandins Leukot Essent Fatty Acids 2020; 162:102183. [PMID: 33038834 PMCID: PMC7527828 DOI: 10.1016/j.plefa.2020.102183] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
Abstract
COVID-19 symptoms vary from silence to rapid death, the latter mediated by both a cytokine storm and a thrombotic storm. SARS-CoV (2003) induces Cox-2, catalyzing the synthesis, from highly unsaturated fatty acids (HUFA), of eicosanoids and docosanoids that mediate both inflammation and thrombosis. HUFA balance between arachidonic acid (AA) and other HUFA is a likely determinant of net signaling to induce a healthy or runaway physiological response. AA levels are determined by a non-protein coding regulatory polymorphisms that mostly affect the expression of FADS1, located in the FADS gene cluster on chromosome 11. Major and minor haplotypes in Europeans, and a specific functional insertion-deletion (Indel), rs66698963, consistently show major differences in circulating AA (>50%) and in the balance between AA and other HUFA (47-84%) in free living humans; the indel is evolutionarily selective, probably based on diet. The pattern of fatty acid responses is fully consistent with specific genetic modulation of desaturation at the FADS1-mediated 20:3→20:4 step. Well established principles of net tissue HUFA levels indicate that the high linoleic acid and low alpha-linoleic acid in populations drive the net balance of HUFA for any individual. We predict that fast desaturators (insertion allele at rs66698963; major haplotype in Europeans) are predisposed to higher risk and pathological responses to SARS-CoV-2 could be reduced with high dose omega-3 HUFA.
Collapse
Affiliation(s)
- Kumar S D Kothapalli
- Dell Pediatric Research Institute, Depts of Pediatrics, of Chemistry, and of Nutrition, University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, United States.
| | - Hui Gyu Park
- Dell Pediatric Research Institute, Depts of Pediatrics, of Chemistry, and of Nutrition, University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, United States.
| | - J Thomas Brenna
- Dell Pediatric Research Institute, Depts of Pediatrics, of Chemistry, and of Nutrition, University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX, United States; Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States.
| |
Collapse
|
48
|
Murphy C, Johnson AP, Koenekoop RK, Seiple W, Overbury O. The Relationship Between Cognitive Status and Known Single Nucleotide Polymorphisms in Age-Related Macular Degeneration. Front Aging Neurosci 2020; 12:586691. [PMID: 33178008 PMCID: PMC7596199 DOI: 10.3389/fnagi.2020.586691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/15/2020] [Indexed: 11/13/2022] Open
Abstract
Recent literature has reported a higher occurrence of cognitive impairment among individuals with Age-related Macular Degeneration (AMD) compared to older adults with normal vision. This pilot study explored potential links between single nucleotide polymorphisms (SNPs) in AMD and cognitive status. Individuals with AMD (N = 21) and controls (N = 18) were genotyped for the SNPs CFHY402H, ARMS2A69S and FADS1 rs174547. Cognitive status was evaluated using the Montreal Cognitive Assessment. The two groups differed significantly on which subscales were most difficult. The control group had difficulty with delayed recall while those with AMD had difficulty on delayed recall in addition to abstraction and orientation. Homozygous carriers of the FADS1 rs174547 SNP had significantly lower scores than heterozygotes or non-carriers on the MoCA. The results suggest that the FADS1 SNP may play a role in visual impairment/cognitive impairment comorbidity as reflected in the poorer cognitive scores among homozygotes with AMD compared to those carrying only one, or no copies of the SNP.
Collapse
Affiliation(s)
- Caitlin Murphy
- Low Vision Lab, School of Optometry, University of Montreal, Montreal, QC, Canada
- Concordia Vision Labs, Department of Psychology, Concordia University, Montreal, QC, Canada
- Centre for Interdisciplinary Research in Rehabilitation of Greater Montreal (CRIR)/Centre de Réadaptation Lethbridge-Layton-Mackay du Centre Intégré Universitaire de Santé et de Services Sociaux du Centre-Ouest-de-l’Ile-de-Montréal (CIUSSS) du Centre-Ouest-de-l’Île-de-Montréal, Montreal, QC, Canada
| | - Aaron P. Johnson
- Concordia Vision Labs, Department of Psychology, Concordia University, Montreal, QC, Canada
- Centre for Interdisciplinary Research in Rehabilitation of Greater Montreal (CRIR)/Centre de Réadaptation Lethbridge-Layton-Mackay du Centre Intégré Universitaire de Santé et de Services Sociaux du Centre-Ouest-de-l’Ile-de-Montréal (CIUSSS) du Centre-Ouest-de-l’Île-de-Montréal, Montreal, QC, Canada
| | - Robert K. Koenekoop
- Paediatric Surgery and Human Genetics and Ophthalmology, Faculty of Medicine, McGill University Health Centre, Montreal QC, Canada
| | - William Seiple
- Arlene R. Gordon Research Institute, Lighthouse Guild, New York, NY, United States
- School of Medicine, New York University, New York, NY, United States
| | - Olga Overbury
- Low Vision Lab, School of Optometry, University of Montreal, Montreal, QC, Canada
- Lady Davis Institute of Medical Research, Montreal, QC, Canada
| |
Collapse
|
49
|
Olivieri O, Speziali G, Castagna A, Pattini P, Udali S, Pizzolo F, Liesinger L, Gindlhuber J, Tomin T, Schittmayer M, Birner-Gruenberger R, Cecconi D, Girelli D, Friso S, Martinelli N. The Positive Association between Plasma Myristic Acid and ApoCIII Concentrations in Cardiovascular Disease Patients Is Supported by the Effects of Myristic Acid in HepG2 Cells. J Nutr 2020; 150:2707-2715. [PMID: 32710763 DOI: 10.1093/jn/nxaa202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/06/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In the settings of primary and secondary prevention for coronary artery disease (CAD), a crucial role is played by some key molecules involved in triglyceride (TG) metabolism, such as ApoCIII. Fatty acid (FA) intake is well recognized as a main determinant of plasma lipids, including plasma TG concentration. OBJECTIVES The aim was to investigate the possible relations between the intakes of different FAs, estimated by their plasma concentrations, and circulating amounts of ApoCIII. METHODS Plasma samples were obtained from 1370 subjects with or without angiographically demonstrated CAD (mean ± SD age: 60.6 ± 11.0 y; males: 75.8%; BMI: 25.9 ± 4.6 kg/m2; CAD: 73.3%). Plasma lipid, ApoCIII, and FA concentrations were measured. Data were analyzed by regression models adjusted for FAs and other potential confounders, such as sex, age, BMI, diabetes, smoking, and lipid-lowering therapies. The in vitro effects of FAs were tested by incubating HepG2 hepatoma cells with increasing concentrations of selected FAs, and the mRNA and protein contents in the cells were quantified by real-time RT-PCR and LC-MS/MS analyses. RESULTS Among all the analyzed FAs, myristic acid (14:0) showed the most robust correlations with both TGs (R = 0.441, P = 2.6 × 10-66) and ApoCIII (R = 0.327, P = 1.1 × 10-31). By multiple regression analysis, myristic acid was the best predictor of both plasma TG and ApoCIII variability. Plasma TG and ApoCIII concentrations increased progressively at increasing concentrations of myristic acid, independently of CAD diagnosis and gender. Consistent with these data, in the in vitro experiments, an ∼2-fold increase in the expression levels of the ApoCIII mRNA and protein was observed after incubation with 250 μM myristic acid. A weaker effect (∼30% increase) was observed for palmitic acid, whereas incubation with oleic acid did not affect ApoCIII protein or gene expression. CONCLUSIONS Plasma myristic acid is associated with increased ApoCIII concentrations in cardiovascular patients. In vitro experiments indicated that myristic acid stimulates ApoCIII expression in HepG2 cells.
Collapse
Affiliation(s)
| | | | | | | | - Silvia Udali
- Department of Medicine, University of Verona, Verona, Italy
| | | | - Laura Liesinger
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,Omics Center Graz, BioTechMed-Graz, Graz, Austria
| | - Juergen Gindlhuber
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,Omics Center Graz, BioTechMed-Graz, Graz, Austria
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,Omics Center Graz, BioTechMed-Graz, Graz, Austria
| | - Matthias Schittmayer
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,Omics Center Graz, BioTechMed-Graz, Graz, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,Omics Center Graz, BioTechMed-Graz, Graz, Austria
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, Verona, Italy
| | | | | | | |
Collapse
|
50
|
Zhao M, Zhang H, Wang J, Shan D, Xu Q. Serum metabolomics analysis of the intervention effect of whole grain oats on insulin resistance induced by high-fat diet in rats. Food Res Int 2020; 135:109297. [DOI: 10.1016/j.foodres.2020.109297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/17/2022]
|