151
|
Zardini Buzatto A, Abdel Jabar M, Nizami I, Dasouki M, Li L, Abdel Rahman AM. Lipidome Alterations Induced by Cystic Fibrosis, CFTR Mutation, and Lung Function. J Proteome Res 2020; 20:549-564. [PMID: 33089695 DOI: 10.1021/acs.jproteome.0c00556] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cystic fibrosis is a genetic pathology characterized by abnormal accumulation of mucus in the respiratory, gastrointestinal, and reproductive tracts, caused by mutations in the CFTR gene. Although the classical presentation of the condition is well known, there is still a need for a better characterization of metabolic alterations related to cystic fibrosis and different genotypic mutations. We employed untargeted, comprehensive lipidomics of blood serum samples to investigate alterations in the lipid metabolism related to the pathology, mutation classes, and lung function decline. Six unique biomarker candidates were able to independently differentiate diseased individuals from healthy controls with excellent performance. Cystic fibrosis patients showed dyslipidemia for most lipid subclasses, with significantly elevated odd-chain and polyunsaturated fatty acyl lipids. Phosphatidic acids and diacylglycerols were particularly affected by different genotypic mutation classes. We selected a biomarker panel composed of four lipids, including two ceramides, one sphingomyelin, and one fatty acid, which correctly classified all validation samples from classes III and IV. A biomarker panel of five oxidized lipids was further selected to differentiate patients with reduced lung function, measured as predicted FEV1%. Our results indicate that cystic fibrosis is deeply related to lipid metabolism and provide new clues for the investigation of the disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
| | - Mai Abdel Jabar
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Imran Nizami
- Lung Transplant Section, Organ Transplant Center, King Faisal Specialist Hospital and Research Center, Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Majed Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Anas M Abdel Rahman
- Department of Genetics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Zahrawi Street, Al Maather, Riyadh 11211, Saudi Arabia.,Department of Biochemistry and Molecular Medicine, College of Medicine, Al Faisal University, Riyadh 11533, Saudi Arabia.,Department of Chemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada
| |
Collapse
|
152
|
Huang Q, Hao S, Yao X, You J, Li X, Lai D, Han C, Schilling J, Hwa KY, Thyparambil S, Whitin J, Cohen HJ, Chubb H, Ceresnak SR, McElhinney DB, Wong RJ, Shaw GM, Stevenson DK, Sylvester KG, Ling XB. High-throughput quantitation of serological ceramides/dihydroceramides by LC/MS/MS: Pregnancy baseline biomarkers and potential metabolic messengers. J Pharm Biomed Anal 2020; 192:113639. [PMID: 33017796 DOI: 10.1016/j.jpba.2020.113639] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/08/2023]
Abstract
Ceramides and dihydroceramides are sphingolipids that present in abundance at the cellular membrane of eukaryotes. Although their metabolic dysregulation has been implicated in many diseases, our knowledge about circulating ceramide changes during the pregnancy remains limited. In this study, we present the development and validation of a high-throughput liquid chromatography-tandem mass spectrometric method for simultaneous quantification of 16 ceramides and 10 dihydroceramides in human serum within 5 min. by using stable isotope-labeled ceramides as internal standards. This method employs a protein precipitation method for high throughput sample preparation, reverse phase isocratic elusion for chromatographic separation, and Multiple Reaction Monitoring for mass spectrometric detection. To qualify for clinical applications, our assay has been validated against the FDA guidelines for Lower Limit of Quantitation (1 nM), linearity (R2>0.99), precision (imprecision<15 %), accuracy (inaccuracy<15 %), extraction recovery (>90 %), stability (>85 %), and carryover (<0.01 %). With enhanced sensitivity and specificity from this method, we have, for the first time, determined the serological levels of ceramides and dihydroceramides to reveal unique temporal gestational patterns. Our approach could have value in providing insights into disorders of pregnancy.
Collapse
Affiliation(s)
| | - Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States; Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, CA, United States
| | | | - Jin You
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiao Li
- mProbe Inc, Mountain View, CA, United States
| | - Donghai Lai
- mProbe Inc, Mountain View, CA, United States
| | - Chunle Han
- mProbe Inc, Mountain View, CA, United States
| | | | | | | | - John Whitin
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Harvey J Cohen
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Henry Chubb
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Scott R Ceresnak
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Doff B McElhinney
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, United States; Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, CA, United States
| | - Ronald J Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - David K Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Karl G Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Xuefeng B Ling
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, CA, United States; Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
153
|
Lee YF, Sim XY, Teh YH, Ismail MN, Greimel P, Murugaiyah V, Ibrahim B, Gam LH. The effects of high-fat diet and metformin on urinary metabolites in diabetes and prediabetes rat models. Biotechnol Appl Biochem 2020; 68:1014-1026. [PMID: 32931602 DOI: 10.1002/bab.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022]
Abstract
High-fat diet (HFD) interferes with the dietary plan of patients with type 2 diabetes mellitus (T2DM). However, many diabetes patients consume food with higher fat content for a better taste bud experience. In this study, we examined the effect of HFD on rats at the early onset of diabetes and prediabetes by supplementing their feed with palm olein oil to provide a fat content representing 39% of total calorie intake. Urinary profile generated from liquid chromatography-mass spectrometry analysis was used to construct the orthogonal partial least squares discriminant analysis (OPLS-DA) score plots. The data provide insights into the physiological state of an organism. Healthy rats fed with normal chow (NC) and HFD cannot be distinguished by their urinary metabolite profiles, whereas diabetic and prediabetic rats showed a clear separation in OPLS-DA profile between the two diets, indicating a change in their physiological state. Metformin treatment altered the metabolomics profiles of diabetic rats and lowered their blood sugar levels. For prediabetic rats, metformin treatment on both NC- and HFD-fed rats not only reduced their blood sugar levels to normal but also altered the urinary metabolite profile to be more like healthy rats. The use of metformin is therefore beneficial at the prediabetes stage.
Collapse
Affiliation(s)
- Yan-Fen Lee
- USM-RIKEN International Centre of Aging Science, USM, Minden, Penang, Malaysia.,School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Xuan-Yi Sim
- USM-RIKEN International Centre of Aging Science, USM, Minden, Penang, Malaysia.,School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Ying-Hui Teh
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Mohd Nazri Ismail
- Analytical Biochemistry Research Centre (ABrC), USM, Minden, Penang, Malaysia
| | - Peter Greimel
- Laboratory for Cell Function Dynamics, RIKEN Centre for Brain Sciences, Wako, Saitama, Japan
| | | | - Baharudin Ibrahim
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| | - Lay-Harn Gam
- USM-RIKEN International Centre of Aging Science, USM, Minden, Penang, Malaysia.,School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang, Malaysia
| |
Collapse
|
154
|
Lopez A, Vasconi M, Battini M, Mattiello S, Moretti VM, Bellagamba F. Intrinsic and Extrinsic Quality Attributes of Fresh and Semi-Hard Goat Cheese from Low- and High-Input Farming Systems. Animals (Basel) 2020; 10:ani10091567. [PMID: 32899239 PMCID: PMC7552214 DOI: 10.3390/ani10091567] [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: 07/30/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 11/16/2022] Open
Abstract
In this study, we investigated the lipid composition of fresh and semi-hard goat cheese produced in three Italian farms as well as the welfare assessment of goats reared in these farms. The fatty acid (FA) profile of cheese samples were found to be strictly related to the livestock system. Cheese collected from farms in which goats were allowed to graze and were fed diets with a higher forage/concentrate (F/C) ratio showed a FA profile represented by higher contents of health-promoting fatty acids. In the same samples, the health lipid indices showed the most favorable values. Conversely, cheese samples collected from a conventional-lowland farm, where goats were fed with higher amounts of concentrates and lower F/C ratio, presented a lower nutritional quality, characterized by the worst results for what concerns the health lipid indices. Then, we built a multivariate model able to discriminate samples coming from farms managed by a low-input system from those coming from farm managed by a high-input system. The comparison of animal welfare measurements and fatty acids data showed that a better intrinsic quality of low-input farms did not always correspond to better extrinsic quality, suggesting that the information on the livestock system is not always enough to provide consumers with complete awareness of the total product quality.
Collapse
Affiliation(s)
- Annalaura Lopez
- Department of Veterinary Medicine, Università degli Studi di Milano, Via dell’Università, 6, 26900 Lodi, Italy; (A.L.); (V.M.M.); (F.B.)
| | - Mauro Vasconi
- Department of Veterinary Medicine, Università degli Studi di Milano, Via dell’Università, 6, 26900 Lodi, Italy; (A.L.); (V.M.M.); (F.B.)
- Correspondence:
| | - Monica Battini
- Department of Agricultural and Environmental Sciences, Production, Landscape, Agroenergy, Università degli Studi di Milano, Via Celoria, 2, 20133 Milano, Italy; (M.B.); (S.M.)
| | - Silvana Mattiello
- Department of Agricultural and Environmental Sciences, Production, Landscape, Agroenergy, Università degli Studi di Milano, Via Celoria, 2, 20133 Milano, Italy; (M.B.); (S.M.)
| | - Vittorio Maria Moretti
- Department of Veterinary Medicine, Università degli Studi di Milano, Via dell’Università, 6, 26900 Lodi, Italy; (A.L.); (V.M.M.); (F.B.)
| | - Federica Bellagamba
- Department of Veterinary Medicine, Università degli Studi di Milano, Via dell’Università, 6, 26900 Lodi, Italy; (A.L.); (V.M.M.); (F.B.)
| |
Collapse
|
155
|
Lytrivi M, Ghaddar K, Lopes M, Rosengren V, Piron A, Yi X, Johansson H, Lehtiö J, Igoillo-Esteve M, Cunha DA, Marselli L, Marchetti P, Ortsäter H, Eizirik DL, Cnop M. Combined transcriptome and proteome profiling of the pancreatic β-cell response to palmitate unveils key pathways of β-cell lipotoxicity. BMC Genomics 2020; 21:590. [PMID: 32847508 PMCID: PMC7448506 DOI: 10.1186/s12864-020-07003-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Prolonged exposure to elevated free fatty acids induces β-cell failure (lipotoxicity) and contributes to the pathogenesis of type 2 diabetes. In vitro exposure of β-cells to the saturated free fatty acid palmitate is a valuable model of lipotoxicity, reproducing features of β-cell failure observed in type 2 diabetes. In order to map the β-cell response to lipotoxicity, we combined RNA-sequencing of palmitate-treated human islets with iTRAQ proteomics of insulin-secreting INS-1E cells following a time course exposure to palmitate. RESULTS Crossing transcriptome and proteome of palmitate-treated β-cells revealed 85 upregulated and 122 downregulated genes at both transcript and protein level. Pathway analysis identified lipid metabolism, oxidative stress, amino-acid metabolism and cell cycle pathways among the most enriched palmitate-modified pathways. Palmitate induced gene expression changes compatible with increased free fatty acid mitochondrial import and β-oxidation, decreased lipogenesis and modified cholesterol transport. Palmitate modified genes regulating endoplasmic reticulum (ER) function, ER-to-Golgi transport and ER stress pathways. Furthermore, palmitate modulated cAMP/protein kinase A (PKA) signaling, inhibiting expression of PKA anchoring proteins and downregulating the GLP-1 receptor. SLC7 family amino-acid transporters were upregulated in response to palmitate but this induction did not contribute to β-cell demise. To unravel critical mediators of lipotoxicity upstream of the palmitate-modified genes, we identified overrepresented transcription factor binding sites and performed network inference analysis. These identified LXR, PPARα, FOXO1 and BACH1 as key transcription factors orchestrating the metabolic and oxidative stress responses to palmitate. CONCLUSIONS This is the first study to combine transcriptomic and sensitive time course proteomic profiling of palmitate-exposed β-cells. Our results provide comprehensive insight into gene and protein expression changes, corroborating and expanding beyond previous findings. The identification of critical drivers and pathways of the β-cell lipotoxic response points to novel therapeutic targets for type 2 diabetes.
Collapse
Affiliation(s)
- Maria Lytrivi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium.,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Kassem Ghaddar
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Miguel Lopes
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Victoria Rosengren
- Diabetes Research Unit, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Anthony Piron
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Henrik Johansson
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, 171 21, Solna, Sweden
| | - Janne Lehtiö
- Clinical Proteomics Mass Spectrometry, Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, 171 21, Solna, Sweden
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Daniel A Cunha
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Henrik Ortsäter
- Diabetes Research Unit, Department of Clinical Science and Education, Sodersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, CP-618, Route de Lennik 808, 1070, Brussels, Belgium. .,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
156
|
Sun L, Zong G, Li H, Lin X. Fatty acids and cardiometabolic health: a review of studies in Chinese populations. Eur J Clin Nutr 2020; 75:253-266. [PMID: 32801302 DOI: 10.1038/s41430-020-00709-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/19/2020] [Accepted: 08/04/2020] [Indexed: 11/09/2022]
Abstract
Rapid nutrition transition from plant-based traditional diet to westernized diet has led to dramatically heightening burdens of cardiometabolic diseases in China in past decades. Recently, national surveys reported that poor dietary quality including low marine n-3 fatty acids and high intakes of red meat and processed meat was associated with considerably elevated cardiometabolic deaths. Previous studies mainly from Western population-based cohorts have indicated that not only fat quantity but also quality linked with different cardiometabolic outcomes. Compared with Western peoples, Asian peoples, including Chinese, are known to have different dietary patterns and lifestyle, as well as genetic heterogeneities, which may modify fatty acid metabolism and disease susceptibility in certain degree. To date, there were limited prospective studies investigating the relationships between fatty acids and cardiometabolic disease outcomes in Chinese, and most existing studies were cross-sectional nature and within one or two region(s). Notably, shifting dietary patterns could change not only amount, types, and ratio of fatty acids accounting for overall energy intake, but also their food sources and ratio to other macronutrients. Moreover, large geographic and urban-rural variations in prevalence of cardiometabolic diseases among Chinese may also reflect the effects of socioeconomic development and local diets on health status. Therefore, current review will summarize available literatures with more focus on the Chinese-based studies which may extend current knowledge about the roles of fatty acids in pathogenesis of cardiometabolic diseases for Asian populations and also provide useful information for trans-ethnic comparisons with other populations.
Collapse
Affiliation(s)
- Liang Sun
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Geng Zong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huaixing Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xu Lin
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China. .,Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
157
|
Kumar AA, Satheesh G, Vijayakumar G, Chandran M, Prabhu PR, Simon L, Kutty VR, Kartha CC, Jaleel A. Plasma leptin level mirrors metabolome alterations in young adults. Metabolomics 2020; 16:87. [PMID: 32772182 DOI: 10.1007/s11306-020-01708-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/03/2020] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Leptin is known to regulate pathways of energy metabolism, reproduction, and control appetite. Whether plasma leptin levels reflect changes in metabolites of these pathways is unknown. OBJECTIVES We aimed to find whether there is an association between leptin levels and levels of metabolites of energy and hormone metabolism. METHODS We performed an untargeted metabolomics analysis of plasma from 110 healthy adults (men: women = 1:1; aged 18-40 years), using liquid chromatography-tandem mass spectrometry. Blood samples were collected from all the study subjects in the fasting state. Clinical features and markers of obesity and Type 2 diabetes mellitus (T2DM) were assessed in all. The association between levels of metabolites and clinical and biochemical parameters was identified using the multivariable-adjusted linear regression model and PLS-DA analysis. RESULTS The leptin level was found to have a significant association with a substantial number of metabolites in women and men. Leptin level was positively associated with glycocholic acid and arachidic acid, metabolites related to energy metabolisms, pregnanediol-3-glucuronide, a metabolite of progesterone metabolism, and quercetin 3'-sulfate, a diet-derived metabolite. Leptin level was negatively associated with ponasteroside A and barringtogenol C levels. Leptin level was positively correlated with adiponectin and negatively with total calorie intake and levels of triglyceride and very-low-density lipoprotein. Leptin levels were associated with lipid and sex hormone metabolism in women, while metabolites involved in amino acid metabolism were correlated to leptin in men. CONCLUSION Our study indicates that leptin level reflects metabolome alterations and hence could be a useful marker to detect early changes in energy and hormone metabolisms.
Collapse
Affiliation(s)
- A Aneesh Kumar
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India
- Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gopika Satheesh
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India
| | - Gadadharan Vijayakumar
- Medical Trust Hospital and Diabetes Care Centre, Kulanada, Pathanamthitta, Kerala, India
| | - Mahesh Chandran
- Mass Spectrometry and Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India
| | - Priya R Prabhu
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India
| | - Leena Simon
- Medical Trust Hospital and Diabetes Care Centre, Kulanada, Pathanamthitta, Kerala, India
| | - Vellappillil Raman Kutty
- Achutha Menon Centre for Health Science Studies, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala, 695012, India
| | - Chandrasekharan C Kartha
- Society for Continuing Medical Education & Research, Kerala Institute of Medical Sciences, Thiruvananthapuram, Kerala, 695029, India
| | - Abdul Jaleel
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India.
- Mass Spectrometry and Proteomics Core Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695012, India.
- Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
158
|
Lai M, Al Rijjal D, Röst HL, Dai FF, Gunderson EP, Wheeler MB. Underlying dyslipidemia postpartum in women with a recent GDM pregnancy who develop type 2 diabetes. eLife 2020; 9:59153. [PMID: 32748787 PMCID: PMC7417169 DOI: 10.7554/elife.59153] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/18/2020] [Indexed: 12/15/2022] Open
Abstract
Approximately, 35% of women with Gestational Diabetes (GDM) progress to Type 2 Diabetes (T2D) within 10 years. However, links between GDM and T2D are not well understood. We used a well-characterised GDM prospective cohort of 1035 women following up to 8 years postpartum. Lipidomics profiling covering >1000 lipids was performed on fasting plasma samples from participants 6–9 week postpartum (171 incident T2D vs. 179 controls). We discovered 311 lipids positively and 70 lipids negatively associated with T2D risk. The upregulation of glycerolipid metabolism involving triacylglycerol and diacylglycerol biosynthesis suggested activated lipid storage before diabetes onset. In contrast, decreased sphingomyelines, hexosylceramide and lactosylceramide indicated impaired sphingolipid metabolism. Additionally, a lipid signature was identified to effectively predict future diabetes risk. These findings demonstrate an underlying dyslipidemia during the early postpartum in those GDM women who progress to T2D and suggest endogenous lipogenesis may be a driving force for future diabetes onset.
Collapse
Affiliation(s)
- Mi Lai
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Dana Al Rijjal
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Hannes L Röst
- Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Ontario, Canada
| | - Feihan F Dai
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Erica P Gunderson
- Kaiser Permanente Northern California, Division of Research, Oakland, United States
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada.,Advanced Diagnostics, Metabolism, Toronto General Research Institute, Ontario, Canada
| |
Collapse
|
159
|
Jiang Z, Hayashi T, Kashima K, Kurotani K, Shirouchi B, Mizoue T, Sato M. Alteration of Serum Phospholipid n‐6 Polyunsaturated Fatty Acid Compositions inNonalcoholicFatty Liver Disease in the Japanese Population: A Cross‐Sectional Study. Lipids 2020; 55:599-614. [DOI: 10.1002/lipd.12251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 01/12/2023]
Affiliation(s)
- Zhe Jiang
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School Kyushu University 744 Motooka Nishi‐ku, Fukuoka 819‐0395 Japan
| | - Takuya Hayashi
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School Kyushu University 744 Motooka Nishi‐ku, Fukuoka 819‐0395 Japan
| | - Kentaro Kashima
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School Kyushu University 744 Motooka Nishi‐ku, Fukuoka 819‐0395 Japan
| | - Kayo Kurotani
- Department of Epidemiology and Prevention, Center for Clinical Sciences National Center for Global Health and Medicine 1‐21‐1, Toyama, Shinjuku‐ku, Tokyo 162‐8655 Japan
- Department of Nutritional Epidemiology and Shokuiku, National Institute of Health and Nutrition National Institutes of Biomedical Innovation, Health and Nutrition 1‐23‐1 Toyama, Shinjuku‐ku, Tokyo 162‐8636 Japan
| | - Bungo Shirouchi
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School Kyushu University 744 Motooka Nishi‐ku, Fukuoka 819‐0395 Japan
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences National Center for Global Health and Medicine 1‐21‐1, Toyama, Shinjuku‐ku, Tokyo 162‐8655 Japan
| | - Masao Sato
- Laboratory of Nutrition Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School Kyushu University 744 Motooka Nishi‐ku, Fukuoka 819‐0395 Japan
| |
Collapse
|
160
|
Jenkins B, Ronis M, Koulman A. LC-MS Lipidomics: Exploiting a Simple High-Throughput Method for the Comprehensive Extraction of Lipids in a Ruminant Fat Dose-Response Study. Metabolites 2020; 10:E296. [PMID: 32709069 PMCID: PMC7407148 DOI: 10.3390/metabo10070296] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/07/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
Typical lipidomics methods incorporate a liquid-liquid extraction with LC-MS quantitation; however, the classic sample extraction methods are not high-throughput and do not perform well at extracting the full range of lipids especially, the relatively polar species (e.g., acyl-carnitines and glycosphingolipids). In this manuscript, we present a novel sample extraction protocol, which produces a single phase supernatant suitable for high-throughput applications that offers greater performance in extracting lipids across the full spectrum of species. We applied this lipidomics pipeline to a ruminant fat dose-response study to initially compare and validate the different extraction protocols but also to investigate complex lipid biomarkers of ruminant fat intake (adjoining onto simple odd chain fatty acid correlations). We have found 100 lipids species with a strong correlation with ruminant fat intake. This novel sample extraction along with the LC-MS pipeline have shown to be sensitive, robust and hugely informative (>450 lipids species semi-quantified): with a sample preparation throughput of over 100 tissue samples per day and an estimated ~1000 biological fluid samples per day. Thus, this work facilitating both the epidemiological involvement of ruminant fat, research into odd chain lipids and also streamlining the field of lipidomics (both by sample preparation methods and data presentation).
Collapse
Affiliation(s)
- Benjamin Jenkins
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Pathology Building Level 4, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| | - Martin Ronis
- College of Medicine, Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Centre, 1901 Perdido Str., New Orleans, LA 70112, USA;
| | - Albert Koulman
- NIHR BRC Core Metabolomics and Lipidomics Laboratory, University of Cambridge, Pathology Building Level 4, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK;
| |
Collapse
|
161
|
Altered Metabolome of Lipids and Amino Acids Species: A Source of Early Signature Biomarkers of T2DM. J Clin Med 2020; 9:jcm9072257. [PMID: 32708684 PMCID: PMC7409008 DOI: 10.3390/jcm9072257] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus, a disease of modern civilization, is considered the major mainstay of mortalities around the globe. A great number of biochemical changes have been proposed to occur at metabolic levels between perturbed glucose, amino acid, and lipid metabolism to finally diagnoe diabetes mellitus. This window period, which varies from person to person, provides us with a unique opportunity for early detection, delaying, deferral and even prevention of diabetes. The early detection of hyperglycemia and dyslipidemia is based upon the detection and identification of biomarkers originating from perturbed glucose, amino acid, and lipid metabolism. The emerging “OMICS” technologies, such as metabolomics coupled with statistical and bioinformatics tools, proved to be quite useful to study changes in physiological and biochemical processes at the metabolic level prior to an eventual diagnosis of DM. Approximately 300–400 such metabolites have been reported in the literature and are considered as predicting or risk factor-reporting metabolic biomarkers for this metabolic disorder. Most of these metabolites belong to major classes of lipids, amino acids and glucose. Therefore, this review represents a snapshot of these perturbed plasma/serum/urinary metabolic biomarkers showing a significant correlation with the future onset of diabetes and providing a foundation for novel early diagnosis and monitoring the progress of metabolic syndrome at early symptomatic stages. As most metabolites also find their origin from gut microflora, metabolism and composition of gut microflora also vary between healthy and diabetic persons, so we also summarize the early changes in the gut microbiome which can be used for the early diagnosis of diabetes.
Collapse
|
162
|
Ojwang AA, Smuts CM, Zec M, Wentzel-Viljoen E, Kruger IM, Kruger HS. Comparison of dietary and plasma phospholipid fatty acids between normal weight and overweight black South Africans according to metabolic health: The PURE study. Prostaglandins Leukot Essent Fatty Acids 2020; 158:102039. [PMID: 31780327 DOI: 10.1016/j.plefa.2019.102039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Information regarding circulating fatty acids (FA) in association with metabolic health in black Africans is scarce, while the usefulness of circulating FAs as biomarkers of dietary fat intake and predictors for medical conditions is increasing. OBJECTIVE We compared eleven dietary and the levels of 26 plasma phospholipid FAs in metabolically healthy and unhealthy phenotypes in black South African adults. METHODS Adults from the South African arm of the Prospective Urban and Rural Epidemiology study baseline (n = 711) were categorised into four groups, namely normal weight without metabolic syndrome (MetS) (MHNW), normal weight with MetS (MUNW), metabolically healthy overweight/obese (MHO) and metabolically unhealthy overweight/obese (MUO). Dietary and plasma phospholipid FAs were measured by a quantitative food frequency questionnaire and gas chromatography-tandem mass spectrometry, respectively. We compared dietary FAs, plasma phospholipid FAs, and estimated desaturase activity between the metabolic status groups using ANCOVA adjusted for age and energy intake. RESULTS MetS was diagnosed in 35% of the participants. After adjustment for age and total energy intake, in comparison to the MHNW reference group, saturated dietary FAs (C14:0 to C18:0) and alpha-linolenic acid intakes were higher in both overweight/obese groups (MHO and MUO), while linoleic acid intakes were higher in the MUO group only. Plasma levels of most saturated FAs (C18:0 to C22:0) and PUFAs were higher, whereas selected MUFAs, palmitic acid, and estimated desaturase activities were lower in the overweight/obese groups. CONCLUSIONS The overweight groups generally had higher fat intakes than normal-weight groups, but lower plasma levels of palmitic, palmitoleic, oleic, cis-vaccenic and estimated desaturase activities. Therefore, in this population, lower plasma levels of palmitic, palmitoleic, oleic, and cis-vaccenic acids and decreased estimated desaturase activities may be biomarkers of abnormal metabolic health in overweight/obese study participants.
Collapse
Affiliation(s)
- A A Ojwang
- Centre of Excellence for Nutrition, North-West University, 2520 Potchefstroom, South Africa; Technical University of Kenya, Kenya.
| | - C M Smuts
- Centre of Excellence for Nutrition, North-West University, 2520 Potchefstroom, South Africa.
| | - M Zec
- Centre of Excellence for Nutrition, North-West University, 2520 Potchefstroom, South Africa; Centre of Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Serbia
| | - E Wentzel-Viljoen
- Centre of Excellence for Nutrition, North-West University, 2520 Potchefstroom, South Africa.
| | - I M Kruger
- Africa Unit for Transdisciplinary Health Research, North-West University, 2520 Potchefstroom, South Africa.
| | - H S Kruger
- Centre of Excellence for Nutrition, North-West University, 2520 Potchefstroom, South Africa; Medical Research Council Extra Mural Unit: Hypertension and CVD, North-West University, 2520 Potchefstroom, South Africa.
| |
Collapse
|
163
|
Deng S, Li D, Liu X, Cai Z, Wei W, Chen J, Zhang L. Serum metabolomic investigations of mulberry leaf powder supplementation in Chinese Erhualian pigs. JOURNAL OF ANIMAL AND FEED SCIENCES 2020. [DOI: 10.22358/jafs/124043/2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
164
|
Chen G, Li Y, Zeng F, Deng G, Liang J, Wang J, Su Y, Chen Y, Mao L, Liu Z, Bao W, Zhang Z. Biomarkers of fatty acids and risk of type 2 diabetes: a systematic review and meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 2020; 61:2705-2718. [PMID: 32598176 DOI: 10.1080/10408398.2020.1784839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We aimed to quantify and generate comprehensive evidence on the associations of different fatty acids (FAs) with the risk of type 2 diabetes mellitus (T2DM). PubMed, MEDLINE, and Embase were searched for prospective cohort studies that examined the association between FA biomarkers and the risk of T2DM published before 18 May 2020. Random-effects meta-analyses of the effects of FA concentration on T2DM were performed. Thirty-three studies involving 95,810 adults (19,904 cases) were included. Divergent associations of different types of FAs with type 2 diabetes were observed. The pooled relative risk (RRs) of T2DM comparing the top versus the bottom tertile of saturated FAs (C14:0, C16:0, C18:0, total saturated FAs), monounsaturated FA (C16:1 n-7), polyunsaturated FA (C20:3 n-6, C22:4 n-6), and Δ-6-desaturase activities ranged from 1.19 to 1.80. Interestingly, unlike previous studies, we found a negative correlation between odd-chain saturated FAs (C15:0, C17:0), trans-FAs (trans-C16:1 n-7), total n-6, Δ-5-desaturase activities and risk of T2DM. The pooled RRs of T2DM comparing the top versus the bottom tertile of these FAs ranged from 0.62 to 0.78. No associations with T2DM were observed for the other FAs. Considerable heterogeneity was observed in our study, and no definitive conclusions can be made until further investigation has been carried out.
Collapse
Affiliation(s)
- Gengdong Chen
- Department of Obstetrics, Foshan Institute of Fetal Medicine, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Yan Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fangfang Zeng
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Guifang Deng
- Department of Clinical Nutrition, Nanshan People's Hospital, Shenzhen, China
| | - Jingjing Liang
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jue Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yixiang Su
- Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuming Chen
- Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Limei Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhengping Liu
- Department of Obstetrics, Foshan Institute of Fetal Medicine, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Wei Bao
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, Iowa, USA.,Obesity Research and Education Initiative, University of Iowa, Iowa City, Iowa, USA.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| | - Zheqing Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
165
|
Saturated Fats and Health: A Reassessment and Proposal for Food-Based Recommendations: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76:844-857. [PMID: 32562735 DOI: 10.1016/j.jacc.2020.05.077] [Citation(s) in RCA: 270] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 12/29/2022]
Abstract
The recommendation to limit dietary saturated fatty acid (SFA) intake has persisted despite mounting evidence to the contrary. Most recent meta-analyses of randomized trials and observational studies found no beneficial effects of reducing SFA intake on cardiovascular disease (CVD) and total mortality, and instead found protective effects against stroke. Although SFAs increase low-density lipoprotein (LDL) cholesterol, in most individuals, this is not due to increasing levels of small, dense LDL particles, but rather larger LDL particles, which are much less strongly related to CVD risk. It is also apparent that the health effects of foods cannot be predicted by their content in any nutrient group without considering the overall macronutrient distribution. Whole-fat dairy, unprocessed meat, and dark chocolate are SFA-rich foods with a complex matrix that are not associated with increased risk of CVD. The totality of available evidence does not support further limiting the intake of such foods.
Collapse
|
166
|
Bishop CA, Schulze MB, Klaus S, Weitkunat K. The branched‐chain amino acids valine and leucine have differential effects on hepatic lipid metabolism. FASEB J 2020; 34:9727-9739. [DOI: 10.1096/fj.202000195r] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/21/2020] [Accepted: 05/15/2020] [Indexed: 01/17/2023]
Affiliation(s)
- Christopher A. Bishop
- Department of Physiology of Energy Metabolism German Institute of Human Nutrition Potsdam‐Rehbruecke (DIfE) Nuthetal Germany
- Institute of Nutrition Science University of Potsdam Nuthetal Germany
| | - Matthias B. Schulze
- Institute of Nutrition Science University of Potsdam Nuthetal Germany
- Department of Molecular Epidemiology German Institute of Human Nutrition Potsdam‐Rehbruecke (DIfE) Nuthetal Germany
| | - Susanne Klaus
- Department of Physiology of Energy Metabolism German Institute of Human Nutrition Potsdam‐Rehbruecke (DIfE) Nuthetal Germany
- Institute of Nutrition Science University of Potsdam Nuthetal Germany
| | - Karolin Weitkunat
- Department of Physiology of Energy Metabolism German Institute of Human Nutrition Potsdam‐Rehbruecke (DIfE) Nuthetal Germany
| |
Collapse
|
167
|
Comprehensive Characterization of Phospholipid Isomers in Human Platelets. JOURNAL OF ANALYSIS AND TESTING 2020. [DOI: 10.1007/s41664-020-00137-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
168
|
Li D, Misialek JR, Jing M, Tsai MY, Eckfeldt JH, Steffen LM, Knopman D, Wruck L, Gottesman R, Mosley TH, Sharrett AR, Alonso A. Plasma phospholipid very-long-chain SFAs in midlife and 20-year cognitive change in the Atherosclerosis Risk in Communities (ARIC): a cohort study. Am J Clin Nutr 2020; 111:1252-1258. [PMID: 32320012 PMCID: PMC7266687 DOI: 10.1093/ajcn/nqaa048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/24/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Very-long-chain SFAs (VLSFAs) have recently gained considerable attention as having beneficial effects on health and aging. OBJECTIVES The objective of this study was to assess the associations of plasma phospholipid VLSFAs [arachidic acid (20:0), behenic acid (22:0), tricosanoic acid (23:0), and lignoceric acid (24:0)] with 20-y cognitive decline in the Atherosclerosis Risk in Communities (ARIC) participants. Furthermore, this study compared the associations of plasma phospholipid VLSFAs with 5 common groups of fatty acids [i.e., total SFAs, total MUFAs, total ω-3 (n-3) PUFAs, total marine-derived ω-3 PUFAs, total ω-6 PUFAs]. METHODS This study used a cohort study design of 3229 ARIC participants enrolled at the Minnesota field center. Fatty acids were measured at visit 1 (1987-1989); and cognition was assessed at visits 2 (1990-1992), 4 (1996-1998), and 5 (2011-2013) using 3 tests: the Delayed Word Recall Test (DWRT), the Digit-Symbol Substitution Test (DSST), and the Word Fluency Test (WFT). RESULTS Higher proportions of plasma phospholipid total VLSFAs and each individual VLSFA were associated with less decline in WFT, a test of verbal fluency. For example, 1 SD higher in total VLSFAs at baseline was associated with 0.057 SD (95% CI: 0.018, 0.096, P = 0.004) less cognitive decline over 20 y as measured by WFT score. None of the 5 common fatty acid groups were associated with change in WFT, but a higher proportion of plasma phospholipid total MUFAs was associated with greater decline in DWRT; higher total ω-6 PUFAs with less decline in DWRT; and higher total ω-3 and total marine-derived ω-3 PUFAs with less decline in DSST. CONCLUSIONS This study suggests that higher proportions of plasma phospholipid VLSFAs in midlife may be associated with less 20-y cognitive decline.
Collapse
Affiliation(s)
- Danni Li
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey R Misialek
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Ma Jing
- Department of Population Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - John H Eckfeldt
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - David Knopman
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Lisa Wruck
- Duke Clinical Research Institute, Duke University, Durham, NC, USA
| | - Rebecca Gottesman
- Department of Neurology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Tom H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - A Richey Sharrett
- Department of Epidemiology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
169
|
Fatty acids in the de novo lipogenesis pathway and incidence of type 2 diabetes: A pooled analysis of prospective cohort studies. PLoS Med 2020; 17:e1003102. [PMID: 32530938 PMCID: PMC7292352 DOI: 10.1371/journal.pmed.1003102] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND De novo lipogenesis (DNL) is the primary metabolic pathway synthesizing fatty acids from carbohydrates, protein, or alcohol. Our aim was to examine associations of in vivo levels of selected fatty acids (16:0, 16:1n7, 18:0, 18:1n9) in DNL with incidence of type 2 diabetes (T2D). METHODS AND FINDINGS Seventeen cohorts from 12 countries (7 from Europe, 7 from the United States, 1 from Australia, 1 from Taiwan; baseline years = 1970-1973 to 2006-2010) conducted harmonized individual-level analyses of associations of DNL-related fatty acids with incident T2D. In total, we evaluated 65,225 participants (mean ages = 52.3-75.5 years; % women = 20.4%-62.3% in 12 cohorts recruiting both sexes) and 15,383 incident cases of T2D over the 9-year follow-up on average. Cohort-specific association of each of 16:0, 16:1n7, 18:0, and 18:1n9 with incident T2D was estimated, adjusted for demographic factors, socioeconomic characteristics, alcohol, smoking, physical activity, dyslipidemia, hypertension, menopausal status, and adiposity. Cohort-specific associations were meta-analyzed with an inverse-variance-weighted approach. Each of the 4 fatty acids positively related to incident T2D. Relative risks (RRs) per cohort-specific range between midpoints of the top and bottom quintiles of fatty acid concentrations were 1.53 (1.41-1.66; p < 0.001) for 16:0, 1.40 (1.33-1.48; p < 0.001) for 16:1n-7, 1.14 (1.05-1.22; p = 0.001) for 18:0, and 1.16 (1.07-1.25; p < 0.001) for 18:1n9. Heterogeneity was seen across cohorts (I2 = 51.1%-73.1% for each fatty acid) but not explained by lipid fractions and global geographical regions. Further adjusted for triglycerides (and 16:0 when appropriate) to evaluate associations independent of overall DNL, the associations remained significant for 16:0, 16:1n7, and 18:0 but were attenuated for 18:1n9 (RR = 1.03, 95% confidence interval (CI) = 0.94-1.13). These findings had limitations in potential reverse causation and residual confounding by imprecisely measured or unmeasured factors. CONCLUSIONS Concentrations of fatty acids in the DNL were positively associated with T2D incidence. Our findings support further work to investigate a possible role of DNL and individual fatty acids in the development of T2D.
Collapse
|
170
|
Gori M, Altomare A, Cocca S, Solida E, Ribolsi M, Carotti S, Rainer A, Francesconi M, Morini S, Cicala M, Pier Luca Guarino M. Palmitic Acid Affects Intestinal Epithelial Barrier Integrity and Permeability In Vitro. Antioxidants (Basel) 2020; 9:antiox9050417. [PMID: 32414055 PMCID: PMC7278681 DOI: 10.3390/antiox9050417] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/05/2020] [Accepted: 05/10/2020] [Indexed: 12/15/2022] Open
Abstract
Palmitic acid (PA), a long-chain saturated fatty acid, might activate innate immune cells. PA plays a role in chronic liver disease, diabetes and Crohn’s disease, all of which are associated with impaired intestinal permeability. We investigated the effect of PA, at physiological postprandial intestinal concentrations, on gut epithelium as compared to lipopolysaccharide (LPS) and ethanol, using an in vitro gut model, the human intestinal epithelial cell line Caco-2 grown on transwell inserts. Cytotoxicity and oxidative stress were evaluated; epithelial barrier integrity was investigated by measuring the paracellular flux of fluorescein, and through RT-qPCR and immunofluorescence of tight junction (TJ) and adherens junction (AJ) mRNAs and proteins, respectively. In PA-exposed Caco-2 monolayers, cytotoxicity and oxidative stress were not detected. A significant increase in fluorescein flux was observed in PA-treated monolayers, after 90 min and up to 360 min, whereas with LPS and ethanol, this was only observed at later time-points. Gene expression and immunofluorescence analysis showed TJ and AJ alterations only in PA-exposed monolayers. In conclusion, PA affected intestinal permeability without inducing cytotoxicity or oxidative stress. This effect seemed to be faster and stronger than those with LPS and ethanol. Thus, we hypothesized that PA, besides having an immunomodulatory effect, might play a role in inflammatory and functional intestinal disorders in which the intestinal permeability is altered.
Collapse
Affiliation(s)
- Manuele Gori
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
- Correspondence: ; Tel: +39-062-2541-9108
| | - Annamaria Altomare
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
| | - Silvia Cocca
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
| | - Eleonora Solida
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
| | - Mentore Ribolsi
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
| | - Simone Carotti
- Microscopic and Ultrastructural Anatomy Unit, Università Campus Bio-Medico di Roma, via Alvaro del Portillo 21, 00128 Rome, Italy; (S.C.); (M.F.); (S.M.)
| | - Alberto Rainer
- Tissue Engineering Laboratory, Department of Engineering, Università Campus Bio-Medico di Roma, via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Maria Francesconi
- Microscopic and Ultrastructural Anatomy Unit, Università Campus Bio-Medico di Roma, via Alvaro del Portillo 21, 00128 Rome, Italy; (S.C.); (M.F.); (S.M.)
| | - Sergio Morini
- Microscopic and Ultrastructural Anatomy Unit, Università Campus Bio-Medico di Roma, via Alvaro del Portillo 21, 00128 Rome, Italy; (S.C.); (M.F.); (S.M.)
| | - Michele Cicala
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
| | - Michele Pier Luca Guarino
- Gastroenterology Unit, Departmental Faculty of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (A.A.); (S.C.); (E.S.); (M.R.); (M.C.); (M.P.L.G.)
| |
Collapse
|
171
|
Saadat N, Lydic TA, Misra DP, Dailey R, Walker DS, Giurgescu C. Lipidome Profiles Are Related to Depressive Symptoms and Preterm Birth Among African American Women. Biol Res Nurs 2020; 22:354-361. [PMID: 32383404 DOI: 10.1177/1099800420923032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
African American women have the highest rate of preterm birth (PTB; <37 completed weeks' gestation) of any racial and ethnic group in the United States (14.1%). Depressive symptoms (DS) have been linked to PTB risk of African American women. We hypothesized that maternal lipidomic profiles are related to prenatal DS and gestational age at birth among African American women. Women were enrolled at 9-25 weeks' gestation, completed questionnaires, and provided plasma samples. Lipidomic profiles were determined by "shotgun" Orbitrap high-resolution/accurate mass spectrometry. Data were analyzed using SIMCA P+ software. There was a clear separation in the orthogonal projections to latent structures discriminant analysis score plot between women with Center for Epidemiologic Studies Depression Scale (CES-D) scores ≥23 and women with CES-D scores ≤22. Similarly, a clear separation was observed in the model between PTB and full-term birth. Corresponding S-plot, loading plot, and variable importance in projection plot/list were used to identify the lipids responsible for the groupings. Higher levels of specific triglyceride (TG) species and lower levels of specific phosphatidylcholines (PCs) PC(37:1), PC(41:6), and PC(39:3) were associated with PTB. PC PC(37:1) levels were also lower among women with CES-D scores ≥23, pointing toward a possible connection between DS and PTB. Although overweight pregnant women showed higher levels of TGs, the PTB model showed specific TGs unique to PTB. Lipidomic profiles in pregnant African American women are related to DS, and our data suggest a role for specific TGs and PCs in PTB.
Collapse
Affiliation(s)
- Nadia Saadat
- College of Nursing, Wayne State University, Detroit, MI, USA
| | - Todd A Lydic
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Dawn P Misra
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Rhonda Dailey
- Department of Family Medicine and Public Health Sciences, School of Medicine, Wayne State University, Detroit, MI, USA
| | | | - Carmen Giurgescu
- College of Nursing, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
172
|
Vissers LET, Soedamah-Muthu SS, van der Schouw YT, Zuithoff NPA, Geleijnse JM, Sluijs I. Consumption of a diet high in dairy leads to higher 15:0 in cholesteryl esters of healthy people when compared to diets high in meat and grain. Nutr Metab Cardiovasc Dis 2020; 30:804-809. [PMID: 32139254 DOI: 10.1016/j.numecd.2020.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS A higher dairy product intake has been associated to higher blood concentrations of 15:0 (pentadecanoic acid), 17:0 (margaric acid), and 14:0 (myristic acid). This study investigates whether a diet high in dairy products influences cholesteryl ester fatty acid concentrations of these specific fatty acids (FA). METHODS AND RESULTS In a randomized multiple cross-over study, 13 men and 17 women aged 22 ± 4 years with a BMI of 21.6 ± 2.2 kg/m2 received 3 isocaloric intervention diets (dairy, meat or grain) in random order. For this post-hoc analysis, FA in plasma cholesteryl esters were measured using gas chromatography. We performed a linear mixed model per centered log-ratio transformed FA, adjusting for period, and the interaction between diet and period. Consumed total fat intake per controlled intervention diet was 31.0 ± 0.9 en%/day (dairy), 31.5 ± 0.6 en%/day (meat), and 28.4 ± 1.2 en%/day (grain), respectively. The dairy diet led to higher relative concentrations of 15:0 when compared to diets high in meat and grain, (β; 0.27, 95%CI: 0.18,0.37; p = 1.2 × 10-5, and β: 0.15; 95%CI: 0.06,0.24; p = 1.2 × 10-2, respectively). The dairy diet also led to higher 14:0 when compared to the meat diet (β: 0.34; 95%CI: 0.21,0.46; p = 6.0 × 10-5), but not when compared to the grain diet. 17:0 did not differ between diets. CONCLUSION The plasma cholesteryl ester fraction after a diet high in dairy was characterized by higher 15:0 levels. Concentrations of 14:0 were only higher when comparing the FA profile after a diet high in dairy when compared to a diet high in meat. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, NCT01314040.
Collapse
Affiliation(s)
- Linda E T Vissers
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Sabita S Soedamah-Muthu
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands; Center of Research on Psychological and Somatic Disorders (CORPS), Department of Medical and Clinical Psychology, Tilburg University, the Netherlands; Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Nicolaas P A Zuithoff
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Johanna M Geleijnse
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Ivonne Sluijs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
173
|
Fretts AM, Jensen PN, Hoofnagle A, McKnight B, Howard BV, Umans J, Yu C, Sitlani C, Siscovick DS, King IB, Sotoodehnia N, Lemaitre RN. Plasma Ceramide Species Are Associated with Diabetes Risk in Participants of the Strong Heart Study. J Nutr 2020; 150:1214-1222. [PMID: 31665380 PMCID: PMC7198314 DOI: 10.1093/jn/nxz259] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/09/2019] [Accepted: 09/27/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Few studies have assessed the associations of ceramides and sphingomyelins (SMs) with diabetes in humans. OBJECTIVE We assessed associations of 15 circulating ceramides and SM species with incident diabetes in 2 studies. METHODS The analysis included 435 American-Indian participants from the Strong Heart Study (nested case-control design for analyses; mean age: 57 y; 34% male; median time until diabetes 4.3 y for cases) and 1902 participants from the Strong Heart Family Study (prospective design for analyses; mean age: 37 y; 39% male; median 12.5 y of follow-up). Sphingolipid species were measured using stored plasma samples by sequential LC and MS. Using logistic regression and parametric survival models within studies, and an inverse-variance-weighted meta-analysis across studies, we examined associations of 15 ceramides and SM species with incident diabetes. RESULTS There were 446 cases of incident diabetes across the studies. Higher circulating concentrations of ceramides containing stearic acid (Cer-18), arachidic acid (Cer-20), and behenic acid (Cer-22) were each associated with a higher risk of diabetes. The RRs for incident diabetes per 1 SD of each log ceramide species (μM) were 1.22 (95% CI: 1.09, 1.37) for Cer-18, 1.18 (95% CI: 1.06, 1.31) for Cer-20, and 1.20 (95% CI: 1.08, 1.32) for Cer-22. Although the magnitude of the risk estimates for the association of ceramides containing lignoceric acid (Cer-24) with diabetes was similar to those for Cer-18, Cer-20, and Cer-22 (RR = 1.13; 95% CI: 1.01, 1.26), the association was not statistically significant after correction for multiple testing (P = 0.007). Ceramides carrying palmitic acid (Cer-16), SMs, glucosyl-ceramides, or a lactosyl-ceramide were not associated with diabetes risk. CONCLUSIONS Higher concentrations of circulating Cer-18, Cer-20, and Cer-22 were associated with a higher risk of developing diabetes in 2 studies of American-Indian adults. This trial was registered at clinicaltrials.gov as NCT00005134.
Collapse
Affiliation(s)
- Amanda M Fretts
- Department of Epidemiology, University of Washington, Seattle, WA, USA,Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Address correspondence to AMF (e-mail: )
| | - Paul N Jensen
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew Hoofnagle
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Barbara McKnight
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Barbara V Howard
- MedStar Health Research Institute, Hyattsville, MD, USA,Georgetown and Howard Universities Center for Translational Science, Washington, DC, USA
| | - Jason Umans
- MedStar Health Research Institute, Hyattsville, MD, USA
| | - Chaoyu Yu
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Colleen Sitlani
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Irena B King
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
174
|
Development of behenic acid-ethyl cellulose oleogel stabilized Pickering emulsions as low calorie fat replacer. Int J Biol Macromol 2020; 150:974-981. [DOI: 10.1016/j.ijbiomac.2019.10.205] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/21/2019] [Accepted: 10/24/2019] [Indexed: 01/08/2023]
|
175
|
Noerman S, Kolehmainen M, Hanhineva K. Profiling of Endogenous and Gut Microbial Metabolites to Indicate Metabotype-Specific Dietary Responses: A Systematic Review. Adv Nutr 2020; 11:1237-1254. [PMID: 32271864 PMCID: PMC7490160 DOI: 10.1093/advances/nmaa031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/21/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
Upon dietary exposure, the endogenous metabolism responds to the diet-derived nutrients and bioactive compounds, such as phytochemicals. However, the responses vary remarkably due to the interplay with other dietary components, lifestyle exposures, and intrinsic factors, which lead to differences in endogenous regulatory metabolism. These physiological processes are evidenced as a signature profile composed of various metabolites constituting metabolic phenotypes, or metabotypes. The metabolic profiling of biological samples following dietary intake hence would provide information about diet-that is, as the intake biomarkers and the ongoing physiological reactions triggered by this intake-thereby enable evaluation of the metabolic basis required to distinguish the different metabotypes. The capacity of nontargeted metabolomics to also encompass the unprecedented metabolite species has enabled the profiling of multiple metabolites and the corresponding metabotypes with a single analysis, decoding the complex interplay between diet, other relevant factors, and health. In this systematic review, we screened 345 articles published in English in January 2007-July 2018, which applied the metabolomics approach to profile the changes of endogenous metabolites in the blood related to dietary interventions, either derived by metabolism of gut microbiota or the human host. We excluded all the compounds that were directly derived from diet, and also the dietary interventions focusing on supplementation with individual compounds. After the removal of less relevant studies and assessment of eligibility, 49 articles were included in this review. First, we mention the contribution of individual factors, either modifiable or nonmodifiable factors, in shaping metabolic profile. Then, how different aspects of the diet would affect the metabolic profiles are disentangled. Next, the classes of endogenous metabolites altered following included dietary interventions are listed. We also discuss the current challenges in the field, along with future research opportunities.
Collapse
Affiliation(s)
- Stefania Noerman
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland,Address correspondence to SN (e-mail: )
| | - Marjukka Kolehmainen
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Kati Hanhineva
- Department of Clinical Nutrition, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland,Address correspondence to KH ()
| |
Collapse
|
176
|
Venn-Watson S, Baird M, Novick B, Parry C, Jensen ED. Modified fish diet shifted serum metabolome and alleviated chronic anemia in bottlenose dolphins (Tursiops truncatus): Potential role of odd-chain saturated fatty acids. PLoS One 2020; 15:e0230769. [PMID: 32259832 PMCID: PMC7138614 DOI: 10.1371/journal.pone.0230769] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/06/2020] [Indexed: 11/18/2022] Open
Abstract
Bottlenose dolphins (Tursiops truncatus) are long-lived mammals that can develop chronic aging-associated conditions similar to humans, including metabolic syndrome. Initial studies suggest that these conditions may be attenuated in dolphins using a modified fish diet. Serum metabolomics, fatty acid panels, and blood-based health indices were compared between 20 dolphins on a modified, 50% wild-type diet (50% mullet, 25% capelin, and 25% squid and/or herring) and 10 dolphins on a baseline diet (75% capelin and 25% squid and/or herring). Blood samples were collected at Months 0, 1, 3 and 6. Dolphins on the modified diet had lower insulin (7.5 ± 4.0 and 14.8 ± 14.0 μIU/ml, P = 0.039), lower cholesterol (160 ± 26 and 186 ± 24 mg/dl, P = 0.015) and higher hematocrit (46 ± 3 and 44 ± 3%, P = 0.043) by Month 1 compared to controls. Dolphins with anemia (hemoglobin ≤ 12.5 g/dl, n = 6) or low-normal hemoglobin (12.5-13.5 g/dl, n = 3) before placed on the modified diet had normal hemoglobin concentrations (> 13.5 g/dl) by Month 3. The modified diet caused a significant shift in the metabolome, which included 664 known metabolites. Thirty prioritized metabolites at Months 1 and 3 were 100% predictive of dolphins on the modified diet. Among 25 prioritized lipids, 10 (40%) contained odd-chain saturated fatty acids (OCFAs); C15:0 was the highest-prioritized OCFA. Increased dietary intake of C15:0 (from 1.3 ± 0.4 to 4.5 ± 1.1 g/day) resulted in increased erythrocyte C15:0 concentrations (from 1.5 ± 0.3 to 5.8 ± 0.8 μg/ml, P < 0.0001), which independently predicted raised hemoglobin. Further, increasing age was associated with declining serum C15:0 (R2 = 0.14, P = 0.04). While higher circulating OCFAs have been previously associated with lower risks of cardiometabolic diseases in humans, further studies are warranted to assess potential active roles of OCFAs, including C15:0, in attenuating anemia.
Collapse
Affiliation(s)
- Stephanie Venn-Watson
- Translational Medicine and Research Program, National Marine Mammal Foundation, San Diego, California, United States of America
- * E-mail:
| | - Mark Baird
- Translational Medicine and Research Program, National Marine Mammal Foundation, San Diego, California, United States of America
| | - Brittany Novick
- Translational Medicine and Research Program, National Marine Mammal Foundation, San Diego, California, United States of America
| | - Celeste Parry
- Translational Medicine and Research Program, National Marine Mammal Foundation, San Diego, California, United States of America
| | - Eric D. Jensen
- U.S. Navy Marine Mammal Program, Naval Information Warfare Center Pacific, San Diego, California, United States of America
| |
Collapse
|
177
|
Impact of management system and lactation stage on fatty acid composition of camel milk. J Food Compost Anal 2020. [DOI: 10.1016/j.jfca.2020.103418] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
178
|
Sun Y, Gao HY, Fan ZY, He Y, Yan YX. Metabolomics Signatures in Type 2 Diabetes: A Systematic Review and Integrative Analysis. J Clin Endocrinol Metab 2020; 105:5645632. [PMID: 31782507 DOI: 10.1210/clinem/dgz240] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/28/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Metabolic signatures have emerged as valuable signaling molecules in the biochemical process of type 2 diabetes (T2D). To summarize and identify metabolic biomarkers in T2D, we performed a systematic review and meta-analysis of the associations between metabolites and T2D using high-throughput metabolomics techniques. METHODS We searched relevant studies from MEDLINE (PubMed), Embase, Web of Science, and Cochrane Library as well as Chinese databases (Wanfang, Vip, and CNKI) inception through 31 December 2018. Meta-analysis was conducted using STATA 14.0 under random effect. Besides, bioinformatic analysis was performed to explore molecule mechanism by MetaboAnalyst and R 3.5.2. RESULTS Finally, 46 articles were included in this review on metabolites involved amino acids, acylcarnitines, lipids, carbohydrates, organic acids, and others. Results of meta-analysis in prospective studies indicated that isoleucine, leucine, valine, tyrosine, phenylalanine, glutamate, alanine, valerylcarnitine (C5), palmitoylcarnitine (C16), palmitic acid, and linoleic acid were associated with higher T2D risk. Conversely, serine, glutamine, and lysophosphatidylcholine C18:2 decreased risk of T2D. Arginine and glycine increased risk of T2D in the Western countries subgroup, and betaine was negatively correlated with T2D in nested case-control subgroup. In addition, slight improvements in T2D prediction beyond traditional risk factors were observed when adding these metabolites in predictive analysis. Pathway analysis identified 17 metabolic pathways may alter in the process of T2D and metabolite-related genes were also enriched in functions and pathways associated with T2D. CONCLUSIONS Several metabolites and metabolic pathways associated with T2D have been identified, which provide valuable biomarkers and novel targets for prevention and drug therapy.
Collapse
Affiliation(s)
- Yue Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Hao-Yu Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Zhi-Yuan Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yan He
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
- Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
179
|
Valença RDL, Silva Sobrinho AGD, Romanzini EP, Andrade ND, Borghi TH, Zeola NMBL, Cirne LGA, Oliveira VDS. Peanut meal and crude glycerin in lamb diets: Meat quality and fatty acid profile. Small Rumin Res 2020. [DOI: 10.1016/j.smallrumres.2020.106076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
180
|
LC-HRMS based approach to identify novel sphingolipid biomarkers in breast cancer patients. Sci Rep 2020; 10:4668. [PMID: 32170160 PMCID: PMC7070000 DOI: 10.1038/s41598-020-61283-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/20/2020] [Indexed: 01/11/2023] Open
Abstract
Perturbations in lipid metabolic pathways to meet the bioenergetic and biosynthetic requirements is a principal characteristic of cancer cells. Sphingolipids (SPLs) are the largest class of bioactive lipids associated to various aspects of tumorigenesis and have been extensively studied in cancer cell lines and experimental models. The clinical relevance of SPLs in human malignancies however is still poorly understood and needs further investigation. In the present study, we adopted a UHPLC-High resolution (orbitrap) Mass spectrometry (HRMS) approach to identify various sphingolipid species in breast cancer patients. A total of 49 SPLs falling into 6 subcategories have been identified. Further, integrating the multivariate analysis with metabolomics enabled us to identify an elevation in the levels of ceramide phosphates and sphingosine phosphates in tumor tissues as compared to adjacent normal tissues. The expression of genes involved in the synthesis of reported metabolites was also determined in local as well as TCGA cohort. A significant upregulation in the expression of CERK and SPHK1 was observed in tumor tissues in local and TCGA cohort. Sphingomyelin levels were found to be high in adjacent normal tissues. Consistent with the above findings, expression of SGMS1 in tumor tissues was downregulated in TCGA cohort only. Clinical correlations of the selected metabolites and their performance as biomarkers was also evaluated. Significant ROC and positive correlation with Ki67 index highlight the diagnostic potential and clinical relevance of ceramide phosphates in breast cancer.
Collapse
|
181
|
Borges MC, Schmidt AF, Jefferis B, Wannamethee SG, Lawlor DA, Kivimaki M, Kumari M, Gaunt TR, Ben-Shlomo Y, Tillin T, Menon U, Providencia R, Dale C, Gentry-Maharaj A, Hughes A, Chaturvedi N, Casas JP, Hingorani AD. Circulating Fatty Acids and Risk of Coronary Heart Disease and Stroke: Individual Participant Data Meta-Analysis in Up to 16 126 Participants. J Am Heart Assoc 2020; 9:e013131. [PMID: 32114887 PMCID: PMC7335585 DOI: 10.1161/jaha.119.013131] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background We aimed at investigating the association of circulating fatty acids with coronary heart disease (CHD) and stroke risk. Methods and Results We conducted an individual-participant data meta-analysis of 5 UK-based cohorts and 1 matched case-control study. Fatty acids (ie, omega-3 docosahexaenoic acid, omega-6 linoleic acid, monounsaturated and saturated fatty acids) were measured at baseline using an automated high-throughput serum nuclear magnetic resonance metabolomics platform. Data from 3022 incident CHD cases (13 104 controls) and 1606 incident stroke cases (13 369 controls) were included. Logistic regression was used to model the relation between fatty acids and odds of CHD and stroke, adjusting for demographic and lifestyle variables only (ie, minimally adjusted model) or with further adjustment for other fatty acids (ie, fully adjusted model). Although circulating docosahexaenoic acid, but not linoleic acid, was related to lower CHD risk in the fully adjusted model (odds ratio, 0.85; 95% CI, 0.76-0.95 per standard unit of docosahexaenoic acid), there was evidence of high between-study heterogeneity and effect modification by study design. Stroke risk was consistently lower with increasing circulating linoleic acid (odds ratio for fully adjusted model, 0.82; 95% CI, 0.75-0.90). Circulating monounsaturated fatty acids were associated with higher CHD risk across all models and with stroke risk in the fully adjusted model (odds ratio, 1.22; 95% CI, 1.03-1.44). Saturated fatty acids were not related to increased CHD risk in the fully adjusted model (odds ratio, 0.94; 95% CI, 0.82-1.09), or stroke risk. Conclusions We found consistent evidence that linoleic acid was associated with decreased risk of stroke and that monounsaturated fatty acids were associated with increased risk of CHD. The different pattern between CHD and stroke in terms of fatty acids risk profile suggests future studies should be cautious about using composite events. Different study designs are needed to assess which, if any, of the associations observed is causal.
Collapse
Affiliation(s)
- Maria Carolina Borges
- MRC Integrative Epidemiology Unit at the University of Bristol United Kingdom.,Population Health Sciences Bristol Medical School University of Bristol United Kingdom
| | - Amand Floriaan Schmidt
- Institute of Cardiovascular Science University College London London United Kingdom.,Groningen Research Institute of Pharmacy University of Groningen the Netherlands.,Division Heart and Lungs Department of Cardiology University Medical Center Utrecht Utrecht The Netherlands
| | - Barbara Jefferis
- UCL Department of Primary Care & Population Health UCL Medical School London United Kingdom
| | - S Goya Wannamethee
- UCL Department of Primary Care & Population Health UCL Medical School London United Kingdom
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol United Kingdom.,Population Health Sciences Bristol Medical School University of Bristol United Kingdom
| | - Mika Kivimaki
- Department of Epidemiology and Public Health University College London London United Kingdom
| | - Meena Kumari
- Department of Epidemiology and Public Health University College London London United Kingdom.,Institute for Social and Economic Research University of Essex United Kingdom
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit at the University of Bristol United Kingdom.,Population Health Sciences Bristol Medical School University of Bristol United Kingdom
| | - Yoav Ben-Shlomo
- Population Health Sciences Bristol Medical School University of Bristol United Kingdom
| | - Therese Tillin
- Cardiometabolic Phenotyping Group Institute of Cardiovascular Science University College London London United Kingdom
| | - Usha Menon
- MRC Clinical Trials Unit at UCL Institute of Clinical Trials & MethodologyUniversity College London London United Kingdom
| | - Rui Providencia
- Farr Institute of Health Informatics University College London London United Kingdom.,Barts Heart Centre St Bartholomew's Hospital Barts Health NHS Trust London United Kingdom
| | - Caroline Dale
- Farr Institute of Health Informatics University College London London United Kingdom
| | - Aleksandra Gentry-Maharaj
- MRC Clinical Trials Unit at UCL Institute of Clinical Trials & MethodologyUniversity College London London United Kingdom
| | - Alun Hughes
- Institute of Cardiovascular Science University College London London United Kingdom
| | - Nish Chaturvedi
- Institute of Cardiovascular Science University College London London United Kingdom
| | - Juan Pablo Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC) VA Boston Healthcare System Boston MA USA
| | - Aroon D Hingorani
- Institute of Cardiovascular Science University College London London United Kingdom.,Farr Institute of Health Informatics University College London London United Kingdom
| | | |
Collapse
|
182
|
Beyene HB, Hamley S, Giles C, Huynh K, Smith A, Cinel M, Mellet NA, Morales-Scholz MG, Kloosterman D, Howlett KF, Kowalski GM, Shaw CS, Magliano DJ, Bruce CR, Meikle PJ. Mapping the Associations of the Plasma Lipidome With Insulin Resistance and Response to an Oral Glucose Tolerance Test. J Clin Endocrinol Metab 2020; 105:5722002. [PMID: 32016362 DOI: 10.1210/clinem/dgaa054] [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: 11/14/2019] [Accepted: 02/02/2020] [Indexed: 02/13/2023]
Abstract
CONTEXT Insulin resistance (IR) remains a global health challenge. Lipidomics offers an opportunity to identify biomarkers and better understand mechanisms of IR associated with abnormal lipid metabolism. OBJECTIVE The objective of this article is to determine plasma lipid species associated with indices of IR and evaluate the lipidome response to an oral glucose tolerance test (OGTT). DESIGN AND SETTING This study was community based and cross-sectional. PARTICIPANTS AND SAMPLE Plasma samples (collected at 0 and 120 min during an OGTT) from nonobese, young adults age 18 to 34 years (n = 246) were analyzed using liquid chromatography-tandem mass spectrometry. MAIN OUTCOME MEASURES The associations between indices of IR and lipid classes and species (with a sex interaction term), or changes in lipid levels during an OGTT, were tested using linear models (adjusted for age, sex, body mass index, total cholesterol, high-density lipoprotein cholesterol, and triglycerides). RESULTS Some (213) and (199) lipid species were associated with the homeostatic model assessment of insulin resistance and insulin area under curve (AUC), respectively. Alkylphosphatidylcholine (10), alkenylphosphatidylcholine (23), and alkylphosphatidylethanolamine (6) species were associated with insulin AUC in men only. Species of phosphatidylcholine (7) and sphingomyelin (5) were associated in women only. In response to an OGTT, a perturbation in the plasma lipidome, particularly in acylcarnitine species, was observed; and the changes in many lipid species were associated with insulin AUC. CONCLUSIONS The plasma lipidome and changes in lipid levels during an OGTT were associated with indices of IR. These findings underlie the involvement of molecular lipid species in the pathogenesis of IR and possibly crosstalk between IR and sex-specific regulation of lipid metabolism.
Collapse
Affiliation(s)
- Habtamu B Beyene
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Steven Hamley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Melbourne, Victoria, Australia
| | - Corey Giles
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Alexander Smith
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Michelle Cinel
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Natalie A Mellet
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Maria G Morales-Scholz
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Melbourne, Victoria, Australia
| | - Danielle Kloosterman
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Melbourne, Victoria, Australia
| | - Kirsten F Howlett
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Melbourne, Victoria, Australia
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Melbourne, Victoria, Australia
| | - Christopher S Shaw
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Melbourne, Victoria, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Clinton R Bruce
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
183
|
Ardisson Korat AV, Malik VS, Furtado JD, Sacks F, Rosner B, Rexrode KM, Willett WC, Mozaffarian D, Hu FB, Sun Q. Circulating Very-Long-Chain SFA Concentrations Are Inversely Associated with Incident Type 2 Diabetes in US Men and Women. J Nutr 2020; 150:340-349. [PMID: 31618417 PMCID: PMC7308624 DOI: 10.1093/jn/nxz240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/30/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Very-long-chain SFAs (VLCSFAs), such as arachidic acid (20:0), behenic acid (22:0), and lignoceric acid (24:0), have demonstrated inverse associations with cardiometabolic conditions, although more evidence is needed to characterize their relation with risk of type 2 diabetes (T2D). In addition, little is known regarding their potential dietary and lifestyle predictors. OBJECTIVE We aimed to examine the association of plasma and erythrocyte concentrations of VLCSFAs with incident T2D risk. METHODS We used existing measurements of fatty acid concentrations in plasma and erythrocytes among 2854 and 2831 participants in the Nurses' Health Study (NHS) and Health Professionals Follow-Up Study (HPFS), respectively. VLCSFAs were measured using GLC, and individual fatty acid concentrations were expressed as a percentage of total fatty acids. Incident T2D cases were identified by self-reports and confirmed by a validated supplementary questionnaire. Cox proportional hazards regression was used to evaluate the association between VLCSFAs and T2D, adjusting for demographic, lifestyle, and dietary variables. RESULTS During 39,941 person-years of follow-up, we documented 243 cases of T2D. Intakes of peanuts, peanut butter, vegetable fat, dairy fat, and palmitic/stearic (16:0-18:0) fatty acids were significantly, albeit weakly, correlated with plasma and erythrocyte VLCSFA concentrations (|rs| ≤ 0.19). Comparing the highest with the lowest quartiles of plasma concentrations, pooled HRs (95% CIs) were 0.51 (0.35, 0.75) for arachidic acid, 0.43 (0.28, 0.64) for behenic acid, 0.40 (0.27, 0.61) for lignoceric acid, and 0.41 (0.27, 0.61) for the sum of VLCSFAs, after multivariate adjustments for demographic, lifestyle, and dietary factors. For erythrocyte VLCSFAs, only arachidic acid and behenic acid concentrations were inversely associated with T2D risk. CONCLUSIONS Our findings suggest that, in US men and women, higher plasma concentrations of VLCSFAs are associated with lower risk of T2D. More research is needed to understand the mechanistic pathways underlying these associations.
Collapse
Affiliation(s)
- Andres V Ardisson Korat
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Vasanti S Malik
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Jeremy D Furtado
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Frank Sacks
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Genetics and Complex Diseases, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Bernard Rosner
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathryn M Rexrode
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dariush Mozaffarian
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Sun
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
184
|
Abstract
PURPOSE OF REVIEW Due to inconsistent findings on dairy consumption and CVD and all-cause mortality, we performed a narrative literature review to the current literature on dairy and its association with CVD. RECENT FINDINGS Due to their complex biochemistry, dairy consumption is a rather heterogeneous exposure. Multiple pathways have been proposed from dairy consumption and CVD. Current guidelines advocate for consumption of low-fat dairy products particularly milk, cheese, and yogurt, although the evidence for this is scant. Randomized clinical trials and large prospective studies on lipid-related cardiometabolic disease risk factors are consistent with results from most meta-analyses of prospective cohort studies, which suggest null or inverse relationship between CVD risk and mortality with dairy consumption although there is no clear dose response relationship. Most of current evidence suggests that dairy products are neutral or positive effect on human cardiovascular diseases.
Collapse
|
185
|
Park YK, Ledesma-Amaro R, Nicaud JM. De novo Biosynthesis of Odd-Chain Fatty Acids in Yarrowia lipolytica Enabled by Modular Pathway Engineering. Front Bioeng Biotechnol 2020; 7:484. [PMID: 32039184 PMCID: PMC6987463 DOI: 10.3389/fbioe.2019.00484] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 12/27/2019] [Indexed: 01/21/2023] Open
Abstract
Microbial oils are regarded as promising alternatives to fossil fuels as concerns over environmental issues and energy production systems continue to mount. Odd-chain fatty acids (FAs) are a type of valuable lipid with various applications: they can serve as biomarkers, intermediates in the production of flavor and fragrance compounds, fuels, and plasticizers. Microorganisms naturally produce FAs, but such FAs are primarily even-chain; only negligible amounts of odd-chain FAs are generated. As a result, studies using microorganisms to produce odd-chain FAs have had limited success. Here, our objective was to biosynthesize odd-chain FAs de novo in Yarrowia lipolytica using inexpensive carbon sources, namely glucose, without any propionate supplementation. To achieve this goal, we constructed a modular metabolic pathway containing seven genes. In the engineered strain expressing this pathway, the percentage of odd-chain FAs out of total FAs was higher than in the control strain (3.86 vs. 0.84%). When this pathway was transferred into an obese strain, which had been engineered to accumulate large amounts of lipids, odd-chain fatty acid production was 7.2 times greater than in the control (0.05 vs. 0.36 g/L). This study shows that metabolic engineering research is making progress toward obtaining efficient cell factories that produce odd-chain FAs.
Collapse
Affiliation(s)
- Young-Kyoung Park
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Rodrigo Ledesma-Amaro
- Imperial College Centre for Synthetic Biology and Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Jean-Marc Nicaud
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
186
|
Metabolomic and Lipidomic Signatures of Metabolic Syndrome and its Physiological Components in Adults: A Systematic Review. Sci Rep 2020; 10:669. [PMID: 31959772 PMCID: PMC6971076 DOI: 10.1038/s41598-019-56909-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
The aim of this work was to conduct a systematic review of human studies on metabolite/lipid biomarkers of metabolic syndrome (MetS) and its components, and provide recommendations for future studies. The search was performed in MEDLINE, EMBASE, EMB Review, CINHAL Complete, PubMed, and on grey literature, for population studies identifying MetS biomarkers from metabolomics/lipidomics. Extracted data included population, design, number of subjects, sex/gender, clinical characteristics and main outcome. Data were collected regarding biological samples, analytical methods, and statistics. Metabolites were compiled by biochemical families including listings of their significant modulations. Finally, results from the different studies were compared. The search yielded 31 eligible studies (2005–2019). A first category of articles identified prevalent and incident MetS biomarkers using mainly targeted metabolomics. Even though the population characteristics were quite homogeneous, results were difficult to compare in terms of modulated metabolites because of the lack of methodological standardization. A second category, focusing on MetS components, allowed comparing more than 300 metabolites, mainly associated with the glycemic component. Finally, this review included also publications studying type 2 diabetes as a whole set of metabolic risks, raising the interest of reporting metabolomics/lipidomics signatures to reflect the metabolic phenotypic spectrum in systems approaches.
Collapse
|
187
|
Abstract
The effect of dietary fats on cardiometabolic diseases, including cardiovascular diseases and type 2 diabetes mellitus, has generated tremendous interest. Many earlier investigations focused on total fat and conventional fat classes (such as saturated and unsaturated fats) and their influence on a limited number of risk factors. However, dietary fats comprise heterogeneous molecules with diverse structures, and growing research in the past two decades supports correspondingly complex health effects of individual dietary fats. Moreover, health effects of dietary fats might be modified by additional factors, such as accompanying nutrients and food-processing methods, emphasizing the importance of the food sources. Accordingly, the rapidly increasing scientific findings on dietary fats and cardiometabolic diseases have generated debate among scientists, caused confusion for the general public and present challenges for translation into dietary advice and policies. This Review summarizes the evidence on the effects of different dietary fats and their food sources on cell function and on risk factors and clinical events of cardiometabolic diseases. The aim is not to provide an exhaustive review but rather to focus on the most important evidence from randomized controlled trials and prospective cohort studies and to highlight current areas of controversy and the most relevant future research directions for understanding how to improve the prevention and management of cardiometabolic diseases through optimization of dietary fat intake.
Collapse
|
188
|
Li X, Huang Y, Zhang W, Yang C, Su W, Wu Y, Chen X, Zhou A, Huo X, Xia W, Xu S, Chen D, Li Y. Association of circulating saturated fatty acids with the risk of pregnancy-induced hypertension: a nested case-control study. Hypertens Res 2020; 43:412-421. [PMID: 31919480 DOI: 10.1038/s41440-019-0383-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/12/2019] [Accepted: 12/03/2019] [Indexed: 11/09/2022]
Abstract
Circulating saturated fatty acids (SFAs) have been associated with cardiovascular disease. However, little is known about the relationship of SFAs with the risk of pregnancy-induced hypertension (PIH). We conducted a nested case-control study to examine the associations between circulating SFAs and the risk of PIH. A total of 92 PIH cases were matched to 184 controls by age (±2 years) and infant sex from a birth cohort study conducted in Wuhan, China. Levels of circulating fatty acids in plasma were measured using gas chromatography-mass spectrometry. Conditional logistic regressions were conducted to calculate odds ratios (ORs) and 95% confidence intervals (95% CIs). Even-chain SFAs, including myristic acid (14:0) and palmitic acid (16:0), were positively associated with the risk of PIH [ORs (95% CIs): 2.92 (1.27, 6.74) for 14:0 and 2.85 (1.18, 6.89) for 16:0, % by wt]. In contrast, higher levels of very-long-chain SFAs, including arachidic acid (20:0), behenic acid (22:0), and lignoceric acid (24:0), were associated with a lower risk of PIH [ORs (95% CIs): 0.40 (0.17, 0.92) for 20:0, 0.30 (0.12, 0.71) for 22:0 and 0.26 (0.11, 0.64) for 24:0, μg/mL]. For odd-chain SFAs, including pentadecanoic acid (15:0) and heptadecanoic acid (17:0), no significant difference was observed. Our results provided convincing evidence that different subclasses of SFAs showed diverse effects on the risk of PIH. This suggests that dietary very-long-chain SFAs may be a novel means by which to prevent hypertension. Future studies are required to confirm these associations and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Xinping Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yichao Huang
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, PR China
| | - Wenxin Zhang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chenhui Yang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Weijie Su
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, PR China
| | - Yi Wu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiaomei Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Aifen Zhou
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan, Hubei, PR China
| | - Xia Huo
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, PR China
| | - Wei Xia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Shunqing Xu
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Da Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, PR China.
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
189
|
Soedamah-Muthu SS, Guo J. Dairy consumption and cardiometabolic diseases: Evidence from prospective studies. MILK AND DAIRY FOODS 2020:1-28. [DOI: 10.1016/b978-0-12-815603-2.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
190
|
Liu L, Xie B, Fan M, Candas-Green D, Jiang JX, Wei R, Wang Y, Chen HW, Hu Y, Li JJ. Low-Level Saturated Fatty Acid Palmitate Benefits Liver Cells by Boosting Mitochondrial Metabolism via CDK1-SIRT3-CPT2 Cascade. Dev Cell 2019; 52:196-209.e9. [PMID: 31866205 DOI: 10.1016/j.devcel.2019.11.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/10/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022]
Abstract
Saturated fatty acids (SFAs) (the "bad" fat), especially palmitate (PA), in the human diet are blamed for potential health risks such as obesity and cancer because of SFA-induced lipotoxicity. However, epidemiological results demonstrate a latent benefit of SFAs, and it remains elusive whether a certain low level of SFAs is physiologically essential for maintaining cell metabolic hemostasis. Here, we demonstrate that although high-level PA (HPA) indeed induces lipotoxic effects in liver cells, low-level PA (LPA) increases mitochondrial functions and alleviates the injuries induced by HPA or hepatoxic agent carbon tetrachloride (CCl4). LPA treatment in mice enhanced liver mitochondrial activity and reduced CCl4 hepatotoxicity with improved blood levels of aspartate aminotransferase (AST), alanine transaminase (ALT), and mitochondrial aspartate transaminase (m-AST). LPA-mediated mitochondrial homeostasis is regulated by CDK1-mediated SIRT3 phosphorylation, which in turn deacetylates and dimerizes CPT2 to enhance fatty acid oxidation. Thus, an advantageous effect is suggested by the consumption of LPA that augments mitochondrial metabolic homeostasis via CDK1-SIRT3-CPT2 cascade.
Collapse
Affiliation(s)
- Lin Liu
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Institute of Liver Diseases, Shuguan Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bowen Xie
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Ming Fan
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Demet Candas-Green
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Joy X Jiang
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Ryan Wei
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Yinsheng Wang
- Department of Chemistry, University of California, Riverside, Riverside, CA, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA; Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA
| | - Yiyang Hu
- Institute of Liver Diseases, Shuguan Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Jian Li
- Department of Radiation Oncology, School of Medicine, University of California, Davis, Sacramento, CA, USA; Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, USA.
| |
Collapse
|
191
|
Lemaitre RN, McKnight B, Sotoodehnia N, Fretts AM, Qureshi WT, Song X, King IB, Sitlani CM, Siscovick DS, Psaty BM, Mozaffarian D. Circulating Very Long-Chain Saturated Fatty Acids and Heart Failure: The Cardiovascular Health Study. J Am Heart Assoc 2019; 7:e010019. [PMID: 30608197 PMCID: PMC6404213 DOI: 10.1161/jaha.118.010019] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Circulating very‐long‐chain saturated fatty acids (VLSFAs) are integrated biomarkers of diet and metabolism that may point to new risk pathways and potential targets for heart failure (HF) prevention. The associations of VLSFA to HF in humans are not known. Methods and Results Using a cohort study design, we studied the associations of serially measured plasma phospholipid VLSFA with incident HF in the Cardiovascular Health Study. We investigated the associations of time‐varying levels of the 3 major circulating VLSFAs, lignoceric acid (24:0), behenic acid (22:0), and arachidic acid (20:0), with the risk of incident HF using Cox regression. During 45030 person‐years among 4249 participants, we identified 1304 cases of incident HF, including 489 with preserved and 310 with reduced ejection fraction. Adjusting for major HF risk factors and other circulating fatty acids, higher levels of each VLSFAs were associated with lower risk of incident HF (P trend≤0.0007 each). The hazard ratio comparing the highest quintile to the lowest quintile was 0.67 (95% confidence interval, 0.55–0.81) for 24:0, 0.72 (95% confidence interval, 0.60–0.87) for 22:0 and 0.72 (95% confidence interval, 0.59–0.88) for 20:0. The associations were similar in subgroups defined by sex, age, body mass index, coronary heart disease, and diabetes mellitus. Among those with ejection fraction data, the associations appeared similar for those with preserved and with reduced ejection fraction. Conclusions Higher levels of circulating VLSFAs are associated with lower risk of incident HF in older adults. These novel associations should prompt further research on the role of VLSFA in HF, including relevant new risk pathways. Clinical Trial Registration URL: https://www.clinicaltrials.gov. Unique identifier: NCT00005133.
Collapse
Affiliation(s)
- Rozenn N Lemaitre
- 1 Department of Medicine Cardiovascular Health Research Unit University of Washington Seattle WA
| | - Barbara McKnight
- 2 Department of Biostatistics University of Washington Seattle WA
| | - Nona Sotoodehnia
- 1 Department of Medicine Cardiovascular Health Research Unit University of Washington Seattle WA
| | - Amanda M Fretts
- 3 Department of Epidemiology University of Washington Seattle WA
| | - Waqas T Qureshi
- 4 School of Medicine Wake Forest University Winston Salem NC
| | - Xiaoling Song
- 5 Public Health Sciences Division Fred Hutchinson Cancer Research Center Seattle WA
| | - Irena B King
- 6 Department of Internal Medicine University of New Mexico Albuquerque NM
| | - Colleen M Sitlani
- 1 Department of Medicine Cardiovascular Health Research Unit University of Washington Seattle WA
| | - David S Siscovick
- 7 Institute for Urban Health New York Academy of Medicine New York NY
| | - Bruce M Psaty
- 1 Department of Medicine Cardiovascular Health Research Unit University of Washington Seattle WA.,3 Department of Epidemiology University of Washington Seattle WA.,8 Department of Health Services University of Washington Seattle WA.,9 Kaiser Permanente Washington Health Research Institute Seattle WA
| | | |
Collapse
|
192
|
Increasing the proportion of plasma MUFA, as a result of dietary intervention, is associated with a modest improvement in insulin sensitivity. J Nutr Sci 2019; 9:e6. [PMID: 32076549 PMCID: PMC7003243 DOI: 10.1017/jns.2019.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The effect of modifying dietary fatty acid (FA) composition on insulin sensitivity remains unclear. We aimed to investigate whether changes in plasma phospholipid (PL) FA composition, as a result of dietary intervention, correspond with changes in insulin sensitivity. The RISCK study was a 6-month randomised controlled dietary intervention study, which assessed the effect of modifying dietary fat and the glycaemic index (GI) of carbohydrates on insulin sensitivity. Total NEFA levels, fasting plasma PL FA profiles and an insulin sensitivity index (Si), derived from intravenous glucose tolerance minimal-model analysis, were available from 533 participants, all at elevated risk of type 2 diabetes. Bivariate correlations between changes in saturated PL FA (SFA), MUFA (as a percentage of total plasma NEFA) and changes in Si were assessed according to treatment group. Age, sex, ethnicity, percentage change in body mass and change in dietary GI were controlled for. Increasing total NEFA concentration was associated with worsening Si (r -0·152; P = 0·001). In the high-MUFA/low-GI diet group, change in PL-MUFA was positively and independently associated with change in Si (r 0·297; P = 0·002). Among MUFA, change in oleic acid (18 : 1) was most strongly correlated with change in Si (r 0·266; P = 0·005), as was change in minor FA 24 : 1 (r 0·244; P = 0·011) and 17 : 1 (r 0·196; P = 0·042). In the high-SFA/high-GI group, change in SFA concentration was not significantly associated with change in Si. In conclusion, increases in the proportion of plasma PL-MUFA following a high-MUFA dietary intervention were associated with improvements in insulin sensitivity.
Collapse
Key Words
- %NEFAtotal, percentage of total NEFA
- FA, fatty acid
- Fatty acids
- GI, glycaemic index
- HGI, high glycaemic index
- HM, high MUFA
- HS, high-SFA
- Insulin resistance
- Insulin sensitivity
- LGI, low glycaemic index
- MUFA
- PL, phospholipid
- RISCK, Reading, Imperial, Surrey, Cambridge and Kings
- Si, insulin sensitivity index
- T2DM, type 2 diabetes mellitus
- Type 2 diabetes mellitus
Collapse
|
193
|
Pertiwi K, Küpers LK, Wanders AJ, de Goede J, Zock PL, Geleijnse JM. Associations of dairy and fiber intake with circulating odd-chain fatty acids in post-myocardial infarction patients. Nutr Metab (Lond) 2019; 16:78. [PMID: 31754368 PMCID: PMC6854617 DOI: 10.1186/s12986-019-0407-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
Background Circulating odd-chain fatty acids pentadecanoic (15:0) and heptadecanoic acid (17:0) are considered to reflect dairy intake. In cohort studies, higher circulating 15:0 and 17:0 were associated with lower type 2 diabetes risk. A recent randomized controlled trial in humans suggested that fiber intake also increased circulating 15:0 and 17:0, potentially resulting from fermentation by gut microbes. We examined the associations of dairy and fiber intake with circulating 15:0 and 17:0 in patients with a history of myocardial infarction (MI). Methods We performed cross-sectional analyses in a subsample of 869 Dutch post-MI patients of the Alpha Omega Cohort who had data on dietary intake and circulating fatty acids. Dietary intakes (g/d) were assessed using a 203-item food frequency questionnaire. Circulating 15:0 and 17:0 (as % of total fatty acids) were measured in plasma phospholipids (PL) and cholesteryl esters (CE). Spearman correlations (rs) were computed between intakes of total dairy, dairy fat, fiber, and circulating 15:0 and 17:0. Results Patients were on average 69 years old, 78% was male and 21% had diabetes. Total dairy intake comprised predominantly milk and yogurt (69%). Dairy fat was mainly derived from cheese (47%) and milk (15%), and fiber was mainly from grains (43%). Circulating 15:0 in PL was significantly correlated with total dairy and dairy fat intake (both rs = 0.19, p < 0.001), but not with dietary fiber intake (rs = 0.05, p = 0.11). Circulating 17:0 in PL was correlated both with dairy intake (rs = 0.14 for total dairy and 0.11 for dairy fat, p < 0.001), and fiber intake (rs = 0.19, p < 0.001). Results in CE were roughly similar, except for a weaker correlation of CE 17:0 with fiber (rs = 0.11, p = 0.001). Circulating 15:0 was highest in those with high dairy intake irrespective of fiber intake, while circulating 17:0 was highest in those with high dairy and fiber intake. Conclusions In our cohort of post-MI patients, circulating 15:0 was associated with dairy intake but not fiber intake, whereas circulating 17:0 was associated with both dairy and fiber intake. These data suggest that cardiometabolic health benefits previously attributed to 17:0 as a biomarker of dairy intake may partly be explained by fiber intake.
Collapse
Affiliation(s)
- Kamalita Pertiwi
- 1Division of Human Nutrition and Health, Wageningen University, PO Box 17, 6700 AA Wageningen, The Netherlands
| | - Leanne K Küpers
- 1Division of Human Nutrition and Health, Wageningen University, PO Box 17, 6700 AA Wageningen, The Netherlands
| | - Anne J Wanders
- 2Future Health & Wellness, Unilever R&D, Vlaardingen, The Netherlands
| | - Janette de Goede
- 1Division of Human Nutrition and Health, Wageningen University, PO Box 17, 6700 AA Wageningen, The Netherlands
| | - Peter L Zock
- 2Future Health & Wellness, Unilever R&D, Vlaardingen, The Netherlands
| | - Johanna M Geleijnse
- 1Division of Human Nutrition and Health, Wageningen University, PO Box 17, 6700 AA Wageningen, The Netherlands
| |
Collapse
|
194
|
Lai HTM, de Oliveira Otto MC, Lee Y, Wu JHY, Song X, King IB, Psaty BM, Lemaitre RN, McKnight B, Siscovick DS, Mozaffarian D. Serial Plasma Phospholipid Fatty Acids in the De Novo Lipogenesis Pathway and Total Mortality, Cause-Specific Mortality, and Cardiovascular Diseases in the Cardiovascular Health Study. J Am Heart Assoc 2019; 8:e012881. [PMID: 31711385 PMCID: PMC6915264 DOI: 10.1161/jaha.119.012881] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Synthesized fatty acids (FAs) from de novo lipogenesis may affect cardiometabolic health, but longitudinal associations between serially measured de novo lipogenesis-related fatty acid biomarkers and mortality or cardiovascular disease (CVD) are not well established. Methods and Results We investigated longitudinal associations between de novo lipogenesis-related fatty acids with all-cause mortality, cause-specific mortality, and incident CVD among 3869 older US adults, mean (SD) age 75 (5) years and free of prevalent CVD at baseline. Levels of plasma phospholipid palmitic (16:0), palmitoleic (16:1n-7), stearic (18:0), oleic acid (18:1n-9), and other risk factors were serially measured at baseline, 6 years, and 13 years. All-cause mortality, cause-specific mortality, and incident fatal and nonfatal CVD were centrally adjudicated. Risk was assessed in multivariable-adjusted Cox models with time-varying FAs and covariates. During 13 years, median follow-up (maximum 22.4 years), participants experienced 3227 deaths (1131 CVD, 2096 non-CVD) and 1753 incident CVD events. After multivariable adjustment, higher cumulative levels of 16:0, 16:1n-7, and 18:1n-9 were associated with higher all-cause mortality, with extreme-quintile hazard ratios (95% CIs) of 1.35 (1.17-1.56), 1.40 (1.21-1.62), and 1.56 (1.35-1.80), respectively, whereas higher levels of 18:0 were associated with lower mortality (hazard ratio=0.76; 95% CI=0.66-0.88). Associations were generally similar for CVD mortality versus non-CVD mortality, as well as total incident CVD. Changes in levels of 16:0 were positively, and 18:0 inversely, associated with all-cause mortality (hazard ratio=1.23, 95% CI=1.08-1.41; and hazard ratio=0.78, 95% CI=0.68-0.90). Conclusions Higher long-term levels of 16:0, 16:1n-7, and 18:1n-9 and changes in 16:0 were positively, whereas long-term levels and changes in 18:0 were inversely, associated with all-cause mortality in older adults.
Collapse
Affiliation(s)
- Heidi T M Lai
- Friedman School of Nutrition Science and Policy Tufts University Boston MA
| | - Marcia C de Oliveira Otto
- Department of Epidemiology Human Genetics and Environmental Sciences University of Texas Health Science Center at Houston TX
| | - Yujin Lee
- Friedman School of Nutrition Science and Policy Tufts University Boston MA
| | - Jason H Y Wu
- The George Institute for Global Health Faculty of Medicine University of New South Wales Newtown NSW Australia
| | | | - Irena B King
- Department of Internal Medicine University of New Mexico Albuquerque NM
| | - Bruce M Psaty
- Department of Medicine, Epidemiology, and Health Services University of Washington Seattle WA.,Cardiovascular Health Research Unit Department of Medicine University of Washington Seattle WA.,Kaiser Permanente Washington Health Research Institute Seattle WA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit Department of Medicine University of Washington Seattle WA
| | | | | | | |
Collapse
|
195
|
Abstract
PURPOSE OF REVIEW We review recent evidence of the relationship between dietary fat intake and risk of type 2 diabetes (T2D), the role of epigenetic alterations as a mediator of this relationship, and the impact of gene-dietary fat interactions in the development of the disease. Based on the observations made, we will discuss whether there is evidence to support genetic personalization of fat intake recommendations in T2D prevention. RECENT FINDINGS Strong evidence suggests that polyunsaturated fatty acids (PUFA) have a protective effect on T2D risk, whereas the roles of saturated and monounsaturated fatty acids (SFA and MUFA) remain unclear. Diets enriched with PUFA vs SFA lead to distinct epigenetic alterations that may mediate their effects on T2D risk by changing gene function. However, it is not currently known which of the epigenetic alterations, if any, are causal for T2D. The current literature shows no replicated evidence of genetic variants modifying the effect of dietary fat intake on T2D risk. There is consistent evidence of a protective role of PUFA in T2D prevention. No evidence supports genetic personalization of dietary recommendations in T2D prevention.
Collapse
Affiliation(s)
- Germán D Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Mærsk Building (8. floor), Copenhagen, Denmark
| | - Hermina Jakupović
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Mærsk Building (8. floor), Copenhagen, Denmark
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Mærsk Building (8. floor), Copenhagen, Denmark.
| |
Collapse
|
196
|
Imamura F, Schulze MB, Sharp SJ, Guevara M, Romaguera D, Bendinelli B, Salamanca-Fernández E, Ardanaz E, Arriola L, Aune D, Boeing H, Dow C, Fagherazzi G, Franks PW, Freisling H, Jakszyn P, Kaaks R, Khaw KT, Kühn T, Mancini FR, Masala G, Chirlaque MD, Nilsson PM, Overvad K, Pala VM, Panico S, Perez-Cornago A, Quirós JR, Ricceri F, Rodríguez-Barranco M, Rolandsson O, Sluijs I, Stepien M, Spijkerman AMW, Tjønneland A, Tong TYN, Tumino R, Vissers LET, Ward HA, Langenberg C, Riboli E, Forouhi NG, Wareham NJ. Estimated Substitution of Tea or Coffee for Sugar-Sweetened Beverages Was Associated with Lower Type 2 Diabetes Incidence in Case-Cohort Analysis across 8 European Countries in the EPIC-InterAct Study. J Nutr 2019; 149:1985-1993. [PMID: 31396627 PMCID: PMC6825826 DOI: 10.1093/jn/nxz156] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/01/2019] [Accepted: 06/11/2019] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Beverage consumption is a modifiable risk factor for type 2 diabetes (T2D), but there is insufficient evidence to inform the suitability of substituting 1 type of beverage for another. OBJECTIVE The aim of this study was to estimate the risk of T2D when consumption of sugar-sweetened beverages (SSBs) was replaced with consumption of fruit juice, milk, coffee, or tea. METHODS In the European Prospective Investigation into Cancer and Nutrition (EPIC)-InterAct case-cohort study of 8 European countries (n = 27,662, with 12,333 cases of incident T2D, 1992-2007), beverage consumption was estimated at baseline by dietary questionnaires. Using Prentice-weighted Cox regression adjusting for other beverages and potential confounders, we estimated associations of substituting 1 type of beverage for another on incident T2D. RESULTS Mean ± SD of estimated consumption of SSB was 55 ± 105 g/d. Means ± SDs for the other beverages were as follows: fruit juice, 59 ± 101 g/d; milk, 209 ± 203 g/d; coffee, 381 ± 372 g/d; and tea, 152 ± 282 g/d. Substituting coffee for SSBs by 250 g/d was associated with a 21% lower incidence of T2D (95% CI: 12%, 29%). The rate difference was -12.0 (95% CI: -20.0, -5.0) per 10,000 person-years among adults consuming SSBs ≥250 g/d (absolute rate = 48.3/10,000). Substituting tea for SSBs was estimated to lower T2D incidence by 22% (95% CI: 15%, 28%) or -11.0 (95% CI: -20.0, -2.6) per 10,000 person-years, whereas substituting fruit juice or milk was estimated not to alter T2D risk significantly. CONCLUSIONS These findings indicate a potential benefit of substituting coffee or tea for SSBs for the primary prevention of T2D and may help formulate public health recommendations on beverage consumption in different populations.
Collapse
Affiliation(s)
- Fumiaki Imamura
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Matthias B Schulze
- Department of Molecular Epidemiology, Germen Institute of Human Nutrition, Potsdam, Germany
| | - Stephen J Sharp
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Marcela Guevara
- IDISNA Navarra Health Research Institute, Pamplona, Spain
- CIBER Epidemiology and Public Health, Madrid, Spain
| | - Dora Romaguera
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Health Research Institute of Balearic Islands (IdISBa), Palma de Mallorca, Spain
- CIBER Physiopathology of Obesity and Nutrition, Madrid, Spain
| | - Benedetta Bendinelli
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Elena Salamanca-Fernández
- CIBER Epidemiology and Public Health, Madrid, Spain
- Andalusian School of Public Health, Institute of Investigation Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University of Granada, Granada, Spain
| | - Eva Ardanaz
- IDISNA Navarra Health Research Institute, Pamplona, Spain
- CIBER Epidemiology and Public Health, Madrid, Spain
| | - Larraitz Arriola
- Public Health Division of Gipuzkoa, Instituto BIO-Donostia, Basque Government, CIBERESP, Gipuzkoa, Spain
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Nutrition, Bjørknes University College, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Heiner Boeing
- Department of Molecular Epidemiology, Germen Institute of Human Nutrition, Potsdam, Germany
| | - Courtney Dow
- Gustave Roussy Institute, Villejuif, France
- University Paris–South, Faculty of Medicine, University Versailles–St Quentin, University Paris-Saclay, Villejuif, France
- National Institute for Health and Medical Research (INSERM), Center for Research in Epidemiology and Population Health, Villejuif, France
| | - Guy Fagherazzi
- Gustave Roussy Institute, Villejuif, France
- University Paris–South, Faculty of Medicine, University Versailles–St Quentin, University Paris-Saclay, Villejuif, France
- National Institute for Health and Medical Research (INSERM), Center for Research in Epidemiology and Population Health, Villejuif, France
| | - Paul W Franks
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Lund University, Skane University Hospital, Malmo, Sweden
| | - Heinz Freisling
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC–WHO), Lyon, France
| | - Paula Jakszyn
- Catalan Institute of Oncology, Barcelona, Spain
- FCS Blanquerna, Universitat Ramon Llull, Barcelona, Spain
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Francesca R Mancini
- Gustave Roussy Institute, Villejuif, France
- University Paris–South, Faculty of Medicine, University Versailles–St Quentin, University Paris-Saclay, Villejuif, France
- National Institute for Health and Medical Research (INSERM), Center for Research in Epidemiology and Population Health, Villejuif, France
| | - Giovanna Masala
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Maria-Dolores Chirlaque
- CIBER Epidemiology and Public Health, Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Department of Health and Social Sciences, Universidad de Murcia, Murcia, Spain
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skane University Hospital, Malmo, Sweden
| | - Kim Overvad
- Department of Cardiology, Aalborg University Hospital, Aarhus, Denmark
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Valeria M Pala
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | | | - Fulvio Ricceri
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- Unit of Epidemiology, Regional Health Service ASL TO3, Grugliasco, Italy
| | - Miguel Rodríguez-Barranco
- CIBER Epidemiology and Public Health, Madrid, Spain
- Andalusian School of Public Health, Institute of Investigation Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), University of Granada, Granada, Spain
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
| | - Ivonne Sluijs
- Julius Center for Health Sciences and Primary Care, Cardiovascular Epidemiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Magdalena Stepien
- Section of Nutrition and Metabolism, International Agency for Research on Cancer (IARC–WHO), Lyon, France
| | | | | | - Tammy Y N Tong
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, “Civic-M.P. Arezzo” Hospital, Ragusa, Italy
| | - Linda E T Vissers
- Julius Center for Health Sciences and Primary Care, Cardiovascular Epidemiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Heather A Ward
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Claudia Langenberg
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nick J Wareham
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
197
|
Drouin-Chartier JP, Li Y, Ardisson Korat AV, Ding M, Lamarche B, Manson JE, Rimm EB, Willett WC, Hu FB. Changes in dairy product consumption and risk of type 2 diabetes: results from 3 large prospective cohorts of US men and women. Am J Clin Nutr 2019; 110:1201-1212. [PMID: 31504094 PMCID: PMC6821541 DOI: 10.1093/ajcn/nqz180] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/10/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Whether changes in dairy product consumption are related to subsequent risk of type 2 diabetes (T2D) remains unknown. OBJECTIVE We evaluated the association of long-term changes in dairy product consumption with subsequent risk of T2D among US men and women. METHODS We followed up 34,224 men in the Health Professionals Follow-Up Study (1986-2012), 76,531 women in the Nurses' Health Study (1986-2012), and 81,597 women in the Nurses' Health Study II (1991-2013). Changes in dairy consumption were calculated from consecutive quadrennial FFQs. Multivariable Cox proportional regression models were used to calculate HRs for T2D associated with changes in dairy product consumption. Results of the 3 cohorts were pooled using an inverse variance-weighted, fixed-effect meta-analysis. RESULTS During 2,783,210 person-years, we documented 11,906 incident T2D cases. After adjustment for initial and changes in diet and lifestyle covariates, decreasing total dairy intake by >1.0 serving/d over a 4-y period was associated with an 11% (95% CI: 3%, 19%) higher risk of T2D in the subsequent 4 y compared with maintaining a relatively stable consumption (i.e., change in intake of ±1.0 serving/wk). Increasing yogurt consumption by >0.5 serving/d was associated with an 11% (95% CI: 4%, 18%) lower T2D risk, whereas increasing cheese consumption by >0.5 serving/d was associated with a 9% (95% CI: 2%, 16%) higher risk compared with maintaining stable intakes. Substituting 1 serving/d of yogurt or reduced-fat milk for cheese was associated with a 16% (95% CI: 10%, 22%) or 12% (95% CI: 8%, 16%) lower T2D risk, respectively. CONCLUSIONS Increasing yogurt consumption was associated with a moderately lower risk of T2D, whereas increasing cheese consumption was associated with a moderately higher risk among US men and women. Our study suggests that substituting yogurt or reduced-fat milk for cheese is associated with a lower risk of T2D.
Collapse
Affiliation(s)
- Jean-Philippe Drouin-Chartier
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Address correspondence to J-PD-C e-mail:
| | - Yanping Li
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Andres Victor Ardisson Korat
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ming Ding
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Benoît Lamarche
- School of Nutrition and Institute of Nutrition and Functional Foods, Laval University, Quebec City, Quebec, Canada
| | - JoAnn E Manson
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA,Address correspondence to FBH e-mail:
| |
Collapse
|
198
|
Murphy AM, Smith CE, Murphy LM, Follis JL, Tanaka T, Richardson K, Noordam R, Lemaitre RN, Kähönen M, Dupuis J, Voortman T, Marouli E, Mook‐Kanamori DO, Raitakari OT, Hong J, Dehghan A, Dedoussis G, de Mutsert R, Lehtimäki T, Liu C, Rivadeneira F, Deloukas P, Mikkilä V, Meigs JB, Uitterlinden A, Ikram MA, Franco OH, Hughes M, O' Gaora P, Ordovás JM, Roche HM. Potential Interplay between Dietary Saturated Fats and Genetic Variants of the NLRP3 Inflammasome to Modulate Insulin Resistance and Diabetes Risk: Insights from a Meta-Analysis of 19 005 Individuals. Mol Nutr Food Res 2019; 63:e1900226. [PMID: 31432628 PMCID: PMC6864231 DOI: 10.1002/mnfr.201900226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/12/2019] [Indexed: 12/13/2022]
Abstract
SCOPE Insulin resistance (IR) and inflammation are hallmarks of type 2 diabetes (T2D). The nod-like receptor pyrin domain containing-3 (NLRP3) inflammasome is a metabolic sensor activated by saturated fatty acids (SFA) initiating IL-1β inflammation and IR. Interactions between SFA intake and NLRP3-related genetic variants may alter T2D risk factors. METHODS Meta-analyses of six Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium (n = 19 005) tested interactions between SFA and NLRP3-related single-nucleotide polymorphisms (SNPs) and modulation of fasting insulin, fasting glucose, and homeostasis model assessment of insulin resistance. RESULTS SFA interacted with rs12143966, wherein each 1% increase in SFA intake increased insulin by 0.0063 IU mL-1 (SE ± 0.002, p = 0.001) per each major (G) allele copy. rs4925663, interacted with SFA (β ± SE = -0.0058 ± 0.002, p = 0.004) to increase insulin by 0.0058 IU mL-1 , per additional copy of the major (C) allele. Both associations are close to the significance threshold (p < 0.0001). rs4925663 causes a missense mutation affecting NLRP3 expression. CONCLUSION Two NLRP3-related SNPs showed potential interaction with SFA to modulate fasting insulin. Greater dietary SFA intake accentuates T2D risk, which, subject to functional validation, may be further elaborated depending on NLRP3-related genetic variants.
Collapse
Affiliation(s)
- Aoife M. Murphy
- Nutrigenomics Research GroupConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - Caren E. Smith
- Jean Mayer USDA Human Nutrition Research Centre on AgingTufts UniversityBostonMA02111USA
| | - Leanne M. Murphy
- UCD School of Biomolecular and Biomedical ScienceConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - Jack L. Follis
- Department of MathematicsUniversity of St. ThomasHoustonTX77006‐4626USA
| | - Toshiko Tanaka
- Translational Gerontology BranchNational Institute on AgingBaltimoreMD21224USA
| | - Kris Richardson
- Jean Mayer USDA Human Nutrition Research Centre on AgingTufts UniversityBostonMA02111USA
| | - Raymond Noordam
- Department of Internal MedicineSection of Gerontology and Geriatrics, Leiden University Medical CenterLeiden2333 ZA.The Netherlands
| | | | - Mika Kähönen
- Department of Clinical PhysiologyTampere University Hospital and University of Tampere School of Medicine33521TampereFinland
| | - Josée Dupuis
- Department of BiostatisticsBoston University School of Public HealthBostonMA02130USA
| | - Trudy Voortman
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Eirini Marouli
- William Harvey Research InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonLondonE1 4NSUK
| | - Dennis O. Mook‐Kanamori
- Department of Clinical Epidemiology and Department of Public Health and Primary CareLeiden University Medical CenterAlbinusdreef 22333 ZALeidenThe Netherlands
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear MedicineTurku University Hospital, and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku20521TurkuFinland
| | - Jaeyoung Hong
- Department of BiostatisticsBoston University School of Public HealthBostonMA02130USA
| | - Abbas Dehghan
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - George Dedoussis
- Department of Nutrition and DieteticsSchool of Health Science and Education, Harokopio UniversityEl. Venizelou 7017671AthensGreece
| | - Renée de Mutsert
- Department of Clinical Epidemiology and Department of Public Health and Primary CareLeiden University Medical CenterAlbinusdreef 22333 ZALeidenThe Netherlands
| | - Terho Lehtimäki
- Department of Clinical ChemistryFimlab Laboratories and Finnish Cardiovascular Research Center–TampereFaculty of Medicine and Life Sciences, University of TampereTampere33520Finland
| | - Ching‐Ti Liu
- Department of BiostatisticsBoston University School of Public HealthBostonMA02130USA
| | - Fernando Rivadeneira
- Department of Internal MedicineErasmus University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Panagiotis Deloukas
- William Harvey Research InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonLondonE1 4NSUK
| | - Vera Mikkilä
- Division of NutritionDepartment of Food and Environmental Sciences00014HelsinkiFinland
| | - James B. Meigs
- Division of General Internal MedicineMassachusetts General HospitalBostonMA02114USA
- Harvard Medical SchoolBostonMA02115USA
- Broad InstituteCambridgeMA02142USA
| | - Andre Uitterlinden
- Department of Internal MedicineErasmus University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Mohammad A. Ikram
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Oscar H. Franco
- Department of EpidemiologyErasmus MC‐University Medical CenterPostbus 2040, 3000 CARotterdamThe Netherlands
| | - Maria Hughes
- Nutrigenomics Research GroupConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - Peadar O' Gaora
- UCD School of Biomolecular and Biomedical ScienceConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
| | - José M. Ordovás
- Jean Mayer USDA Human Nutrition Research Centre on AgingTufts UniversityBostonMA02111USA
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)28029MadridSpain
- IMDEA Food Institute, CEI UAM + CSICE ‐ 28049MadridSpain
| | - Helen M. Roche
- Nutrigenomics Research GroupConway Institute of Biomedical and Biomolecular SciencesUniversity College DublinBelfieldDublin 4, D04 V1W8Ireland
- Institute For Global Food SecurityQueen's University BelfastNorthern Ireland
| |
Collapse
|
199
|
Seah JYH, Ong CN, Koh WP, Yuan JM, van Dam RM. A Dietary Pattern Derived from Reduced Rank Regression and Fatty Acid Biomarkers Is Associated with Lower Risk of Type 2 Diabetes and Coronary Artery Disease in Chinese Adults. J Nutr 2019; 149:2001-2010. [PMID: 31386157 PMCID: PMC6825830 DOI: 10.1093/jn/nxz164] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/10/2019] [Accepted: 06/18/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Combinations of circulating fatty acids may affect the risk of type 2 diabetes (T2D) and coronary artery disease (CAD). No previous studies have identified a dietary pattern predicting fatty acid profiles using reduced rank regression (RRR) and evaluated its associations with the risk of T2D and CAD. OBJECTIVE The aim of this study was to derive a dietary pattern to explain variation in plasma fatty acid concentrations using RRR and evaluate these in relation to risk of T2D and CAD. METHODS We derived a dietary pattern using fatty acid concentrations from 711 controls of a nested case-control study in the Singapore Chinese Health Study using RRR with 36 food and beverages as predictors and 19 fatty acid biomarkers as responses. Dietary pattern scores were then calculated for the full cohort of men and women (mean age: 56 y). We followed up 45,411 and 58,065 participants for incident T2D and CAD mortality, respectively. Multivariable Cox regression models were used to estimate HRs and 95% CIs. RESULTS We identified a dietary pattern high in soy, vegetables, fruits, tea, tomato products, bread, fish, margarine and dairy, and low in rice, red meat, coffee, alcohol, sugar-sweetened beverages, and eggs. This pattern predicted higher circulating n-3 (ω-3) PUFAs (18:3n-3, 20:3n-3, 20:5n-3), odd-chain fatty acids (15:0, 17:0), 18:2n-6 and 20:1, and lower 20:4n-6 and 16:1. During a mean follow-up of 11 y and 19 y, 5207 T2D and 3016 CAD mortality events, respectively, were identified. Higher dietary pattern scores were associated with a lower risk of T2D [multivariable-adjusted HR comparing extreme quintiles, 0.86 (95% CI: 0.79, 0.95); P-trend <0.001] and CAD mortality [HR, 0.76 (95% CI: 0.68, 0.86); P-trend <0.001]. CONCLUSIONS Dietary patterns reflecting higher circulating n-3 PUFAs, odd-chain fatty acids, and linoleic acid may be associated with lower T2D and CAD risk in Chinese adults. This trial was registered at www.clinicaltrials.gov as NCT03356340.
Collapse
Affiliation(s)
- Jowy Y H Seah
- Saw Swee Hock School of Public Health, National University of Singapore (NUS), Singapore
- NUS Graduate School for Integrative Sciences and Engineering, NUS, Singapore
| | - Choon Nam Ong
- Saw Swee Hock School of Public Health, National University of Singapore (NUS), Singapore
- NUS Environmental Research Institute, National University of Singapore, Singapore
| | - Woon-Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore (NUS), Singapore
- Health Services and Systems Research, Duke-NUS Medical School, Singapore
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore (NUS), Singapore
- NUS Graduate School for Integrative Sciences and Engineering, NUS, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, NUS and National University Health System, Singapore
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
200
|
Guo J, Givens DI, Astrup A, Bakker SJL, Goossens GH, Kratz M, Marette A, Pijl H, Soedamah‐Muthu SS. The Impact of Dairy Products in the Development of Type 2 Diabetes: Where Does the Evidence Stand in 2019? Adv Nutr 2019; 10:1066-1075. [PMID: 31124561 PMCID: PMC6855942 DOI: 10.1093/advances/nmz050] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/26/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased rapidly. Adopting a heathy diet is suggested as one of the effective behaviors to prevent or delay onset of T2D. Dairy consumption has been recommended as part of a healthy diet, but there remains uncertainty in both the scientific community and the public about the effect of different dairy products on T2D risk. In a recent workshop, the evidence on dairy products and T2D risk was presented and discussed by a group of experts. The main conclusions from the workshop are presented in this position paper and are as follows. 1) Available evidence from large prospective cohort studies and limited randomized controlled trials (RCTs) suggests that total dairy consumption has a neutral or moderately beneficial effect on T2D risk. 2) Increasing evidence from prospective cohort studies indicates that yogurt is most strongly associated with a lower T2D risk, but evidence from RCTs is scarce. 3) Fatty acids from dairy (medium-chain, odd, and very long-chain SFAs as well as trans-palmitoleic acid) are associated with lower T2D risk and improved metabolic health, but more research is needed on studies that explore cause and effect relations to exclude the possibility that the dairy fatty acids simply serve as markers of overall dairy consumption. 4) The food matrix can be a stronger determinant of health effects than SFA content. This review further identifies research gaps in the existing knowledge and highlights key research questions that need to be addressed to better understand the impact of dairy consumption on future T2D risk.
Collapse
Affiliation(s)
- Jing Guo
- Institute for Food, Nutrition and Health, University of Reading, Reading, United Kingdom
| | - D Ian Givens
- Institute for Food, Nutrition and Health, University of Reading, Reading, United Kingdom
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, Netherlands
| | - Mario Kratz
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - André Marette
- Faculty of Medicine, Laval University, Quebec, Canada
| | - Hanno Pijl
- Section of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Sabita S Soedamah‐Muthu
- Institute for Food, Nutrition and Health, University of Reading, Reading, United Kingdom
- Department of Medical and Clinical Psychology, Center of Research on Psychology in Somatic Diseases, Tilburg University, Tilburg, Netherlands
| |
Collapse
|