1
|
Slurink IAL, Kupper N, Smeets T, Soedamah-Muthu SS. Dairy consumption and risk of prediabetes and type 2 diabetes in the Fenland study. Clin Nutr 2024; 43:69-79. [PMID: 39353264 DOI: 10.1016/j.clnu.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 08/25/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND & AIMS Limited observational evidence suggests that a higher intake of high-fat dairy may be associated with lower prediabetes risk, while opposite associations have been observed for low-fat milk intake. This study aimed to examine associations between baseline and changes in dairy consumption, risk of prediabetes, and glycaemic status. METHODS 7521 participants from the prospective UK Fenland study were included (mean age 48.7 ± 2.0 years, 51.9 % female). Dairy intake was measured using self-reported food frequency questionnaires. Associations with prediabetes risk and glycaemic status were analysed using Poisson regression models adjusted for social demographics, health behaviours, family history of diabetes and food group intake. RESULTS At a mean follow-up of 6.7 ± 2.0 years, 290 participants developed prediabetes (4.3 %). Most dairy products were not significantly associated with prediabetes risk. A higher baseline intake of high-fat dairy (RRservings/day 1.20, 95%CI 1.03-1.39) and high-fat milk (RRservings/day 1.22, 1.01-1.47) were associated with higher prediabetes risk. Conversely, low-fat milk was associated with lower prediabetes risk (RRservings/day 0.86, 0.75-0.98). In the analyses evaluating dietary changes over time, increases in high-fat milk were inversely associated with risk of progressing from normoglycaemia to prediabetes or type 2 diabetes (RRservings/day 0.86, 95%CI 0.75-0.99). CONCLUSIONS This population-based study showed that most dairy products are not associated with prediabetes risk or progression in glycaemic status. Positive associations of high-fat dairy, high-fat milk, and the inverse association of low-fat milk with prediabetes risk found were inconsistent with prior literature and suggestive of the need for future research on environmental, behavioural, and biological factors that explain the available evidence.
Collapse
Affiliation(s)
- Isabel A L Slurink
- Center of Research on Psychological Disorders and Somatic Diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, PO Box 90153, 5000 LE, Tilburg, the Netherlands.
| | - Nina Kupper
- Center of Research on Psychological Disorders and Somatic Diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, PO Box 90153, 5000 LE, Tilburg, the Netherlands
| | - Tom Smeets
- Center of Research on Psychological Disorders and Somatic Diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, PO Box 90153, 5000 LE, Tilburg, the Netherlands
| | - Sabita S Soedamah-Muthu
- Center of Research on Psychological Disorders and Somatic Diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, PO Box 90153, 5000 LE, Tilburg, the Netherlands; Institute for Food, Nutrition and Health, University of Reading, Reading RG6 6AR, United Kingdom
| |
Collapse
|
2
|
Marklund M, Billyrose S, Orji IA, Ikechukwu-Orji MU, Okoro C, Obagha C, Iyer G, Jamro EL, Ojo A, Harris WS, Wu JH, Hirschhorn LR, Van Horn L, Huffman MD, Ojji DB. Blood biomarkers of trans-fatty acid intake among Nigerian adults in the Federal Capital Territory: a cross-sectional study. Am J Clin Nutr 2024:S0002-9165(24)00860-8. [PMID: 39490795 DOI: 10.1016/j.ajcnut.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Intake of trans-fatty acids (TFAs) is an established risk factor for cardiovascular disease. In April 2023, Nigeria passed regulations limiting TFA content in foods, fats, and oils, but the current level of TFA exposure in the Nigerian population is unknown. We assessed TFA biomarkers in dried blood spots from Nigerian adults in the Federal Capital Territory before policy enforcement to establish a baseline for future evaluation. METHODS We used gas chromatography to measure TFA content in dried blood spots from adults participating in a cross-sectional household survey using a representative sampling frame. Individual TFA (t-16:1, t-18:1, and t- 18:2) and their total were expressed as percentage of total fatty acids. We assessed differences in TFA levels between subgroups based on sex, age, body mass index (BMI), education, income, and local government area using multivariable-adjusted linear regression models. Mean TFA levels were compared with samples from individuals in 30 countries. RESULTS In 213 adults (62% females; mean age: 36 y, mean BMI: 25.9 kg/m2), the mean TFA level in dried blood spots was 0.61% of total fatty acids (range: 0.23%-1.31%). In multivariable-adjusted models, TFA levels were higher in younger adults {<30 y compared with ≥42 y, 0.07% [95% confidence interval (CI): 0.00, 0.15], P = 0.047}, those without a high school degree [compared with higher education, 0.08% (95% CI: 0.01, 0.16), P = 0.023], and residents of Abuja Municipal Area Council [compared with residents in Gwagwalada, 0.12% (95% CI: 0.05, 0.20), P = 0.001]. Total TFA levels were comparable with international samples, but t-16:1 and t-18:1 appeared lower, whereas t-18:2 appeared greater (52% of all TFA), in the Nigerian samples. CONCLUSIONS These results provide a baseline assessment of TFA exposure in Nigerian adults to evaluate implementation and effect of national regulation passed in 2023. The observed subgroup differences may help identify subpopulations for targeted interventions to reduce TFA intake.
Collapse
Affiliation(s)
- Matti Marklund
- Center for Human Nutrition, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, MD, United States; The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Clinical Nutrition and Metabolism Unit, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden.
| | - Soji Billyrose
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Ikechukwu A Orji
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Mercy U Ikechukwu-Orji
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Clementina Okoro
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Chijioke Obagha
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - Guhan Iyer
- Washington University in St. Louis, Saint Louis, MO, United States
| | - Erica L Jamro
- Washington University in St. Louis, Saint Louis, MO, United States
| | - Adedayo Ojo
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria
| | - William S Harris
- Fatty Acid Research Institute, Sioux Falls, SD, United States; University of South Dakota, Sioux Falls, SD, United States
| | - Jason Hy Wu
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; School of Population Health, UNSW Sydney, Sydney, NSW, Australia
| | - Lisa R Hirschhorn
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Robert J Havey Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Linda Van Horn
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Robert J Havey Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Mark D Huffman
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Washington University in St. Louis, Saint Louis, MO, United States; Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Dike B Ojji
- Cardiovascular Research Unit, University of Abuja, and University of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria; Department of Internal Medicine, Faculty of Clinical Sciences, University of Abuja, Abuja, Nigeria
| |
Collapse
|
3
|
Sowah SA, Koulman A, Sharp SJ, Imamura F, Khaw KT, Forouhi NG. Effects of coconut oil, olive oil, and butter on plasma fatty acids and metabolic risk factors: a randomised trial. J Lipid Res 2024:100681. [PMID: 39490924 DOI: 10.1016/j.jlr.2024.100681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/19/2024] [Accepted: 10/13/2024] [Indexed: 11/05/2024] Open
Abstract
There is limited evidence on the effects of different dietary sources of fats on detailed blood fatty acids (FAs). We aimed to evaluate the effects of coconut oil, olive oil and butter on circulating FA concentrations, and examine the associations between changes in plasma FAs and changes in metabolic markers. We conducted secondary analyses in the COB (coconut oil, olive oil and butter) Trial that evaluated 96 healthy adults in a 4-week parallel randomised controlled trial of three dietary interventions: 50 g/d of extra-virgin coconut oil (n=30), extra-virgin olive oil (n=33) or unsalted butter (n=33). We measured plasma phospholipid FA concentrations (mol% of total) using gas-chromatography. Using linear regression, we estimated the effects of the interventions on changes in FAs and the associations of changes in selected FAs with changes in metabolic markers. Coconut oil doubled lauric acid (C12:0) and myristic acid (C14:0), butter increased those to a lesser extent, and olive oil reduced those. β (95% confidence interval) for changes in C12:0 comparing coconut oil to butter and olive oil were +0.04 (0.03-0.05) and +0.05 (0.04-0.06) mol%, respectively; for C14:0, +0.24 (0.17-0.32) and +0.37 (0.29-0.45), respectively. Olive oil increased oleic acid (OA) approximately by 1 mol%, while coconut oil and butter had little effect on OA. Butter increased odd-chain SFAs and trans-FAs while olive oil and coconut oil decreased them. Changes in FAs mostly showed no significant associations with changes in metabolic markers. The interventions of equal amounts of different food FA sources altered circulating FA concentrations differently.
Collapse
Affiliation(s)
- Solomon A Sowah
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK (SAS, AK, SJS, FI, KTK, NGF)
| | - Albert Koulman
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK (SAS, AK, SJS, FI, KTK, NGF)
| | - Stephen J Sharp
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK (SAS, AK, SJS, FI, KTK, NGF)
| | - Fumiaki Imamura
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK (SAS, AK, SJS, FI, KTK, NGF)
| | - Kay-Tee Khaw
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK (SAS, AK, SJS, FI, KTK, NGF)
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK (SAS, AK, SJS, FI, KTK, NGF).
| |
Collapse
|
4
|
Wang J, Zheng S, Li Z, Tang Y, Huang Y, Wang J, Li R, Peng J. Pentadecanoic acid (C15:0, PA) induces mild maternal glucose intolerance and promotes the growth of the offspring partly through up-regulating liver PPARα and MAPK signaling pathways. Food Funct 2024. [PMID: 39434548 DOI: 10.1039/d4fo03970j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Gestational diabetes mellitus (GDM) is one of the most common metabolic disturbances during pregnancy, which poses a serious threat to both maternal and offspring health. Pentadecanoic acid (C15:0, PA) is one of the most common odd-chain saturated fatty acids (OCS-FAs). However, its safety and nutritional value are yet to be verified. Herein, we provide a systematic assessment of the effects of PA on maternal and progeny health and insulin sensitivity for the first time. Our results showed that consumption of 1% PA during pregnancy could increase the contents of PA and heptadecanoic acid (C17:0) in maternal plasma, fetal tissue and offspring plasma, but it had no effect on embryonic development. During pregnancy, PA treatment caused mild insulin resistance, while it had little effect on the maternal body composition. During lactation, PA treatment caused mild insulin resistance and oxidative stress. Maternal body fat deposition was also reduced, but the growth rate of the offspring was faster. It is worth noting that PA treatment decreased plasma and liver TG content and increased the antioxidant capacity of the offspring. The effect of PA on the transcription and expression genes in the liver of pregnant mice was investigated using RNA-seq. PPARα and MAPK signaling pathways, both closely related to lipolysis, inflammation, oxidative stress, and insulin resistance were significantly increased. The expression of c-JUN, ERK, JNK and P65 proteins was also significantly up-regulated. In conclusion, our results suggest that 1% PA can induce a mild decrease in the maternal glucose tolerance and lipolysis mainly by activated MAPK and PPARα signaling. Moreover, low concentrations of PA may be an effective nutrient to alleviate the oxidative stress and reduce blood lipid levels of offspring.
Collapse
Affiliation(s)
- Jun Wang
- Animal Husbandry and Fisheries Research Center of Guangdong Haid Group Co., Ltd., Guangzhou 511400, China.
| | - Shiqi Zheng
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Ziying Li
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Yimei Tang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Yanhua Huang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| | - Junwen Wang
- Division of AOS & CDC, Faculty of Dentistry, and State Key Lab of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077 China
| | - Rui Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China.
| | - Jie Peng
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong, 510225, China
| |
Collapse
|
5
|
Ciesielski V, Legrand P, Blat S, Rioux V. New insights on pentadecanoic acid with special focus on its controversial essentiality: A mini-review. Biochimie 2024:S0300-9084(24)00234-7. [PMID: 39395658 DOI: 10.1016/j.biochi.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Pentadecanoic acid (C15:0, PDA) is an odd and minor fatty acid that has been neglected in the literature until the last decade. Indeed, as a specific fatty acid of dairy fat, PDA was only used as a biomarker of dairy fat consumption. Lately, PDA was first correlated negatively with the incidence of metabolic syndrome disorder, then its physiological effects have been investigated as a protective fatty acid. PDA supplementation has been demonstrated as negatively correlated with elevated levels of leptin, plasminogen activator inhibitor-1 and insulin, and has been shown to exhibit sensitizing insulin effects with activation of AMPK pathway. PDA also reduced the severity of metabolic dysfunction-associated steatohepatitis (MASH), notably through reduced alanine transaminase and pro-inflammatory cytokines levels. The final effect described for PDA is its ability to display anti-inflammatory properties in several pathology models. Hence, considering these multiple effects, the presence of PDA could be associated with a healthier physiological state, this raises the question of whether the presence of PDA in the body, in adequate quantities, is needed to participate to health maintenance. PDA is not synthesized in sufficient quantities endogenously, so it must be provided by the diet, mainly through dairy fat, although other types of food can also contribute to the dietary intake of PDA. Essential fatty acids are described as not being endogenously synthesized in sufficient and required quantities to maintain physiological health. Thus, PDA might gather both conditions to be described as essential, yet further investigations on both criteria are needed to enhance knowledge on this odd chain fatty acid with promising impact as potential protective supplement nutrient.
Collapse
Affiliation(s)
- Vincent Ciesielski
- Institut Agro Rennes Angers, Rennes, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Philippe Legrand
- Institut Agro Rennes Angers, Rennes, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Sophie Blat
- Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France
| | - Vincent Rioux
- Institut Agro Rennes Angers, Rennes, France; Institut Numecan, INRAE, INSERM, Univ Rennes, Rennes, France.
| |
Collapse
|
6
|
Eichelmann F, Prada M, Sellem L, Jackson KG, Salas Salvadó J, Razquin Burillo C, Estruch R, Friedén M, Rosqvist F, Risérus U, Rexrode KM, Guasch-Ferré M, Sun Q, Willett WC, Martinez-Gonzalez MA, Lovegrove JA, Hu FB, Schulze MB, Wittenbecher C. Lipidome changes due to improved dietary fat quality inform cardiometabolic risk reduction and precision nutrition. Nat Med 2024; 30:2867-2877. [PMID: 38992128 PMCID: PMC11485259 DOI: 10.1038/s41591-024-03124-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 06/11/2024] [Indexed: 07/13/2024]
Abstract
Current cardiometabolic disease prevention guidelines recommend increasing dietary unsaturated fat intake while reducing saturated fats. Here we use lipidomics data from a randomized controlled dietary intervention trial to construct a multilipid score (MLS), summarizing the effects of replacing saturated fat with unsaturated fat on 45 lipid metabolite concentrations. In the EPIC-Potsdam cohort, a difference in the MLS, reflecting better dietary fat quality, was associated with a significant reduction in the incidence of cardiovascular disease (-32%; 95% confidence interval (95% CI): -21% to -42%) and type 2 diabetes (-26%; 95% CI: -15% to -35%). We built a closely correlated simplified score, reduced MLS (rMLS), and observed that beneficial rMLS changes, suggesting improved dietary fat quality over 10 years, were associated with lower diabetes risk (odds ratio per standard deviation of 0.76; 95% CI: 0.59 to 0.98) in the Nurses' Health Study. Furthermore, in the PREDIMED trial, an olive oil-rich Mediterranean diet intervention primarily reduced diabetes incidence among participants with unfavorable preintervention rMLS levels, suggestive of disturbed lipid metabolism before intervention. Our findings indicate that the effects of dietary fat quality on the lipidome can contribute to a more precise understanding and possible prediction of the health outcomes of specific dietary fat modifications.
Collapse
Affiliation(s)
- Fabian Eichelmann
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Marcela Prada
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Laury Sellem
- Hugh Sinclair Unit of Human Nutrition, Institute for Cardiovascular and Metabolic Research and Institute for Food, Nutrition and Health, Reading, UK
| | - Kim G Jackson
- Hugh Sinclair Unit of Human Nutrition, Institute for Cardiovascular and Metabolic Research and Institute for Food, Nutrition and Health, Reading, UK
| | - Jordi Salas Salvadó
- Human Nutrition Unit, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Cristina Razquin Burillo
- Human Nutrition Unit, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, Pamplona, Spain
| | - Ramon Estruch
- Human Nutrition Unit, Department of Biochemistry and Biotechnology, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
- Department of Internal Medicine, Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Michael Friedén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Frederik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Kathryn M Rexrode
- Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marta Guasch-Ferré
- Department of Public Health and Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. 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 T.H. Chan School of Public Health, Boston, MA, USA
| | - Miguel Angel Martinez-Gonzalez
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Preventive Medicine and Public Health, IdiSNA (Instituto de Investigación Sanitaria de Navarra), University of Navarra, Pamplona, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Institute for Cardiovascular and Metabolic Research and Institute for Food, Nutrition and Health, Reading, UK
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Clemens Wittenbecher
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany.
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
7
|
Martínez-González MA, Planes FJ, Ruiz-Canela M, Toledo E, Estruch R, Salas-Salvadó J, Valdés-Más R, Mena P, Castañer O, Fitó M, Clish C, Landberg R, Wittenbecher C, Liang L, Guasch-Ferré M, Lamuela-Raventós RM, Wang DD, Forouhi N, Razquin C, Hu FB. Recent advances in precision nutrition and cardiometabolic diseases. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2024:S1885-5857(24)00279-2. [PMID: 39357800 DOI: 10.1016/j.rec.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Abstract
A growing body of research on nutrition omics has led to recent advances in cardiovascular disease epidemiology and prevention. Within the PREDIMED trial, significant associations between diet-related metabolites and cardiovascular disease were identified, which were subsequently replicated in independent cohorts. Some notable metabolites identified include plasma levels of ceramides, acyl-carnitines, branched-chain amino acids, tryptophan, urea cycle pathways, and the lipidome. These metabolites and their related pathways have been associated with incidence of both cardiovascular disease and type 2 diabetes. Future directions in precision nutrition research include: a) developing more robust multimetabolomic scores to predict long-term risk of cardiovascular disease and mortality; b) incorporating more diverse populations and a broader range of dietary patterns; and c) conducting more translational research to bridge the gap between precision nutrition studies and clinical applications.
Collapse
Affiliation(s)
- Miguel A Martínez-González
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States.
| | - Francisco J Planes
- Tecnun Escuela de Ingeniería, Departamento de Ingeniería Biomédica y Ciencias, Universidad de Navarra, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Estefanía Toledo
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Departamento de Medicina Interna, Instituto de Investigaciones Biomédicas August Pi Sunyer (IDIBAPS), Hospital Clínico, Universidad de Barcelona, Barcelona, Spain
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria Pere i Virgili, Departamento de Bioquímica y Biotecnología, Unidad de Nutrición Humana Universidad Rovira i Virgili, Reus, Tarragona, Spain
| | - Rafael Valdés-Más
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Pedro Mena
- Dipartimento di Scienze degli Alimenti e del Farmaco, Universitá di Parma, Parma, Italy
| | - Olga Castañer
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Spain
| | - Montse Fitó
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Unidad de Riesgo Cardiovascular y Nutrición, Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States
| | - Rikard Landberg
- Division of Food and Nutrition Science, Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Department of Public Health and Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Rosa M Lamuela-Raventós
- Grup de recerca antioxidants naturals: polifenols, Departament de Nutrició, Ciències de l'Alimentació i Gastronomia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain; Institut de Nutrició i Seguretat Alimentària (INSA), Universitat de Barcelona (UB), Barcelona, Spain
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Nita Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Cristina Razquin
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Universidad de Navarra, Departamento de Medicina Preventiva y Salud Pública, Pamplona, Navarra, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
8
|
Śledziński M, Gołębiewska J, Mika A. The Long-Term Effect of Kidney Transplantation on the Serum Fatty Acid Profile. Nutrients 2024; 16:3319. [PMID: 39408286 PMCID: PMC11478970 DOI: 10.3390/nu16193319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Epidemiologic evidence has demonstrated the prevalence of metabolic disorders and increased cardiovascular risk related to lipid metabolism disorders in kidney transplant recipients. Therefore, it is of great importance to understand lipid alterations and to look for ways to reduce cardiovascular risk in this patient group. Methods: Our study included 25 patients with chronic kidney disease undergoing kidney transplantation (KTx). Three blood samples were taken from each patient: before KTx, 3 months after KTx and 6-12 months after KTx. A series of biochemical blood tests and a detailed analysis of the serum fatty acid profile were performed. Results: In our previous study, the effects of kidney transplantation on serum fatty acid (FA) profile 3 months after the procedure were investigated. The current study shows the longer-term (6-12 months) effects of the procedure on the serum FA profile. We found that although n-3 polyunsaturated FA levels started to decrease 3 months after surgery, they normalized over a longer period of time (6-12 months). Furthermore, we observed a strong decrease in ultra-long-chain FAs and an increase in odd-chain FAs over a longer time after kidney transplantation. All of the above FAs may have an important impact on human health, including inflammation, cardiovascular risk or cancer risk. Conclusions: The changes in serum FA profiles after kidney transplantation are a dynamic process and that more detailed studies could provide an accurate indication for supplementation with some FAs or diet modification.
Collapse
Affiliation(s)
- Maciej Śledziński
- Department of General, Endocrine and Transplant Surgery, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Justyna Gołębiewska
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland;
| | - Adriana Mika
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdansk, Poland
- Department of Environmental Analytics, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland
| |
Collapse
|
9
|
Yuzbashian E, Berg E, de Campos Zani SC, Chan CB. Cow's Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies. Foods 2024; 13:2837. [PMID: 39272602 PMCID: PMC11395457 DOI: 10.3390/foods13172837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Obesity disrupts glucose metabolism, leading to insulin resistance (IR) and cardiometabolic diseases. Consumption of cow's milk and other dairy products may influence glucose metabolism. Within the complex matrix of cow's milk, various carbohydrates, lipids, and peptides act as bioactive molecules to alter human metabolism. Here, we summarize data from human studies and rodent experiments illustrating how these bioactive molecules regulate insulin and glucose homeostasis, supplemented with in vitro studies of the mechanisms behind their effects. Bioactive carbohydrates, including lactose, galactose, and oligosaccharides, generally reduce hyperglycemia, possibly by preventing gut microbiota dysbiosis. Milk-derived lipids of the milk fat globular membrane improve activation of insulin signaling pathways in animal trials but seem to have little impact on glycemia in human studies. However, other lipids produced by ruminants, including polar lipids, odd-chain, trans-, and branched-chain fatty acids, produce neutral or contradictory effects on glucose metabolism. Bioactive peptides derived from whey and casein may exert their effects both directly through their insulinotropic effects or renin-angiotensin-aldosterone system inhibition and indirectly by the regulation of incretin hormones. Overall, the results bolster many observational studies in humans and suggest that cow's milk intake reduces the risk of, and can perhaps be used in treating, metabolic disorders. However, the mechanisms of action for most bioactive compounds in milk are still largely undiscovered.
Collapse
Affiliation(s)
- Emad Yuzbashian
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Emily Berg
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | - Catherine B Chan
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
10
|
Wang S, Hu C, Lin H, Jia X, Hu R, Zheng R, Li M, Xu Y, Xu M, Zheng J, Zhao X, Li Y, Chen L, Zeng T, Ye Z, Shi L, Su Q, Chen Y, Yu X, Yan L, Wang T, Zhao Z, Qin G, Wan Q, Chen G, Dai M, Zhang D, Qiu B, Zhu X, Liu R, Wang X, Tang X, Gao Z, Shen F, Gu X, Luo Z, Qin Y, Chen L, Hou X, Huo Y, Li Q, Wang G, Zhang Y, Liu C, Wang Y, Wu S, Yang T, Deng H, Zhao J, Mu Y, Xu G, Lai S, Li D, Ning G, Wang W, Bi Y, Lu J. Association of circulating long-chain free fatty acids and incident diabetes risk among normoglycemic Chinese adults: a prospective nested case-control study. Am J Clin Nutr 2024; 120:336-346. [PMID: 38729573 DOI: 10.1016/j.ajcnut.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Long-chain free fatty acids (FFAs) are associated with risk of incident diabetes. However, a comprehensive assessment of the associations in normoglycemic populations is lacking. OBJECTIVES Our study aimed to comprehensively investigate the prospective associations and patterns of FFA profiles with diabetes risk among normoglycemic Chinese adults. METHODS This is a prospective nested case-control study from the China Cardiometabolic Disease and Cancer Cohort (4C) study. We quantitatively measured 53 serum FFAs using a targeted metabolomics approach in 1707 incident diabetes subjects and 1707 propensity score-matched normoglycemic controls. Conditional logistic regression models were employed to estimate odds ratios (ORs) for associations. Least Absolute Shrinkage and Selection Operator (LASSO) penalty regression and quantile g-computation (qg-comp) analyses were implemented to estimate the association between multi-FFA exposures and incident diabetes. RESULTS The majority of odd-chain FFAs exhibited an inverse association with incident diabetes, wherein the ORs per SD increment of all 7 saturated fatty acids (SFAs), monounsaturated fatty acid (MUFA) 15:1, and polyunsaturated fatty acid (PUFA) 25:2 were ranging from 0.79 to 0.88 (95% CIs ranging between 0.71 and 0.97). Even-chain FFAs comprised 99.3% of total FFAs and displayed heterogeneity with incident diabetes. SFAs with 18-26 carbon atoms are inversely linked to incident diabetes, with ORs ranging from 0.81 to 0.86 (95% CIs ranging between 0.73 and 0.94). MUFAs 26:1 (OR: 0.85; 95% CI: 0.76, 0.94), PUFAs 20:4 (OR: 0.84; 95% CI: 0.75, 0.94), and 24:2 (OR: 0.87; 95% CI: 0.78, 0.97) demonstrated significant associations. In multi-FFA exposure model, 24 FFAs were significantly associated with incident diabetes, most of which were consistent with univariate results. The mixture OR was 0.78 (95% CI: 0.61, 0.99; P = 0.04159). Differential correlation network analysis revealed pre-existing perturbations in intraclass and interclass FFA coregulation before diabetes onset. CONCLUSIONS These findings underscore the variations in diabetes risk associated with FFAs across chain length and unsaturation degree, highlighting the importance of recognizing FFA subtypes in the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Hu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruying Hu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinjie Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yanli Li
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Lulu Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianshu Zeng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Ye
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Lixin Shi
- Affiliated Hospital of Guiyang Medical College, Guiyang, China
| | - Qing Su
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuefeng Yu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yan
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guijun Qin
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qin Wan
- The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Chen
- Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Meng Dai
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bihan Qiu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xulei Tang
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhengnan Gao
- Dalian Municipal Central Hospital, Dalian, China
| | - Feixia Shen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuejiang Gu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zuojie Luo
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingfen Qin
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Chen
- Qilu Hospital of Shandong University, Jinan, China
| | - Xinguo Hou
- Qilu Hospital of Shandong University, Jinan, China
| | - Yanan Huo
- Jiangxi provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Qiang Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guixia Wang
- The First Hospital of Jilin University, Changchun, China
| | - Yinfei Zhang
- Central Hospital of Shanghai Jiading District, Shanghai, China
| | - Chao Liu
- Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Youmin Wang
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shengli Wu
- Karamay Municipal People's Hospital, Xinjiang, China
| | - Tao Yang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huacong Deng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiajun Zhao
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yiming Mu
- Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guowang Xu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Shenghan Lai
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MA, United States
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Sanjulian L, Lamas A, Barreiro R, Martínez I, García-Alonso L, Cepeda A, Fente C, Regal P. Investigating the Dietary Impact on Trans-Vaccenic Acid (Trans-C18:1 n-7) and Other Beneficial Fatty Acids in Breast Milk and Infant Formulas. Foods 2024; 13:2164. [PMID: 39063248 PMCID: PMC11275335 DOI: 10.3390/foods13142164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Maternal diet plays a significant role in the fatty acid composition of breast milk. Dietary products such as milk and meat are the primary sources of natural TFAs for humans. These peculiar fatty acids hold nutritional significance as they not only lack the detrimental effects of industrially produced trans fats on the endothelium characteristic, but they also exhibit anti-inflammatory properties. The relationship between the presence of eight fatty acids in breast milk (including natural TFAs trans-vaccenic and conjugated linoleic acid) and the maternal diet has been explored, and their abundance has been compared to that of infant formulas. Two cohorts of lactating women, originating from a Spanish region, participated in this study; they adhered to the Southern European Atlantic diet or the Atlantic diet. While the consumption of conventional meat or dairy products does not seem to increase the abundance of TFAs in breast milk, trans-vaccenic and oleic acid are among the most distinctive features of breast milk fat in mothers consuming naturally improved dairy products with an improved fatty acid profile. The most significant differences between natural breastfeeding and formula feeding lie in natural TFAs, since formulas are notably deficient in natural TFAs while being overfortified in alpha-linolenic acid in comparison to breast milk. We suggest an improvement in the formulation of these products through using cow's milk with an optimal fatty acid profile that better mimics the fatty acid composition found in human milk.
Collapse
Affiliation(s)
- Laura Sanjulian
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Medicine, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (A.L.); (R.B.); (A.C.); (P.R.)
| | - Alexandre Lamas
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Medicine, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (A.L.); (R.B.); (A.C.); (P.R.)
| | - Rocío Barreiro
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Medicine, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (A.L.); (R.B.); (A.C.); (P.R.)
| | - Ismael Martínez
- Feiraco Sociedade Cooperativa Galega, Ponte Maceira s/n, 15864 Ames, Spain;
| | | | - Alberto Cepeda
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Medicine, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (A.L.); (R.B.); (A.C.); (P.R.)
| | - Cristina Fente
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Medicine, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (A.L.); (R.B.); (A.C.); (P.R.)
| | - Patricia Regal
- Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Veterinary Medicine, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (L.S.); (A.L.); (R.B.); (A.C.); (P.R.)
| |
Collapse
|
12
|
Venn-Watson S. The Cellular Stability Hypothesis: Evidence of Ferroptosis and Accelerated Aging-Associated Diseases as Newly Identified Nutritional Pentadecanoic Acid (C15:0) Deficiency Syndrome. Metabolites 2024; 14:355. [PMID: 39057678 PMCID: PMC11279173 DOI: 10.3390/metabo14070355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Ferroptosis is a newly discovered form of cell death caused by the peroxidation of fragile fatty acids in cell membranes, which combines with iron to increase reactive oxygen species and disable mitochondria. Ferroptosis has been linked to aging-related conditions, including type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease (NAFLD). Pentadecanoic acid (C15:0), an odd-chain saturated fat, is an essential fatty acid with the primary roles of stabilizing cell membranes and repairing mitochondrial function. By doing so, C15:0 reverses the underpinnings of ferroptosis. Under the proposed "Cellular Stability Hypothesis", evidence is provided to show that cell membranes optimally need >0.4% to 0.64% C15:0 to support long-term health and longevity. A pathophysiology of a newly identified nutritional C15:0 deficiency syndrome ("Cellular Fragility Syndrome") is provided that demonstrates how C15:0 deficiencies (≤0.2% total circulating fatty acids) can increase susceptibilities to ferroptosis, dysmetabolic iron overload syndrome, type 2 diabetes, cardiovascular disease, and NAFLD. Further, evidence is provided that C15:0 supplementation can reverse the described C15:0 deficiency syndrome, including the key components of ferroptosis. Given the declining dietary intake of C15:0, especially among younger generations, there is a need for extensive studies to understand the potential breadth of Cellular Fragility Syndrome across populations.
Collapse
Affiliation(s)
- Stephanie Venn-Watson
- Seraphina Therapeutics Inc., San Diego, CA 92106, USA;
- Epitracker Inc., San Diego, CA 92106, USA
| |
Collapse
|
13
|
Dhillon J, Pandey S, Newman JW, Fiehn O, Ortiz RM. Metabolic Responses to an Acute Glucose Challenge: The Differential Effects of Eight Weeks of Almond vs. Cracker Consumption in Young Adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.19.24307571. [PMID: 38826341 PMCID: PMC11142291 DOI: 10.1101/2024.05.19.24307571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study investigated the dynamic responses to an acute glucose challenge following chronic almond versus cracker consumption for 8 weeks (clinicaltrials.gov ID: NCT03084003). Seventy-three young adults (age: 18-19 years, BMI: 18-41 kg/m2) participated in an 8-week randomized, controlled, parallel-arm intervention and were randomly assigned to consume either almonds (2 oz/d, n=38) or an isocaloric control snack of graham crackers (325 kcal/d, n=35) daily for 8 weeks. Twenty participants from each group underwent a 2-hour oral glucose tolerance test (oGTT) at the end of the 8-week intervention. Metabolite abundances in the oGTT serum samples were quantified using untargeted metabolomics, and targeted analyses for free PUFAs, total fatty acids, oxylipins, and endocannabinoids. Multivariate, univariate, and chemical enrichment analyses were conducted to identify significant metabolic shifts. Findings exhibit a biphasic lipid response distinguished by higher levels of unsaturated triglycerides in the earlier periods of the oGTT followed by lower levels in the latter period in the almond versus cracker group (p-value<0.05, chemical enrichment analyses). Almond (vs. cracker) consumption was also associated with higher AUC120 min of aminomalonate, and oxylipins (p-value<0.05), but lower AUC120 min of L-cystine, N-acetylmannosamine, and isoheptadecanoic acid (p-value<0.05). Additionally, the Matsuda Index in the almond group correlated with AUC120 min of CE 22:6 (r=-0.46; p-value<0.05) and 12,13 DiHOME (r=0.45; p-value<0.05). Almond consumption for 8 weeks leads to dynamic, differential shifts in response to an acute glucose challenge, marked by alterations in lipid and amino acid mediators involved in metabolic and physiological pathways.
Collapse
Affiliation(s)
- Jaapna Dhillon
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia
- Department of Molecular and Cell Biology, University of California, Merced
| | - Saurabh Pandey
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia
- Jaypee University of Information Technology, Waknaghat, India
| | - John W. Newman
- West Coast Metabolomics Center, University of California, Davis
- Department of Nutrition, University of California, Davis
- Obesity and Metabolism Research Unit, USDA Agricultural Research Service Western Human Nutrition Research Center, University of California, Davis
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California, Davis
| | - Rudy M. Ortiz
- Department of Molecular and Cell Biology, University of California, Merced
| |
Collapse
|
14
|
Chen XY, Yang MT, Huang SY, Qiu YZ, Wei W, Jiang CY, Song S, Zhu HL, Lan QY. Concentration and composition of odd-chain fatty acids in phospholipids and triacylglycerols in Chinese human milk throughout lactation. Food Funct 2024; 15:5352-5363. [PMID: 38635214 DOI: 10.1039/d4fo00396a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Human milk represents the gold standard for infant nutrition, with approximately 50% of the energy in human milk derived from lipids. Odd-chain fatty acids (OCFAs) have been recognized as a category of bioactive milk fatty acids in recent research; however, limited data exist on OCFAs in human milk. This study collected human milk samples spanning the postpartum period from 0 to 400 days. Phospholipids containing OCFAs (PL-OCFAs) were determined in 486 human milk samples using hydrophilic liquid chromatography-electrospray ionization-triquadrupole-mass spectrometry. Triacylglycerols containing OCFAs (TAG-OCFAs) were analyzed in 296 human milk samples using ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The average total concentration of PL-OCFA ranged from 30.89 ± 14.27 mg L-1 to 93.48 ± 36.55 mg L-1 during lactation, and the average total TAG-OCFA content ranged from 103.1 ± 147.15 mg L-1 to 965.41 ± 651.67 mg L-1. Despite the lower absolute concentration of PL-OCFA, its relative concentration (8.75%-11.75%) was significantly higher than that of TAG-OCFA (0.37%-1.85%) throughout lactation. PC-OCFA, SM-OCFA and PE-OCFA are major sub-classes of PL-OCFA. Furthermore, C17:0 was the major chain length in both PL-OCFA and TAG-OCFA, followed by C15:0. C17:1 was characteristic of TAG-OCFA, while long-chain fatty acids C19:0, C21:0 and C23:0 were characteristic of PL-OCFA. Our findings highlighted the importance of bioactive lipids in human milk, suggesting that OCFAs could be targeted in future studies in relation to the health and development of infants.
Collapse
Affiliation(s)
- Xiao-Yan Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Meng-Tao Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Si-Yu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Ying-Zhen Qiu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Wei Wei
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chen-Yu Jiang
- Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui-Lian Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Qiu-Ye Lan
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
15
|
Wu S, Luo H, Zhong J, Su M, Lai X, Zhang Z, Zhou Q. Differential Associations of Erythrocyte Membrane Saturated Fatty Acids with Glycemic and Lipid Metabolic Markers in a Chinese Population: A Cross-Sectional Study. Nutrients 2024; 16:1507. [PMID: 38794744 PMCID: PMC11123842 DOI: 10.3390/nu16101507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Mounting evidence indicates a complex link between circulating saturated fatty acids (SFAs) and cardiovascular disease (CVD) risk factors, but research on erythrocyte membrane SFA associations with metabolic markers remains limited. Our study sought to investigate the correlations between erythrocyte membrane SFAs and key metabolic markers within glycemic and lipid metabolism in a Chinese population of 798 residents aged 41 to 71 from Guangzhou. Using gas chromatography-mass spectrometry, we assessed the erythrocyte membrane saturated fatty acid profile and performed multiple linear regression to evaluate the relationship between different SFA subtypes and metabolic markers. Our findings revealed that the odd-chain SFA group (C15:0 + C17:0) exhibited negative associations with fasting blood glucose (FBG), homeostatic model assessment for insulin resistance (HOMA-IR), and triglycerides (TG). Conversely, the very-long-chain SFA group (C20:0 + C22:0 + C23:0 + C24:0) exhibited positive associations with fasting insulins (FINS), HOMA-IR, total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C). Furthermore, there was no evidence supporting an association between the even-chain group (C14:0 + C16:0 + C18:0) and metabolic markers. Our findings suggest that different subtypes of SFAs have diverse effects on glycemic and lipid metabolic markers, with odd-chain SFAs associated with a lower metabolic risk. However, the results concerning the correlations between even-chain SFAs and very-long-chain SFAs with markers of glycemic and lipid metabolism pathways are confusing, highlighting the necessity for further exploration and investigation.
Collapse
Affiliation(s)
- Shixin Wu
- School of Public Health, Guangzhou Medical University, Guangzhou 510180, China; (S.W.); (H.L.)
| | - Huiru Luo
- School of Public Health, Guangzhou Medical University, Guangzhou 510180, China; (S.W.); (H.L.)
| | - Juncheng Zhong
- School of Public Health, Guangzhou Medical University, Guangzhou 510180, China; (S.W.); (H.L.)
| | - Mengyang Su
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China;
| | - Xiaoying Lai
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
| | - Zheqing Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China;
| | - Quan Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510180, China; (S.W.); (H.L.)
| |
Collapse
|
16
|
Bahadoran Z, Mirmiran P, Azizi F. Usual intake of dairy products and the chance of pre-diabetes regression to normal glycemia or progression to type 2 diabetes: a 9-year follow-up. Nutr Diabetes 2024; 14:15. [PMID: 38594262 PMCID: PMC11004158 DOI: 10.1038/s41387-024-00257-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 03/16/2023] [Accepted: 01/09/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND We assessed the possible effect of usual dairy consumption on pre-diabetes (Pre-DM) remission or progression to type 2 diabetes (T2D). METHODS Pre-DM adults (n = 334, mean age of 49.4 years, and 51.5% men) were assessed for dairy intakes (2006-2008) and followed up to 9 years for incidence of T2D or normal glycemia (NG). All biochemical measurements were done at baseline and all subsequent examinations with 3-y follow-up intervals. Multinomial regression models with adjustment of confounding variables were used to estimate odds ratios (OR) and 95% confidence intervals (CIs) of incident T2D and NG for each serving/d dairy consumption. RESULTS The odds of NG was significantly elevated by 69% (OR = 1.69, 95% CI = 1.00-2.86, P = 0.05) per 200 g/d increased high-fat dairy intake, while the amount of total dairy or low-fat dairy was not related to the outcomes. Higher intakes of yogurt were more likely to be associated with an increased odds of NG (OR = 1.82, 95% CI = 1.20-2.74, P = 0.01). Usual intakes of milk, cheese, or cream-butter were not associated to Pre-DM remission or progression to T2D. CONCLUSION Regular dairy consumption may increase the chance of Pre-DM regression to NG.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Mitchell E, Comerford K, Knight M, McKinney K, Lawson Y. A review of dairy food intake for improving health among black adults in the US. J Natl Med Assoc 2024; 116:253-273. [PMID: 38378306 DOI: 10.1016/j.jnma.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/22/2024]
Abstract
The adult life stage encompasses a range of new experiences, opportunities, and responsibilities that impact health and well-being. During this life stage, health disparities continue to increase for Black Americans, with Black adults having a disproportionate burden of obesity, chronic diseases, comorbidities, and worse treatment outcomes compared to their White peers. While many of the underlying factors for these disparities can be linked to longstanding sociopolitical factors such as systemic racism, food insecurity, and poor access to healthcare, there are also several modifiable risk factors that are known to significantly impact health outcomes, such as improving diet quality, increasing physical activity, and not smoking. Of all the modifiable risk factors known to impact health, improving dietary habits is the factor most consistently associated with better outcomes for body weight and chronic disease. Of the major food groups recommended by the 2020-2025 Dietary Guidelines for Americans (DGA) for achieving healthier dietary patterns, dairy foods have a nutrient profile which matches most closely to what Black Americans are inadequately consuming (e.g., vitamin A, vitamin D, calcium, magnesium). However, Black adults tend to consume less than half the recommended daily servings of dairy foods, in part, due to issues with lactose intolerance, making higher intake of dairy foods an ideal target for improving diet quality and health in this population. This review examines the current body of evidence exploring the links between dairy intake, obesity, cardiometabolic disease risk, chronic kidney disease, and the most common types of cancer, with a special focus on health and disparities among Black adults. Overall, the evidence from most systematic reviews and/or meta-analyses published in the last decade on dairy intake and health outcomes has been conducted on White populations and largely excluded research on Black populations. The findings from this extensive body of research indicate that when teamed with an energy-restricted diet, meeting or exceeding the DGA recommended 3 daily servings of dairy foods is associated with better body weight and composition outcomes and lower rates of most common chronic diseases than lower intake (<2 servings per day). In addition to the number of daily servings consumed, the specific types (e.g., milk, yogurt, cheese) and subtypes (e.g., low-fat, fermented, fortified) consumed have also been shown to play major roles in how these foods impact health. For example, higher intake of fermented dairy foods (e.g., yogurt) and vitamin D fortified dairy products appear to have the most protective effects for reducing chronic disease risk. Along with lactose-free milk and cheese, yogurt is also generally low in lactose, making it an excellent option for individuals with lactose intolerance, who are trying to meet the DGA recommendations for dairy food intake.
Collapse
Affiliation(s)
- Edith Mitchell
- Sidney Kimmel Cancer at Jefferson, Philadelphia, PA, United States
| | - Kevin Comerford
- OMNI Nutrition Science, California Dairy Research Foundation, Davis, CA, United States.
| | - Michael Knight
- The George Washington University School of Medicine and Health Sciences, Washington D.C., United States
| | - Kevin McKinney
- University of Texas Medical Branch, Department of Internal Medicine, Division of Endocrinology, Galveston, TX, United States
| | - Yolanda Lawson
- Baylor University Medical Center, Dallas, TX, United States
| |
Collapse
|
18
|
Doumatey AP, Shriner D, Zhou J, Lei L, Chen G, Oluwasola-Taiwo O, Nkem S, Ogundeji A, Adebamowo SN, Bentley AR, Gouveia MH, Meeks KAC, Adebamowo CA, Adeyemo AA, Rotimi CN. Untargeted metabolomic profiling reveals molecular signatures associated with type 2 diabetes in Nigerians. Genome Med 2024; 16:38. [PMID: 38444015 PMCID: PMC10913364 DOI: 10.1186/s13073-024-01308-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) has reached epidemic proportions globally, including in Africa. However, molecular studies to understand the pathophysiology of T2D remain scarce outside Europe and North America. The aims of this study are to use an untargeted metabolomics approach to identify: (a) metabolites that are differentially expressed between individuals with and without T2D and (b) a metabolic signature associated with T2D in a population of Sub-Saharan Africa (SSA). METHODS A total of 580 adult Nigerians from the Africa America Diabetes Mellitus (AADM) study were studied. The discovery study included 310 individuals (210 without T2D, 100 with T2D). Metabolites in plasma were assessed by reverse phase, ultra-performance liquid chromatography and mass spectrometry (RP)/UPLC-MS/MS methods on the Metabolon Platform. Welch's two-sample t-test was used to identify differentially expressed metabolites (DEMs), followed by the construction of a biomarker panel using a random forest (RF) algorithm. The biomarker panel was evaluated in a replication sample of 270 individuals (110 without T2D and 160 with T2D) from the same study. RESULTS Untargeted metabolomic analyses revealed 280 DEMs between individuals with and without T2D. The DEMs predominantly belonged to the lipid (51%, 142/280), amino acid (21%, 59/280), xenobiotics (13%, 35/280), carbohydrate (4%, 10/280) and nucleotide (4%, 10/280) super pathways. At the sub-pathway level, glycolysis, free fatty acid, bile metabolism, and branched chain amino acid catabolism were altered in T2D individuals. A 10-metabolite biomarker panel including glucose, gluconate, mannose, mannonate, 1,5-anhydroglucitol, fructose, fructosyl-lysine, 1-carboxylethylleucine, metformin, and methyl-glucopyranoside predicted T2D with an area under the curve (AUC) of 0.924 (95% CI: 0.845-0.966) and a predicted accuracy of 89.3%. The panel was validated with a similar AUC (0.935, 95% CI 0.906-0.958) in the replication cohort. The 10 metabolites in the biomarker panel correlated significantly with several T2D-related glycemic indices, including Hba1C, insulin resistance (HOMA-IR), and diabetes duration. CONCLUSIONS We demonstrate that metabolomic dysregulation associated with T2D in Nigerians affects multiple processes, including glycolysis, free fatty acid and bile metabolism, and branched chain amino acid catabolism. Our study replicated previous findings in other populations and identified a metabolic signature that could be used as a biomarker panel of T2D risk and glycemic control thus enhancing our knowledge of molecular pathophysiologic changes in T2D. The metabolomics dataset generated in this study represents an invaluable addition to publicly available multi-omics data on understudied African ancestry populations.
Collapse
Affiliation(s)
- Ayo P Doumatey
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA.
| | - Daniel Shriner
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | - Jie Zhou
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | - Lin Lei
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | - Guanjie Chen
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | | | - Susan Nkem
- Center for Bioethics & Research, Ibadan, Nigeria
| | | | - Sally N Adebamowo
- Department of Epidemiology and Public Health, and the Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amy R Bentley
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | - Mateus H Gouveia
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | - Karlijn A C Meeks
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| | - Clement A Adebamowo
- Department of Epidemiology and Public Health, and the Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Adebowale A Adeyemo
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA.
| | - Charles N Rotimi
- Center for Research On Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, 12 South Drive, Building 12 A, Room 1025A, Bethesda, MD, 20892, USA
| |
Collapse
|
19
|
Chopra AK. Dietary management of dyslipidemia. Indian Heart J 2024; 76 Suppl 1:S65-S72. [PMID: 38122980 PMCID: PMC11019336 DOI: 10.1016/j.ihj.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
The rising burden of cardiovascular disease (CVD) has made the achievement of optimal lipoprotein levels a major public health priority. As nearly a fifth of global mortality is associated with dietary factors, and recommendations have been mired in controversy, a fresh look on the available data is attempted. Well established concepts regarding nutrition and cardiometabolic health, role of macronutrients, calories, and controversial foods are discussed followed by recommendations in the Indian context. A healthy dietary pattern rather than individual foods or nutrients is emphasized, and this is generally plant based with optional consumption of dairy, eggs, and meats within the suggested limits. Suggestions/recommendations are given for consumption of individual foods, remembering that choosing appropriate replacement foods is as important as restricting unhealthy foods.
Collapse
|
20
|
Muñoz-Alvarez KY, Gutiérrez-Aguilar R, Frigolet ME. Metabolic effects of milk fatty acids: A literature review. NUTR BULL 2024; 49:19-39. [PMID: 38226553 DOI: 10.1111/nbu.12657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024]
Abstract
Milk and dairy products are known to have a significant role in human development and tissue maintenance due to their high nutritional value. With the higher incidence of obesity and metabolic diseases, nutrition and public health authorities have recommended the intake of fat-free or low-fat dairy due to the saturated fatty acid content of whole-fat products and their effect on serum cholesterol levels. However, recent studies have questioned the association between milk fat consumption and cardiometabolic risk. This literature review aims to compile the scientific evidence of the metabolic effects of milk fatty acids in clinical and basic research studies, as well as their relationship with metabolic disorders and gut microbiota composition. Research shows that various milk fatty acids exert effects on metabolic alterations (obesity, type 2 diabetes and cardiovascular diseases) by modifying glucose homeostasis, inflammation and lipid profile-related factors. Additionally, recent studies have associated the consumption of milk fatty acids with the production of metabolites and the promotion of healthy gut microbiota. From mainly observational studies, evidence suggests that milk and dairy fatty acids are not directly linked to cardiometabolic risk, but further controlled research is necessary to clarify such findings and to assess whether dietary recommendations to choose low-fat dairy foods are necessary for the population for the prevention of obesity and cardiometabolic disease.
Collapse
Affiliation(s)
- Karla Y Muñoz-Alvarez
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
| | - Ruth Gutiérrez-Aguilar
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - María E Frigolet
- Laboratorio de Investigación en Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México 'Federico Gómez' (HIMFG), Mexico City, Mexico
| |
Collapse
|
21
|
Cetin E, Pedersen B, Porter LM, Adler GK, Burak MF. Protocol for a randomized placebo-controlled clinical trial using pure palmitoleic acid to ameliorate insulin resistance and lipogenesis in overweight and obese subjects with prediabetes. Front Endocrinol (Lausanne) 2024; 14:1306528. [PMID: 38313838 PMCID: PMC10835623 DOI: 10.3389/fendo.2023.1306528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
Palmitoleic acid (POA), a nonessential, monounsaturated omega-7 fatty acid (C16:1n7), is a lipid hormone secreted from adipose tissue and has beneficial effects on distant organs, such as the liver and muscle. Interestingly, POA decreases lipogenesis in toxic storage sites such as the liver and muscle, and paradoxically increases lipogenesis in safe storage sites, such as adipose tissue. Furthermore, higher POA levels in humans are correlated with better insulin sensitivity, an improved lipid profile, and a lower incidence of type-2 diabetes and cardiovascular pathologies, such as myocardial infarction. In preclinical animal models, POA improves glucose intolerance, dyslipidemia, and steatosis of the muscle and liver, while improving insulin sensitivity and secretion. This double-blind placebo-controlled clinical trial tests the hypothesis that POA increases insulin sensitivity and decreases hepatic lipogenesis in overweight and obese adult subjects with pre-diabetes. Important to note, that this is the first study ever to use pure (>90%) POA with < 0.3% palmitic acid (PA), which masks the beneficial effects of POA. The possible positive findings may offer a therapeutic and/or preventative pathway against diabetes and related immunometabolic diseases.
Collapse
Affiliation(s)
- Ecesu Cetin
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Brian Pedersen
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Lindsey M. Porter
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Gail K. Adler
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Mehmet Furkan Burak
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Sabri Ulker Center, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| |
Collapse
|
22
|
Retterstøl K, Rosqvist F. Fat and fatty acids - a scoping review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2024; 68:9980. [PMID: 38327998 PMCID: PMC10845901 DOI: 10.29219/fnr.v68.9980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/05/2023] [Accepted: 08/30/2023] [Indexed: 02/09/2024] Open
Abstract
Two de novo NNR2022 systematic reviews (SRs) as well as 21 qualified SRs (qSRs) were available. A literature search yielded an additional ~70 SRs, meta-analyses and biomarker papers. Diets lower in total fat are associated with reductions in body weight and blood pressure compared with diets higher in total fat in adults. Partial replacement of saturated fatty acid (SFA) with n-6 polyunsaturated fatty acid (PUFA) improves blood lipid profile, decreases the risk of cardiovascular disease (CVD), improves glucose-insulin homeostasis and may decrease the risk of total mortality. Long-chain n-3 PUFAs (eicosapentaenoic acid [EPA] and docosahexaenoic acid [DHA]) decrease triglycerides and are associated with lower risk of CVD. Dietary PUFAs, both n-3 and n-6, may be associated with reduced risk of type 2 diabetes (T2D). There is inconclusive evidence to suggest that the type of dietary fat is associated with blood pressure, risk of hypertension or musculoskeletal health. Higher intake of total PUFA is associated with lower mortality from any cancer. Long-chain n-3 PUFA is associated with reduced risk of breast cancer, whereas biomarker levels of n-6 PUFA are associated with lower risk of any cancer. Intake of long-chain n-3 PUFA during pregnancy increases length of gestation and child birth weight and reduces the risk of preterm delivery, but there is inconclusive evidence to suggest that it may influence child neurodevelopment, growth or development of allergic disease. In studies with higher versus lower dietary cholesterol intake levels, total blood cholesterol increased or were unaffected by the dietary cholesterol, resulting in inconclusive results. Trans fatty acid (TFA), regardless of source, impairs blood lipid profile compared to unsaturated fat. In observational studies, TFA is positively associated with CVD and total mortality but whether associations differ by source is inconclusive. Ruminant TFA, as well as biomarker levels of odd-chain fatty acids, might be associated with lower risk of T2D.
Collapse
Affiliation(s)
- Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Trichia E, Koulman A, Stewart ID, Brage S, Griffin SJ, Griffin JL, Khaw K, Langenberg C, Wareham NJ, Imamura F, Forouhi NG. Plasma Metabolites Related to the Consumption of Different Types of Dairy Products and Their Association with New-Onset Type 2 Diabetes: Analyses in the Fenland and EPIC-Norfolk Studies, United Kingdom. Mol Nutr Food Res 2024; 68:e2300154. [PMID: 38054622 PMCID: PMC10909549 DOI: 10.1002/mnfr.202300154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/07/2023] [Indexed: 12/07/2023]
Abstract
SCOPE To identify metabolites associated with habitual dairy consumption and investigate their associations with type 2 diabetes (T2D) risk. METHODS AND RESULTS Metabolomics assays were conducted in the Fenland (n = 10,281) and EPIC-Norfolk (n = 1,440) studies. Using 82 metabolites assessed in both studies, we developed metabolite scores to classify self-reported consumption of milk, yogurt, cheese, butter, and total dairy (Fenland Study-discovery set; n = 6035). Internal and external validity of the scores was evaluated (Fenland-validation set, n = 4246; EPIC-Norfolk, n = 1440). The study assessed associations between each metabolite score and T2D incidence in EPIC-Norfolk (n = 641 cases; 16,350 person-years). The scores classified low and high consumers for all dairy types with internal validity, and milk, butter, and total dairy with external validity. The scores were further associated with lower incident T2D: hazard ratios (95% confidence interval) per standard deviation: milk 0.71 (0.65, 0.77); butter 0.62 (0.57, 0.68); total dairy 0.66 (0.60, 0.72). These associations persisted after adjustment for known dairy-fat biomarkers. CONCLUSION Metabolite scores identified habitual consumers of milk, butter, and total dairy products, and were associated with lower T2D risk. These findings hold promise for identifying objective indicators of the physiological response to dairy consumption.
Collapse
Affiliation(s)
- Eirini Trichia
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Albert Koulman
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Isobel D. Stewart
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Soren Brage
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Simon J. Griffin
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | | | - Kay‐Tee Khaw
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Claudia Langenberg
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Nicholas J. Wareham
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Fumiaki Imamura
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| | - Nita G. Forouhi
- MRC Epidemiology UnitInstitute of Metabolic ScienceUniversity of Cambridge School of Clinical MedicineCambridgeCB2 0SLUK
| |
Collapse
|
24
|
Roy BC, Coleman P, Markowsky M, Wang K, She Y, Richard C, Proctor SD, Bruce HL. Muscle Fiber, Connective Tissue and Meat Quality Characteristics of Pork from Low Birth Weight Pigs as Affected by Diet-Induced Increased Fat Absorption and Preferential Muscle Marbling. Food Sci Anim Resour 2024; 44:51-73. [PMID: 38229859 PMCID: PMC10789550 DOI: 10.5851/kosfa.2023.e56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 01/18/2024] Open
Abstract
This study investigated how birth weight differences in piglets affected carcass and muscle fiber properties as well as meat quality at slaughter. Within litters, piglets were grouped according to their birth weight as either normal (NBW; 1.62-1.73 kg) or low (LBW; 1.18-1.29 kg). At 5 weeks of age, NBW piglets were randomly transitioned to control (C) or isocaloric high fat diets derived from non-dairy (HF), while LBW piglets were randomly transitioned to high fat diets derived from non-dairy (HF) or dairy sources (HFHD). Piglets were reared in individual pens under standardized housing and feeding conditions. Live weight was recorded weekly, and pigs were slaughtered at 12 weeks of age. Hot carcass weights, dressing percentages, lean meat yield, and primal cut proportions were determined. The m. longissimus thoracis was collected from the right side of the carcass for measurement of physical and chemical properties of meat and muscle fiber characteristics. Results indicated that LBW pigs compensated for their live weight compared to NBW pigs at 6 weeks of age. The mean muscle fiber diameter of LBW-HFHD group is significantly higher than NBW-C and NBW-HF group, and the type I muscle fiber diameter is significantly higher than NBW-C group. Dairy fat inclusion in LBW pig diet reduced carcass back fat thickness. This increased the calculated lean meat yield to be comparable to that of NBW pigs fed a commercial diet. Incorporating dairy-sourced high-fat into LBW pigs' diets appears to be an effective strategy for producing carcasses equivalent to NBW pigs.
Collapse
Affiliation(s)
- Bimol C. Roy
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Patience Coleman
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Meghan Markowsky
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Kun Wang
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Yongbo She
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Caroline Richard
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Spencer D. Proctor
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
- Division of Human Nutrition, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| | - Heather L. Bruce
- Division of Animal Science, Department of
Agricultural, Food and Nutritional Science, University of
Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
25
|
Khajeh pour S, Blanton C, Ghimire B, Aghazadeh‐Habashi A. Development of a rapid, sensitive, and selective LC-MS/MS method for quantifying curcumin levels in healthy human urine: Effect of pepper on curcumin bioavailability. Food Sci Nutr 2023; 11:7732-7741. [PMID: 38107126 PMCID: PMC10724617 DOI: 10.1002/fsn3.3691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 12/19/2023] Open
Abstract
Curcumin (CCM), a culinary spice, is widely consumed for its health benefits for managing oxidative and inflammatory conditions, metabolic syndrome, arthritis, and hyperlipidemia. However, due to its extensive metabolism, the oral bioavailability of CCM is very low. In this study, we developed a rapid, sensitive, and selective assay to examine the hypothesis that piperine improves CCM bioavailability after piperine co-ingestion. We developed a selective, sensitive, and robust LC-MS/MS method to quantify CCM in human urine. The method was linear over a concentration range 0.625-40 ng/mL with LLOQ and LLOD of 0.625 ng/mL and 0.312 ng/mL, respectively. Healthy volunteers have consumed test meals of CCM as turmeric powder with and without black pepper with 1 week wash out. Urine samples were collected for 24 hours and analyzed for CCM excretion. Black pepper increased CCM half-life from 2.2 ± 0.79 h (CCM alone) to 4.5 ± 0.80 h (CCM + pepper). The CCM 24-h urinary excreted amount was higher in individuals consuming CCM + pepper (218.14 ± 94.98 μg) than those who received CCM only (49.45 ± 12.94 μg). This preliminary study indicates that piperine significantly increased CCM oral absorption, reduced systemic clearance, and improved bioavailability.
Collapse
Affiliation(s)
- Sana Khajeh pour
- Department of Biomedical and Pharmaceutical SciencesIdaho State UniversityPocatelloIdahoUSA
| | - Cynthia Blanton
- Department of Nutrition and DieteticsIdaho State UniversityPocatelloIdahoUSA
| | - Biwash Ghimire
- Department of Biomedical and Pharmaceutical SciencesIdaho State UniversityPocatelloIdahoUSA
| | - Ali Aghazadeh‐Habashi
- Department of Biomedical and Pharmaceutical SciencesIdaho State UniversityPocatelloIdahoUSA
| |
Collapse
|
26
|
Venn-Watson S, Schork NJ. Pentadecanoic Acid (C15:0), an Essential Fatty Acid, Shares Clinically Relevant Cell-Based Activities with Leading Longevity-Enhancing Compounds. Nutrients 2023; 15:4607. [PMID: 37960259 PMCID: PMC10649853 DOI: 10.3390/nu15214607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/24/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Pentadecanoic acid (C15:0) is an essential odd-chain saturated fatty acid with broad activities relevant to protecting cardiometabolic, immune, and liver health. C15:0 activates AMPK and inhibits mTOR, both of which are core components of the human longevity pathway. To assess the potential for C15:0 to enhance processes associated with longevity and healthspan, we used human cell-based molecular phenotyping assays to compare C15:0 with three longevity-enhancing candidates: acarbose, metformin, and rapamycin. C15:0 (n = 36 activities in 10 of 12 cell systems) and rapamycin (n = 32 activities in 12 of 12 systems) had the most clinically relevant, dose-dependent activities. At their optimal doses, C15:0 (17 µM) and rapamycin (9 µM) shared 24 activities across 10 cell systems, including anti-inflammatory (e.g., lowered MCP-1, TNFα, IL-10, IL-17A/F), antifibrotic, and anticancer activities, which are further supported by previously published in vitro and in vivo studies. Paired with prior demonstrated abilities for C15:0 to target longevity pathways, hallmarks of aging, aging rate biomarkers, and core components of type 2 diabetes, heart disease, cancer, and nonalcoholic fatty liver disease, our results support C15:0 as an essential nutrient with activities equivalent to, or surpassing, leading longevity-enhancing candidate compounds.
Collapse
Affiliation(s)
- Stephanie Venn-Watson
- Epitracker Inc., San Diego, CA 92106, USA
- Seraphina Therapeutics, Inc., San Diego, CA 92106, USA;
| | - Nicholas J. Schork
- Seraphina Therapeutics, Inc., San Diego, CA 92106, USA;
- Translational Genomics Research Institute (TGen), City of Hope, Phoenix, AZ 85004, USA
| |
Collapse
|
27
|
Bernard L, Chen J, Kim H, Huang Z, Bazzano L, Qi L, He J, Rao VS, Potts KS, Kelly TN, Wong KE, Steffen LM, Yu B, Rhee EP, Rebholz CM. Serum Metabolomic Markers of Dairy Consumption: Results from the Atherosclerosis Risk in Communities Study and the Bogalusa Heart Study. J Nutr 2023; 153:2994-3002. [PMID: 37541543 PMCID: PMC10613758 DOI: 10.1016/j.tjnut.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/14/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Dairy consumption is related to chronic disease risk; however, the measurement of dairy consumption has largely relied upon self-report. Untargeted metabolomics allows for the identification of objective markers of dietary intake. OBJECTIVES We aimed to identify associations between dietary dairy intake (total dairy, low-fat dairy, and high-fat dairy) and serum metabolites in 2 independent study populations of United States adults. METHODS Dietary intake was assessed with food frequency questionnaires. Multivariable linear regression models were used to estimate cross-sectional associations between dietary intake of dairy and 360 serum metabolites analyzed in 2 subgroups of the Atherosclerosis Risk in Communities study (ARIC; n = 3776). Results from the 2 subgroups were meta-analyzed using fixed effects meta-analysis. Significant meta-analyzed associations in the ARIC study were then tested in the Bogalusa Heart Study (BHS; n = 785). RESULTS In the ARIC study and BHS, the mean age was 54 and 48 years, 61% and 29% were Black, and the mean dairy intake was 1.7 and 1.3 servings/day, respectively. Twenty-nine significant associations between dietary intake of dairy and serum metabolites were identified in the ARIC study (total dairy, n = 14; low-fat dairy, n = 10; high-fat dairy, n = 5). Three associations were also significant in BHS: myristate (14:0) was associated with high-fat dairy, and pantothenate was associated with total dairy and low-fat dairy, but 23 of the 27 associations significant in the ARIC study and tested in BHS were not associated with dairy in BHS. CONCLUSIONS We identified metabolomic associations with dietary intake of dairy, including 3 associations found in 2 independent cohort studies. These results suggest that myristate (14:0) and pantothenate (vitamin B5) are candidate biomarkers of dairy consumption.
Collapse
Affiliation(s)
- Lauren Bernard
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Lu Qi
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Varun S Rao
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Kaitlin S Potts
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, United States
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States; Division of Nephrology, Department of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Kari E Wong
- Metabolon, Research Triangle Park, Morrisville, NC, United States
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, United States
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston School of Public Health, Houston, TX, United States
| | - Eugene P Rhee
- Division of Nephrology and Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
28
|
Li A, Han X, Liu L, Zhang G, Du P, Zhang C, Li C, Chen B. Dairy products and constituents: a review of their effects on obesity and related metabolic diseases. Crit Rev Food Sci Nutr 2023:1-21. [PMID: 37724572 DOI: 10.1080/10408398.2023.2257782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Obesity has become a global public health problem that seriously affects the quality of life. As an important part of human diet, dairy products contain a large number of nutrients that are essential for maintaining human health, such as proteins, peptides, lipids, vitamins, and minerals. A growing number of epidemiological investigations provide strong evidence on dairy interventions for weight loss in overweight/obese populations. Therefore, this paper outlines the relationship between the consumption of different dairy products and obesity and related metabolic diseases. In addition, we dive into the mechanisms related to the regulation of glucose and lipid metabolism by functional components in dairy products and the interaction with gut microbes. Lastly, the role of dairy products on obesity of children and adolescents is revisited. We conclude that whole dairy products exert more beneficial effect than single milk constituent on alleviating obesity and that dairy matrix has important implications for metabolic health.
Collapse
Affiliation(s)
- Aili Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Xueting Han
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Libo Liu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Guofang Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Peng Du
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Chao Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
| | - Chun Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin, China
- Heilongjiang Green Food Research Institute, Harbin, China
| | - Bingcan Chen
- Department of Plant Sciences, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|
29
|
Nestel PJ, Mori TA. Dairy Foods: Beneficial Effects of Fermented Products on Cardiometabolic Health. Curr Nutr Rep 2023; 12:478-485. [PMID: 37226031 DOI: 10.1007/s13668-023-00476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2023] [Indexed: 05/26/2023]
Abstract
PURPOSE OF REVIEW This review of recent observational studies and understanding of the complex nature of dairy foods is intended to reappraise the effects of different types of dairy foods on cardiovascular disease (CVD). RECENT FINDINGS Recent guidelines from major cardiovascular societies suggest that apart from the adverse effect of butter, consumption of more complex dairy products notably fermented varieties and yogurt in particular, appear to be inversely associated with outcomes of CVD and type 2 diabetes (T2D). Reduced fat in dairy food remains preferred for people at increased CVD risk. Changed evidence has led to new advice regarding consumption of some dairy foods. The apparent beneficial effects of fermented milk products, particularly yogurt, allow for increased consumption of nutritious staple foods. Recent national guidelines reflect this view.
Collapse
Affiliation(s)
- Paul J Nestel
- Baker Heart and Diabetes Institute, 75 Commercial Rd, Melbourne, 3004, Australia.
| | - Trevor A Mori
- Medical School, University of Western Australia, Perth, Australia
| |
Collapse
|
30
|
Givens DI. Dairy foods and cardiometabolic diseases: an update and a reassessment of the impact of SFA. Proc Nutr Soc 2023; 82:329-345. [PMID: 36740241 DOI: 10.1017/s0029665123000083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes (T2D) and CVD are major causes of mortality and chronic morbidity. Whilst mortality from CVD has decreased they remain the largest cause of death in Europe and the prevalence of T2D is increasing rapidly. A consistent component of public health advice is to reduce intake of SFA to reduce CVD in particular, which implies limiting dairy food consumption. The prospective studies and randomised controlled trials included in this review show that for dairy foods at least, SFA are not consistently associated with CVD or T2D risk. For CVD the association with dairy foods is generally neutral despite dairy foods being the major source of SFA in many diets. This creates considerable doubt, at least for dairy foods, concerning the validity of the traditional diet-heart hypothesis which positively relates SFA intake to increased serum LDL-cholesterol and subsequent increased CVD. There is now emerging evidence to explain this which is highly relevant to dairy foods. These include the potentially counterbalancing effect of SFA-stimulated HDL-cholesterol and specific food matrix factors. In addition, SFA are associated with the less atherogenic large buoyant LDL particles and possible counterbalancing hypotensive effects of dairy proteins. Overall, dairy foods have either a neutral or beneficial association with CVD and T2D. Beneficial associations are seen for blood pressure and the reduced T2D risk linked to yoghurt consumption, a subject that needs urgent attention given the sharp rise in T2D prevalence in many countries.
Collapse
Affiliation(s)
- D Ian Givens
- Institute for Food, Nutrition and Health, University of Reading, Reading RG6 6EU, UK
| |
Collapse
|
31
|
Okami H, Kawaharada R, Yoshizaki H, Toriumi A, Tsutsumi S, Nakamura A. Maternal n-7 Unsaturated Fatty Acids Protect the Fetal Brain from Neuronal Degeneration in an Intrauterine Hyperglycemic Animal Model. Nutrients 2023; 15:3434. [PMID: 37571372 PMCID: PMC10421171 DOI: 10.3390/nu15153434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
We previously reported that glycation induces insulin resistance in the hearts of newborn pups from a gestational diabetes mellitus (GDM) rat model. Administration of n-3 unsaturated fatty acids suppressed glycation and improved signaling in GDM rat pups. In this study, we investigated their effects on cranial neurons using the GDM rat model and PC12 cells derived from rat adrenal pheochromocytomas. Additionally, we examined whether n-3 and n-7 unsaturated fatty acids (cis-palmitoleic acid [CPA] and trans-palmitoleic acid [TPA]) ameliorate the detrimental effects of high glucose exposure on rats. In the neonatal cerebrum of GDM rats, increased levels of advanced glycation end products (AGEs) inhibited Akt phosphorylation; however, CPA and TPA intake during pregnancy ameliorated these abnormalities. Furthermore, exposure to high-glucose-induced apoptosis in PC12 cells compared to the cells cultured in control glucose. PC12 cells exposed to high-glucose with fatty acids exhibited reduced AGE production and apoptosis induction compared to the high-glucose group. These findings suggest that a hyperglycemic environment during pregnancy promotes AGE formation in brain neuronal proteins and induces apoptosis. Both TPA and CPA mitigated these abnormalities; however, CPA is cytotoxic, highlighting its safety in pregnant women.
Collapse
Affiliation(s)
- Haruka Okami
- Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan;
| | - Ritsuko Kawaharada
- Department of Health and Nutrition, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan;
| | - Hitomi Yoshizaki
- Department of Bioregulatory Science (Physiology), Nippon Medical School, Tokyo 113-8602, Japan;
| | - Akiyo Toriumi
- Department of Public Health, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan;
| | - Saki Tsutsumi
- Department of Neurophysiology & Neural Repair, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan;
| | - Akio Nakamura
- Department of Molecular Nutrition, Faculty of Human Life Sciences, Jissen Women’s University, Hino 191-8510, Japan
| |
Collapse
|
32
|
Lampousi AM, Carlsson S, Löfvenborg JE, Cabrera-Castro N, Chirlaque MD, Fagherazzi G, Franks PW, Hampe CS, Jakszyn P, Koulman A, Kyrø C, Moreno-Iribas C, Nilsson PM, Panico S, Papier K, van der Schouw YT, Schulze MB, Weiderpass E, Zamora-Ros R, Forouhi NG, Sharp SJ, Rolandsson O, Wareham NJ. Interaction between plasma phospholipid odd-chain fatty acids and GAD65 autoantibodies on the incidence of adult-onset diabetes: the EPIC-InterAct case-cohort study. Diabetologia 2023; 66:1460-1471. [PMID: 37301794 PMCID: PMC10317878 DOI: 10.1007/s00125-023-05948-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/25/2023] [Indexed: 06/12/2023]
Abstract
AIMS/HYPOTHESIS Islet autoimmunity may progress to adult-onset diabetes. We investigated whether circulating odd-chain fatty acids (OCFA) 15:0 and 17:0, which are inversely associated with type 2 diabetes, interact with autoantibodies against GAD65 (GAD65Ab) on the incidence of adult-onset diabetes. METHODS We used the European EPIC-InterAct case-cohort study including 11,124 incident adult-onset diabetes cases and a subcohort of 14,866 randomly selected individuals. Adjusted Prentice-weighted Cox regression estimated HRs and 95% CIs of diabetes in relation to 1 SD lower plasma phospholipid 15:0 and/or 17:0 concentrations or their main contributor, dairy intake, among GAD65Ab-negative and -positive individuals. Interactions between tertiles of OCFA and GAD65Ab status were estimated by proportion attributable to interaction (AP). RESULTS Low concentrations of OCFA, particularly 17:0, were associated with a higher incidence of adult-onset diabetes in both GAD65Ab-negative (HR 1.55 [95% CI 1.48, 1.64]) and GAD65Ab-positive (HR 1.69 [95% CI 1.34, 2.13]) individuals. The combination of low 17:0 and high GAD65Ab positivity vs high 17:0 and GAD65Ab negativity conferred an HR of 7.51 (95% CI 4.83, 11.69), with evidence of additive interaction (AP 0.25 [95% CI 0.05, 0.45]). Low dairy intake was not associated with diabetes incidence in either GAD65Ab-negative (HR 0.98 [95% CI 0.94, 1.02]) or GAD65Ab-positive individuals (HR 0.97 [95% CI 0.79, 1.18]). CONCLUSIONS/INTERPRETATION Low plasma phospholipid 17:0 concentrations may promote the progression from GAD65Ab positivity to adult-onset diabetes.
Collapse
Affiliation(s)
- Anna-Maria Lampousi
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Sofia Carlsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josefin E Löfvenborg
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Risk and Benefit Assessment, Swedish Food Agency, Uppsala, Sweden
| | | | - María-Dolores Chirlaque
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Health and Social Sciences, Murcia University, Murcia, Spain
| | - Guy Fagherazzi
- Deep Digital Phenotyping Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Paul W Franks
- Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital, Lund University, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
| | - Christiane S Hampe
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Paula Jakszyn
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
- Blanquerna School of Health Sciences, Ramon Llull University, Barcelona, Spain
| | - Albert Koulman
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- National Institute for Health Research Biomedical Research Centre Core Nutritional Biomarker Laboratory, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Cecilie Kyrø
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Conchi Moreno-Iribas
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Peter M Nilsson
- Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Salvatore Panico
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Keren Papier
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - Nita G Forouhi
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Stephen J Sharp
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Family Medicine, Umeå University, Umeå, Sweden
| | - Nicholas J Wareham
- Medical Research Council Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
33
|
Mozaffarian D. Identifying nutritional priorities for global health: time for a more PURE focus on protective foods. Eur Heart J 2023; 44:2580-2582. [PMID: 37414412 PMCID: PMC10361008 DOI: 10.1093/eurheartj/ehad325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2023] Open
Affiliation(s)
- Dariush Mozaffarian
- Friedman School of Nutrition Science & Policy, Tufts University, 150 Harrison Ave, Boston, MA 02111, USA
| |
Collapse
|
34
|
Yehia NA, Isai L, Semnani-Azad Z, Lai KZH, Retnakaran R, Harris SB, Beaudry JL, Bazinet RP, Hanley AJ. Association of circulating branched chain fatty acids with insulin sensitivity and beta cell function in the PROMISE cohort. Lipids 2023; 58:171-183. [PMID: 37165723 DOI: 10.1002/lipd.12373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/29/2023] [Accepted: 04/17/2023] [Indexed: 05/12/2023]
Abstract
Branched chain fatty acids (BCFAs) are mainly saturated fatty acids with a methyl branch on the penultimate or antepenultimate carbon atom. While BCFAs are endogenously produced via the catabolism of branched chain amino acids, the primary exogenous source of BCFAs in the human body is via the diet, including dairy products. Recently, BCFAs have been identified as having a potentially protective role in the etiology of cardiometabolic disorders although current literature is limited. We aimed to investigate the longitudinal associations of circulating BCFAs across four serum pools with insulin sensitivity, beta cell function, and glucose concentrations in the PROMISE Cohort. Estimates of insulin sensitivity were assessed using Matsuda's insulin sensitivity index (ISI) and the homeostasis model assessment of insulin sensitivity (HOMA2). Estimates of beta cell function were determined using the insulinogenic index divided by HOMA insulin resistance and the insulin secretion-sensitivity index-2 (ISSI-2). Baseline serum samples were analyzed for BCFAs using gas-chromatography flame ionization detection. Longitudinal associations were determined using generalized estimating equations. In the free fatty acid (FFA) pool, iso15:0 and anteiso15:0 were positively associated with logHOMA2 (iso15:0 logHOMA2-%S: β = 6.86, 95% CI: [1.64, 12.36], p < 0.05, anteiso15:0 logHOMA2-%S: β = 6.36, 95% CI: [0.63, 12.42], p < 0.05) while anteiso14:0 was inversely associated with measures of insulin sensitivity (iso14:0 logHOMA2-%S: β = -2.35, 95% CI: [-4.26, -0.40], p < 0.05, logISI: β = -2.30, 95% CI: [-4.32, -0.23], p < 0.05, anteiso14:0 logHOMA2-%S: β = -4.72, 95% CI: [-7.81, -1.52], p < 0.05, logISI: β = -6.13, 95% CI: [-9.49, -2.66], p < 0.01). Associations in other pools were less consistent. We identified the potential importance of specific BCFAs, specifically iso14:0, anteiso14:0, iso15:0, anteiso15:0, in cardiometabolic phenotypes underlying type 2 diabetes.
Collapse
Affiliation(s)
- Nagam A Yehia
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Liridona Isai
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhila Semnani-Azad
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kira Zhi Hua Lai
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ravi Retnakaran
- Division of Endocrinology and Metabolism, University of Toronto, Toronto, Ontario, Canada
- Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Stewart B Harris
- Department of Family Medicine, Western University, London, Canada
| | - Jacqueline L Beaudry
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anthony J Hanley
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, University of Toronto, Toronto, Ontario, Canada
- Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Family Medicine, Western University, London, Canada
| |
Collapse
|
35
|
Bishop CA, Machate T, Henkel J, Schulze MB, Klaus S, Piepelow K. Heptadecanoic Acid Is Not a Key Mediator in the Prevention of Diet-Induced Hepatic Steatosis and Insulin Resistance in Mice. Nutrients 2023; 15:2052. [PMID: 37432205 DOI: 10.3390/nu15092052] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 07/12/2023] Open
Abstract
Epidemiological studies found that the intake of dairy products is associated with an increased amount of circulating odd-chain fatty acids (OCFA, C15:0 and C17:0) in humans and further indicate that especially C17:0 is associated with a lower incidence of type 2 diabetes. However, causal relationships are not elucidated. To provide a mechanistic link, mice were fed high-fat (HF) diets supplemented with either milk fat or C17:0 for 20 weeks. Cultured primary mouse hepatocytes were used to distinguish differential effects mediated by C15:0 or C17:0. Despite an induction of OCFA after both dietary interventions, neither long-term milk fat intake nor C17:0 supplementation improved diet-induced hepatic lipid accumulation and insulin resistance in mice. HF feeding with milk fat actually deteriorates liver inflammation. Treatment of primary hepatocytes with C15:0 and C17:0 suppressed JAK2/STAT3 signaling, but only C15:0 enhanced insulin-stimulated phosphorylation of AKT. Overall, the data indicate that the intake of milk fat and C17:0 do not mediate health benefits, whereas C15:0 might be promising in further studies.
Collapse
Affiliation(s)
- Christopher A Bishop
- Department Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
| | - Tina Machate
- Department Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
| | - Janin Henkel
- Department of Nutritional Biochemistry, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, 95326 Kulmbach, Germany
- Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Matthias B Schulze
- Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
- Department Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
| | - Susanne Klaus
- Department Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Karolin Piepelow
- Department Physiology of Energy Metabolism, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
| |
Collapse
|
36
|
Kwon C, Ediriweera MK, Kim Cho S. Interplay between Phytochemicals and the Colonic Microbiota. Nutrients 2023; 15:nu15081989. [PMID: 37111207 PMCID: PMC10145007 DOI: 10.3390/nu15081989] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/08/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Phytochemicals are natural compounds found in food ingredients with a variety of health-promoting properties. Phytochemicals improve host health through their direct systematic absorption into the circulation and modulation of the gut microbiota. The gut microbiota increases the bioactivity of phytochemicals and is a symbiotic partner whose composition and/or diversity is altered by phytochemicals and affects host health. In this review, the interactions of phytochemicals with the gut microbiota and their impact on human diseases are reviewed. We describe the role of intestinal microbial metabolites, including short-chain fatty acids, amino acid derivatives, and vitamins, from a therapeutic perspective. Next, phytochemical metabolites produced by the gut microbiota and the therapeutic effect of some selected metabolites are reviewed. Many phytochemicals are degraded by enzymes unique to the gut microbiota and act as signaling molecules in antioxidant, anti-inflammatory, anticancer, and metabolic pathways. Phytochemicals can ameliorate diseases by altering the composition and/or diversity of the gut microbiota, and they increase the abundance of some gut microbiota that produce beneficial substances. We also discuss the importance of investigating the interactions between phytochemicals and gut microbiota in controlled human studies.
Collapse
Affiliation(s)
- Chohee Kwon
- Department of Environmental Biotechnology, Graduate School of Industry, Jeju National University, Jeju 63243, Republic of Korea
| | - Meran Keshawa Ediriweera
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Colombo, Colombo 008, Sri Lanka
| | - Somi Kim Cho
- Department of Environmental Biotechnology, Graduate School of Industry, Jeju National University, Jeju 63243, Republic of Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
37
|
Harlan TS, Gow RV, Kornstädt A, Alderson PW, Lustig RH. The Metabolic Matrix: Re-engineering ultraprocessed foods to feed the gut, protect the liver, and support the brain. Front Nutr 2023; 10:1098453. [PMID: 37063330 PMCID: PMC10097968 DOI: 10.3389/fnut.2023.1098453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
Ultraprocessed food is established as a metabolic disruptor acting to increase adiposity, reduce mitochondrial efficiency, drive insulin resistance, alter growth, and contribute to human morbidity and mortality. Consumer packaged goods (CPG) companies are beginning to understand the detrimental impact of the food they market, and have employed substitution strategies to reduce salt, sugar, and fat. However, the harms of ultraprocessed foods are far more complex than any single component, and are not ameliorated by such simple substitutions. Over the past 2 years, the authors have worked with the Kuwaiti Danish Dairy Company (KDD) to conduct a comprehensive scientific evaluation of their entire commercial food and beverage portfolio. Assay of the macronutrients, micronutrients, additives, and toxins contained in each of their products was undertaken to determine the precise nature of each product's ingredients as well as the health impacts of processing. The authors formed a Scientific Advisory Team (SAT) and developed a tiered "Metabolic Matrix" founded in three science-based principles: (1) protect the liver, (2) feed the gut, and (3) support the brain. The Metabolic Matrix categorizes each product and provides the criteria, metrics, and recommendations for improvement or reformulation. Real-time consultation with the KDD Executive and Operations teams was vital to see these procedures through to fruition. This scientific exercise has enabled KDD to lay the groundwork for improving the health, well-being, and sustainability of their entire product line, while maintaining flavor, economic, and fiscal viability. This process is easily transferrable, and we are sharing this effort and its approaches as a proof-of-concept. The key aim of our work is to not only make ultraprocessed food healthier but to urge other food companies to implement similar analysis and reformulation of their product lines to improve the metabolic health and well-being of consumers worldwide.
Collapse
Affiliation(s)
- Timothy S. Harlan
- Division of General Internal Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| | - Rachel V. Gow
- Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | | | - P. Wolfram Alderson
- Human & Environmental Health Department, Kuwaiti Danish Dairy Company, Kuwait City, Kuwait
| | - Robert H. Lustig
- Department of Pediatrics and Institute for Health Policy Studies, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
38
|
Slurink IA, Chen L, Magliano DJ, Kupper N, Smeets T, Soedamah-Muthu SS. Dairy product consumption and incident prediabetes in the Australian Diabetes, Obesity and Lifestyle Study with 12 years follow up. J Nutr 2023:S0022-3166(23)35505-6. [PMID: 37003506 DOI: 10.1016/j.tjnut.2023.03.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/07/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Investigating modifiable risk factors of early stages of the development of type 2 diabetes is essential for effective prevention. Some studies show protective associations between dairy and prediabetes, yet associations are heterogenous by type and fat content of dairy foods. OBJECTIVE To examine the relationship between the consumption of dairy, including different types of dairy products and the risk of prediabetes. METHODS The study included 4,891 participants with normal glucose tolerance (aged 49.0±12.3 years, 57% female) of the Australian Diabetes, Obesity and Lifestyle (AusDiab) study, a longitudinal population-based study. Dairy intake was measured at baseline using a food frequency questionnaire. Prediabetes at 5-year and 12-year follow-up was defined according to WHO criteria as fasting plasma glucose levels of 110-125 mg/dl or 2-hour plasma glucose levels of 140-199 mg/dl. Associations were analyzed using Poisson regression, adjusted for social demographics, lifestyle behaviors, family history of diabetes, and food group intake. RESULTS 765 (15.6%) incident cases of prediabetes were observed. The mean intake of dairy foods was 2.4±1.2 servings/day, mostly consisting of low-fat milk (0.70±0.78) and high-fat milk (0.47±0.72). A higher intake of high-fat dairy (RRservings/day, 0.92, 95%CI 0.85-1.00), high-fat milk (0.89, 0.80-0.99), and total cheese (0.74, 0.56-0.96)was associated with lower prediabetes risk. Low-fat milk intake was associated non-linearly with prediabetes risk. Low-fat dairy foods, total milk, yogurt, low-fat cheese, and ice cream were not associated with prediabetes risk. CONCLUSION In this large Australian cohort, protective associations were found for high-fat dairy types, while neutral associations were seen for low-fat dairy. Studies with more detail on sugar content of types of dairy foods and products eaten with dairy foods (e.g., cereals or jam), as well as studies into potential causal mechanisms of the health effects of dairy intake are required.
Collapse
Affiliation(s)
- Isabel Al Slurink
- Center of Research on Psychological disorders and Somatic diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, P.O. Box 90153, 5000 LE, Tilburg, The Netherlands.
| | - Lei Chen
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| | - Dianna J Magliano
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia; School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Nina Kupper
- Center of Research on Psychological disorders and Somatic diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, P.O. Box 90153, 5000 LE, Tilburg, The Netherlands
| | - Tom Smeets
- Center of Research on Psychological disorders and Somatic diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, P.O. Box 90153, 5000 LE, Tilburg, The Netherlands
| | - Sabita S Soedamah-Muthu
- Center of Research on Psychological disorders and Somatic diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, P.O. Box 90153, 5000 LE, Tilburg, The Netherlands; Institute for Food, Nutrition and Health, University of Reading, Reading, RG6 6AR United Kingdom
| |
Collapse
|
39
|
She Y, Wang K, Makarowski A, Mangat R, Tsai S, Willing BP, Proctor SD, Richard C. Low-fat dairy consumption improves intestinal immune function more than high-fat dairy in a diet-induced swine model of insulin resistance. Eur J Nutr 2023; 62:699-711. [PMID: 36197467 DOI: 10.1007/s00394-022-03013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/22/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE To understand the effects of consuming high-fat and low-fat dairy products on postprandial cardiometabolic risk factors and intestinal immune function, we used an established low birthweight (LBW) swine model of diet-induced insulin resistance. METHODS LBW piglets were randomized to consume one of the 3 experimental high fat diets and were fed for a total of 7 weeks: (1) Control high fat (LBW-CHF), (2) CHF diet supplemented with 3 servings of high-fat dairy (LBW-HFDairy) and (3) CHF diet supplemented with 3 servings of low-fat dairy (LBW-LFDairy). As comparison groups, normal birthweight (NBW) piglets were fed a CHF (NBW-CHF) or standard pig grower diet (NBW-Chow). At 11 weeks of age, all piglets underwent an established modified oral glucose and fat tolerance test. At 12 weeks of age, piglets were euthanized and ex vivo cytokine production by cells isolated from mesenteric lymph node (MLN) stimulated with mitogens was assessed. RESULTS Dairy consumption did not modulate postprandial plasma lipid, inflammatory markers and glucose concentrations. A lower production of IL-2 and TNF-α after pokeweed mitogen (PWM) stimulation was observed in LBW-CHF vs NBW-Chow (P < 0.05), suggesting impaired MLN T cell function. While feeding high-fat dairy had minimal effects, feeding low-fat dairy significantly improved the production of IL-2 and TNF-α after PWM stimulation (P < 0.05). CONCLUSIONS Irrespective of fat content, dairy had a neutral effect on postprandial cardiometabolic risk factors. Low-fat dairy products improved intestinal T cell function to a greater extent than high-fat dairy in this swine model of obesity and insulin resistance.
Collapse
Affiliation(s)
- Yongbo She
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Kun Wang
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Alexander Makarowski
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Rabban Mangat
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Benjamin P Willing
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Spencer D Proctor
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Caroline Richard
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Science, 4-002G Li Ka Shing (LKS) Centre for Health Research Innovation, University of Alberta, Edmonton, AB, T6G 2E, Canada.
| |
Collapse
|
40
|
Poli A, Agostoni C, Visioli F. Dietary Fatty Acids and Inflammation: Focus on the n-6 Series. Int J Mol Sci 2023; 24:ijms24054567. [PMID: 36901998 PMCID: PMC10003459 DOI: 10.3390/ijms24054567] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Among the polyunsaturated fatty acids (PUFAs), those belonging to the n-3 (or ω3) series, i.e., alpha-linolenic (ALA), eicosapentaenoic (EPA), and docosahexaenoic (DHA) acids have been studied for decades from a pharma-nutritional viewpoint, namely in relation to cardiovascular health. More recent research is focusing on n-6 PUFAs, e.g., linoleic acid (LA), whose levels of consumption are much higher than those of n-3 and that cannot be used "pharmacologically". Perhaps because of this, the biological actions of n-6 PUFAs have not been investigated in details as those of their n-3 counterparts. However, an increasing body of evidence underscores their healthful actions on the cardiovascular system. Among the critiques to n-6 PUFAs and, particularly, LA there is the fact that they are precursors of pro-inflammatory eicosanoids. Hence, the hypothesis posits that we should reduce their intakes precisely to avoid increasing systemic, low-grade inflammation, i.e., one of the major etiological agents in degenerative diseases. In this narrative review, we address the issue of whether n-6 PUFAs are indeed pro-inflammatory, we discuss the most recent evidence of their role(s) in human health and prognosis, and we conclude that adequate intakes of n-6 fatty acids are associated with better cardiovascular health and child development.
Collapse
Affiliation(s)
- Andrea Poli
- Nutrition Foundation of Italy, 20124 Milano, Italy
| | - Carlo Agostoni
- Pediatric Area, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Francesco Visioli
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy
- IMDEA-Food, CEI UAM + CSIC, 28049 Madrid, Spain
- Correspondence: ; Tel.: +39-0498276107
| |
Collapse
|
41
|
MetaboVariation: Exploring Individual Variation in Metabolite Levels. Metabolites 2023; 13:metabo13020164. [PMID: 36837783 PMCID: PMC9965648 DOI: 10.3390/metabo13020164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
To date, most metabolomics biomarker research has focused on identifying disease biomarkers. However, there is a need for biomarkers of early metabolic dysfunction to identify individuals who would benefit from lifestyle interventions. Concomitantly, there is a need to develop strategies to analyse metabolomics data at an individual level. We propose "MetaboVariation", a method that models repeated measurements on individuals to explore fluctuations in metabolite levels at an individual level. MetaboVariation employs a Bayesian generalised linear model to flag individuals with intra-individual variations in their metabolite levels across multiple measurements. MetaboVariation models repeated metabolite levels as a function of explanatory variables while accounting for intra-individual variation. The posterior predictive distribution of metabolite levels at the individual level is available, and is used to flag individuals with observed metabolite levels outside the 95% highest posterior density prediction interval at a given time point. MetaboVariation was applied to a dataset containing metabolite levels for 20 metabolites, measured once every four months, in 164 individuals. A total of 28% of individuals with intra-individual variations in three or more metabolites were flagged. An R package for MetaboVariation was developed with an embedded R Shiny web application. To summarize, MetaboVariation has made considerable progress in developing strategies for analysing metabolomics data at the individual level, thus paving the way toward personalised healthcare.
Collapse
|
42
|
Yehia NA, Lai KZH, Semnani-Azad Z, Blanco Mejia S, Bazinet RP, Beaudry JL, Hanley AJ. Association of branched chain fatty acids with cardiometabolic disorders in humans: a systematic review. Nutr Rev 2023; 81:180-190. [PMID: 36029228 DOI: 10.1093/nutrit/nuac051] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
CONTEXT Despite advances in treatments for cardiometabolic disorders such as type 2 diabetes mellitus and obesity, the increasing frequency of these conditions is of major clinical and public health concern. Therefore, primary prevention including diet and lifestyle approaches continues to play a key role in risk reduction. Meta-analyses of prospective cohort studies have documented inverse associations of dairy consumption with the incidence of different cardiometabolic disorders. Dairy is the largest dietary contributor of branched chain fatty acids (BCFAs), which have been suggested to not only serve as biomarkers of dairy consumption but may also have bioactive properties contributing to reducing the risk of cardiometabolic outcomes. To date, however, the literature on this topic has not been systematically reviewed. OBJECTIVE The aim here was to report the results of a systematic review of the association of BCFAs with cardiometabolic disorders in humans. DATA SOURCES Search terms were developed and run through the Ovid MEDLINE, Ovid Embase, and the Cochrane Library databases. DATA EXTRACTION Articles were selected on the basis of prespecified inclusion criteria and assessed for risk of bias by independent reviewers. RESULTS Four studies (n = 2 cross sectional; n = 1 randomized feeding trial and n = 1 pre-post study) were identified. Two studies reported significant inverse associations between serum BCFAs and insulin resistance, triglycerides and/or body mass index. One study identified an inverse association between adipose tissue monomethyl BCFAs and skeletal muscle insulin resistance. In contrast, the randomized feeding trial reported no significant differences to stool BCFA concentrations or body mass index in obese participants following assignment to fruit-vegetable or whole-grain diet groups compared with a refined-grain control group. CONCLUSIONS Current evidence suggests beneficial associations of circulating BCFAs with cardiometabolic risk phenotypes, although data in human participants are limited, indicating that additional research is required. PROSPERO REGISTRATION NO CRD42021224975.
Collapse
Affiliation(s)
- Nagam Anna Yehia
- are with the Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kira Zhi Hua Lai
- are with the Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhila Semnani-Azad
- with the Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sonia Blanco Mejia
- are with the Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,is with the Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Risk Factor Modification Centre, St Michael's Hospital, Toronto, Ontario, Canada
| | - Richard P Bazinet
- are with the Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jacqueline L Beaudry
- are with the Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anthony J Hanley
- are with the Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,with the Department of Medicine, Division of Endocrinology and Metabolism, University of Toronto, Toronto, Ontario, Canada.,is with the Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Wang K, She Y, Mangat R, Makarowski A, Roy BC, Bruce HL, Dyck MK, Richard C, Proctor SD. Preferential deposition of dairy derived fatty acids in muscle tissue is partially due to the upregulation of CD36 in a low-birth-weight swine model. J Anim Sci 2023; 101:skad113. [PMID: 37086180 PMCID: PMC10184697 DOI: 10.1093/jas/skad113] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/21/2023] [Indexed: 04/23/2023] Open
Abstract
Metabolic syndrome is a worldwide health issue. Previous research has revealed that low-birth weight (LBW) swine fed a high-fat (HF) diet were susceptible to insulin resistance (IR) and developed a preferential intestinal lipid absorption, hypertriglyceridemia, and muscle steatosis. We hypothesized that fatty acid transporters such as CD36, FATP4, and FABP2 could potentially explain the development of these conditions. In addition, dairy-derived fatty acids have been shown to be valid biomarkers to assess dairy intake, which can be utilized to investigate muscle lipid deposition in LBW swine. The overall aim of this study was to delineate molecular transport candidates responsible for intestinal lipid absorption and muscle lipid deposition in LBW swine; and secondly to determine what dietary fatty acids might accumulate preferentially in pork muscle when consuming dairy products. At 5 weeks of age, normal birth weight (NBW) and LBW piglets were randomly assigned to three experimental diets: 1-chow diet, 2-HF diet, or 3-isocaloric HF diet supplemented with full fat dairy products. At 12 weeks of age, piglets were euthanized, and carcass, fasting plasma, biceps femoris and jejunum mucosal scrapings were collected. Results showed that HF-fed LBW swine exhibited early signs of IR (fasting glucose, P < 0.05; fasting insulin, P = 0.091; HOMA-IR, P = 0.086) compared with NBW-Chow, which were attenuated with increased dairy intake. Muscle samples from HF-fed LBW swine contained significantly more triglyceride compared to Chow-fed NBW swine (P < 0.05). Increased dairy intake significantly increased myristic acid (C14:0) and DPA (C22:5n3) relative to HF feeding alone (P < 0.05). All HF-fed LBW swine (regardless of dairy intake) exhibited an upregulation of CD36 expression (but not FABP2) compared with NBW littermates in both the small intestine and muscle (P < 0.05). Interestingly, increased dairy intake significantly increased the Canadian Lean Yield percentage in LBW swine fed an HF diet (P < 0.05). Findings from this study provide evidence on the mechanistic pathway of intestinal and muscle lipid metabolism in an innovative LBW swine model. We have also revealed that increasing dairy intake can enhance the incorporation of dietary long-chain polyunsaturated fatty acids into pork, as well as increasing the predicted lean yield of the carcass.
Collapse
Affiliation(s)
- Kun Wang
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Yongbo She
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Rabban Mangat
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Makarowski
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Bimol C Roy
- Division of Animal Science, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Heather L Bruce
- Division of Animal Science, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Michael K Dyck
- Division of Animal Science, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Spencer D Proctor
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
- Metabolic and Cardiovascular Diseases Laboratory, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
44
|
We are what we eat: The role of lipids in metabolic diseases. ADVANCES IN FOOD AND NUTRITION RESEARCH 2023. [PMID: 37516463 DOI: 10.1016/bs.afnr.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lipids play a fundamental role, both structurally and functionally, for the correct functioning of the organism. In the last two decades, they have evolved from molecules involved only in energy storage to compounds that play an important role as components of cell membranes and signaling molecules that regulate cell homeostasis. For this reason, their interest as compounds involved in human health has been gaining weight. Indeed, lipids derived from dietary sources and endogenous biosynthesis are relevant for the pathophysiology of numerous diseases. There exist pathological conditions that are characterized by alterations in lipid metabolism. This is particularly true for metabolic diseases, such as liver steatosis, type 2 diabetes, cancer and cardiovascular diseases. The main issue to be considered is lipid homeostasis. A precise control of fat homeostasis is required for a correct regulation of metabolic pathways and safe and efficient energy storage in adipocytes. When this fails, a deregulation occurs in the maintenance of systemic metabolism. This happens because an increased concentrations of lipids impair cellular homeostasis and disrupt tissue function, giving rise to lipotoxicity. Fat accumulation results in many alterations in the physiology of the affected organs, mainly in metabolic tissues. These alterations include the activation of oxidative and endoplasmic reticulum stress, mitochondrial dysfunction, increased inflammation, accumulation of bioactive molecules and modification of gene expression. In this chapter, we review the main metabolic diseases in which alterations in lipid homeostasis are involved and discuss their pathogenic mechanisms.
Collapse
|
45
|
Correddu F, Carta S, Mazza A, Nudda A, Rassu SPG. Effect of extruded linseed on sarda donkey milk quality. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2022.2104176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Fabio Correddu
- Dipartimento di Agraria, Sezione di Scienze Zootecniche, University of Sassari, Sassari, Italy
| | - Silvia Carta
- Dipartimento di Agraria, Sezione di Scienze Zootecniche, University of Sassari, Sassari, Italy
| | - Antonio Mazza
- Dipartimento di Agraria, Sezione di Scienze Zootecniche, University of Sassari, Sassari, Italy
| | - Anna Nudda
- Dipartimento di Agraria, Sezione di Scienze Zootecniche, University of Sassari, Sassari, Italy
| | | |
Collapse
|
46
|
Sellem L, Jackson KG, Paper L, Givens ID, Lovegrove JA. Can individual fatty acids be used as functional biomarkers of dairy fat consumption in relation to cardiometabolic health? A narrative review. Br J Nutr 2022; 128:2373-2386. [PMID: 35086579 PMCID: PMC9723489 DOI: 10.1017/s0007114522000289] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/25/2021] [Accepted: 01/20/2022] [Indexed: 12/30/2022]
Abstract
In epidemiological studies, dairy food consumption has been associated with minimal effect or decreased risk of some cardiometabolic diseases (CMD). However, current methods of dietary assessment do not provide objective and accurate measures of food intakes. Thus, the identification of valid and reliable biomarkers of dairy product intake is an important challenge to best determine the relationship between dairy consumption and health status. This review investigated potential biomarkers of dairy fat consumption, such as odd-chain, trans- and branched-chain fatty acids (FA), which may improve the assessment of full-fat dairy product consumption. Overall, the current use of serum/plasma FA as biomarkers of dairy fat consumption is mostly based on observational evidence, with a lack of well-controlled, dose-response intervention studies to accurately assess the strength of the relationship. Circulating odd-chain SFA and trans-palmitoleic acid are increasingly studied in relation to CMD risk and seem to be consistently associated with a reduced risk of type 2 diabetes in prospective cohort studies. However, associations with CVD are less clear. Overall, adding less studied FA such as vaccenic and phytanic acids to the current available evidence may provide a more complete assessment of dairy fat intake and minimise potential confounding from endogenous synthesis. Finally, the current evidence base on the direct effect of dairy fatty acids on established biomarkers of CMD risk (e.g. fasting lipid profiles and markers of glycaemic control) mostly derives from cross-sectional, animal and in vitro studies and should be strengthened by well-controlled human intervention studies.
Collapse
Affiliation(s)
- Laury Sellem
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Reading, RG6 6DZ, UK
- Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Kim G. Jackson
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Reading, RG6 6DZ, UK
- Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Laura Paper
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Reading, RG6 6DZ, UK
- Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Ian D. Givens
- Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Science, University of Reading, Whiteknights, Pepper Lane, Reading, RG6 6DZ, UK
- Institute for Food, Nutrition and Health, University of Reading, Reading, UK
| |
Collapse
|
47
|
Carta S, Correddu F, Battacone G, Pulina G, Nudda A. Comparison of Milk Odd- and Branched-Chain Fatty Acids among Human, Dairy Species and Artificial Substitutes. Foods 2022; 11:foods11244118. [PMID: 36553860 PMCID: PMC9778152 DOI: 10.3390/foods11244118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The aim of the study was to compare odd and branched-chain fatty acids (OBCFA) of milk from sheep, goat, cow, buffalo, donkey, human, and formula milk. Ruminant, monogastric, and human milks have different concentrations of these fatty acids (FA). To highlight the differences on OBCFA, a total of 282 individual milk samples were analyzed by gas chromatography. The OBCFA were found higher in ruminant than non-ruminant milks (p < 0.05). Among ruminants, sheep milk had the highest OBCFA (4.5 g/100 g of total FAME), whereases the lowest values were found in formula milk (0.18 g/100 g of total FAME). Regarding individual linear odd-chain FA (linear-OCFA), C11:0 was found higher in donkey milk than others, while sheep and buffalo milks had the greatest concentration of C15:0. Among BCFA, the iso-BCFA were higher than anteiso-BCFA in all considered milks. The isoC17:0 showed the highest concentration in all milks except for donkey and buffalo, which showed higher concentration of isoC16:0 than others. In conclusion, ruminant milks are different in terms of these FA compared to human milk and its substitutes. However, the greatest differences were found with formula milk, suggesting that this product needs the implementation of these FA to be more similar to human milk composition.
Collapse
|
48
|
Yuzbashian E, Pakseresht M, Vena J, Chan CB. Association of dairy consumption patterns with the incidence of type 2 diabetes: Findings from Alberta's Tomorrow Project. Nutr Metab Cardiovasc Dis 2022; 32:2760-2771. [PMID: 36333201 DOI: 10.1016/j.numecd.2022.09.022] [Citation(s) in RCA: 1] [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: 05/31/2022] [Revised: 08/22/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND AIMS We aimed to extract dairy consumption patterns of men and women from a population-based cohort and then assess the association of each consumption pattern with incident T2D risk. METHODS AND RESULTS This prospective study was conducted within the framework of Alberta's Tomorrow Project (ATP), in which 8615 men and 15,016 women provided information on dietary intake by completing a food-frequency questionnaire at baseline, and then were followed up over time to determine the incidence of T2D via questionnaires. Principal Component Analysis (PCA) was used to extract dairy consumption patterns (DCPs). The association between each extracted pattern and T2D incidence was estimated using multivariable logistic regression models.The incidence of T2D among men and women was 3.8 and 3.2%, respectively, and the mean duration of follow-up was 5.2 years. Three major DCPs were identified. After controlling for potential confounders, the OR for risk of T2D in men in the highest compared with those in the lowest quartile of the DCP3 (whole milk, regular cheese, and non-fat milk as a beverage and in cereal) was 0.64 (95%CI: 0.47 to 0.88, P-trend=0.001), whereas it was not significant for women. DCP1 and DCP2 were not associated with incident T2D in men or women. CONCLUSION Adherence to a DCP characterized by higher consumption of whole milk, regular cheese, and non-fat milk was associated with decreased risk of incident T2D only in men. Our results support current evidence that a combination of different dairy products, regardless of their fat content, might be favorable for health maintenance, at least in men.
Collapse
Affiliation(s)
- Emad Yuzbashian
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Mohammadreza Pakseresht
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; Cancer Research & Analytics, Cancer Care Alberta, Alberta Health Services, Alberta, Canada
| | - Jennifer Vena
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; Cancer Research & Analytics, Cancer Care Alberta, Alberta Health Services, Alberta, Canada
| | - Catherine B Chan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada; Department of Physiology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
49
|
Variations in the milk lipidomes of two dairy cow herds fed hay- or silage-based diets over a full year. Food Chem 2022; 390:133091. [DOI: 10.1016/j.foodchem.2022.133091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/15/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022]
|
50
|
Dietary fat quality impacts metabolic impairments of type 2 diabetes risk differently in male and female CD-1 ® mice. Br J Nutr 2022; 128:1013-1028. [PMID: 34605388 DOI: 10.1017/s0007114521004001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Metabolic impairments associated with type 2 diabetes, including insulin resistance and loss of glycaemic control, disproportionately impact the elderly. Lifestyle interventions, such as manipulation of dietary fat quality (i.e. fatty acid (FA) composition), have been shown to favourably modulate metabolic health. Yet, whether or not chronic consumption of beneficial FAs can protect against metabolic derangements and disease risk during ageing is not well defined. We sought to evaluate whether long-term dietary supplementation of fish-, dairy- or echium-derived FAs to the average FA profile in a U.S. American diet may offset metabolic impairments in males and females during ageing. One-month-old CD-1® mice were fed isoenergetic, high-fat (40 %) diets with the fat content composed of either 100 % control fat blend (CO) or 70 % CO with 30 % fish oil, dairy fat or echium oil for 13 months. Every 3 months, parameters of glucose homoeostasis were evaluated via glucose and insulin tolerance tests. Glucose tolerance improved in males consuming a diet supplemented with fish oil or echium oil as ageing progressed, but not in females. Yet, females were more metabolically protected than males regardless of age. Additionally, Spearman correlations were performed between indices of glucose homoeostasis and previously reported measurements of diet-derived FA content in tissues and colonic bacterial composition, which also revealed sex-specific associations. This study provides evidence that long-term dietary fat quality influences risk factors of metabolic diseases during ageing in a sex-dependent manner; thus, sex is a critical factor to be considered in future dietary strategies to mitigate type 2 diabetes risk.
Collapse
|