1
|
Lin N, Zeleznik OA, Vitonis AF, Laliberte A, Shafrir AL, Avila-Pacheco J, Clish C, Terry KL, Missmer SA, Sasamoto N. Plasma metabolites associated with endometriosis in adolescents and young adults. Hum Reprod 2025; 40:843-854. [PMID: 40107296 PMCID: PMC12046077 DOI: 10.1093/humrep/deaf040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/04/2025] [Indexed: 03/22/2025] Open
Abstract
STUDY QUESTION What are the plasma metabolomics profiles associated with endometriosis in adolescents and young adults? SUMMARY ANSWER Our findings show dysregulation of plasma metabolomic profiles in adolescents and young adults with endometriosis, revealing systemic elevation of fatty acyls and ceramides in endometriosis cases compared to controls. WHAT IS KNOWN ALREADY Endometriosis is a gynecologic disease often presenting with severe pelvic pain impacting around 200 million reproductive-aged women worldwide. However, little is known about the pathophysiology and molecular features of endometriosis diagnosed during adolescence and young adulthood. STUDY DESIGN, SIZE, DURATION We conducted a cross-sectional analysis including 190 laparoscopically confirmed endometriosis cases and 120 controls who participated in The Women's Health Study: From Adolescence to Adulthood, which enrolled participants from 2012 to 2018. Control participants were females without a diagnosis of endometriosis enrolled from the same clinics as the cases or recruited from the general population. Among the cases, 81 had blood samples collected before and after surgery. PARTICIPANTS/MATERIALS, SETTING, METHODS Plasma metabolites were measured in blood collected at enrollment using liquid chromatography-tandem mass spectrometry, and a total of 430 known metabolites were evaluated in our analysis. We used linear regression adjusting for age at blood draw, BMI, hormone use, and fasting status at blood draw. Metabolite set enrichment analysis (MSEA) was used to identify metabolite classes. Number of effective tests (NEF) and false discovery rate (FDR) were used for multiple testing correction. MAIN RESULTS AND THE ROLE OF CHANCE The median age was 17 years for endometriosis cases and 22 years for controls. The majority of endometriosis cases had rASRM stage I or II (>95%). We identified 63 plasma metabolites associated with endometriosis (NEF < 0.05). Endometriosis cases had higher levels of plasma metabolites associated with proinflammatory response [e.g. eicosatrienoic acid (β = 0.61, 95% CI = 0.37, 0.86)], increased oxidative stress response [e.g. xanthine (β = 0.64, 95% CI = 0.39, 0.88)], and downregulation of metabolites related to apoptosis [glycocholic acid (β = -0.80, 95% CI = -1.04, -0.56)]. MSEA revealed increased fatty acyls (FDR = 2.3e-4) and ceramides (FDR = 6.0e-3) and decreased steroids and steroid derivatives (FDR = 1.3e-4) in endometriosis cases compared to controls. When we examined the changes in plasma metabolite profiles before and after surgery among endometriosis cases, 55 endometriosis-associated metabolites significantly changed from before to after surgery. MSEA revealed steroids and steroid derivatives (FDR = 8.1e-4) significantly increased after surgery, while fatty acyls (FDR = 1.2e-4) significantly decreased after surgery. Ceramides did not change from pre- to post-surgery and were elevated in post-surgical blood compared to controls (FDR = 3.9e-3). LIMITATIONS, REASONS FOR CAUTION Our study population mainly consists of self-reported non-Hispanic, white individuals and endometriosis cases with superficial peritoneal lesions only, so the generalizability may be limited. Furthermore, despite our large study population of adolescents and young adults with endometriosis, sample size was limited to conduct detailed stratified analyses of plasma metabolomic profiles, especially by post-surgical pelvic pain outcomes. WIDER IMPLICATIONS OF THE FINDINGS Our study includes the utilization of state-of-the-art metabolomics technology with high reproducibility to comprehensively investigate the metabolites that were associated with endometriosis diagnosed in adolescents and young adults. Our results suggest a positive impact of endometriosis-related surgery for some, but not all, on systemic metabolic dysregulation in young patients with endometriosis. These results warrant further investigation on whether and how persistent systemic changes despite treatment may lead to long-term chronic disease risk among those diagnosed with endometriosis. STUDY FUNDING/COMPETING INTEREST(S) Financial support for establishment of and data collection within the A2A cohort was provided by the J. Willard and Alice S. Marriott Foundation, and support for assay costs was in part provided by the Peery family. This project was funded by Eunice Kennedy Shriver National Institute of Child Health and Human Development R21HD107266. S.A.M., A.L.S., and K.L.T. were supported by Eunice Kennedy Shriver National Institute of Child Health and Human Development R01HD094842. S.A.M. received grant funding from AbbVie, National Institutes of Health, Department of Defense, and Marriott Family Foundation; received honoraria from WERF, Huilun Shanghai, and University of Kansas Medical Center; travel support from SRI, ESHRE, FWGBD, University of Michigan, MIT, ASRM, LIDEA Registry, Taiwan Endometriosis Society, SEUD, Japan Endometriosis Society, NASEM, Endometriosis Foundation of America, Gedeon Richter Symposium at ESHRE; Board member receiving financial remuneration from AbbVie, Roche, LIDEA Registry, Editor of Frontiers in Reproductive Health, Roundtable participation for Abbott; Board member without financial remuneration from NextGen Jane and Statistical Advisory Board member of Human Reproduction; leadership role in Society for Women's Health Research, World Endometriosis Society, World Endometriosis Research Foundation, ASRM, ESHRE. N.S. and K.L.T. receive grant funding from Aspira Women's Health unrelated to this project. The remaining authors have no disclosures relevant to this manuscript. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Nan Lin
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Allison F Vitonis
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
| | - Ashley Laliberte
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
| | - Amy L Shafrir
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Department of Health Sciences and Nutrition, School of Nursing and Health Sciences, Merrimack College, North Andover, MA, USA
- Division of Adolescent and Young Adult Medicine, Department of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Clary Clish
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Kathryn L Terry
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Stacey A Missmer
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Naoko Sasamoto
- Department of Obstetrics and Gynecology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Boston Center for Endometriosis, Boston Children’s Hospital and Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
2
|
Morales G, Bugueño C, Valenzuela R, Chamorro R, Leiva C, Gotteland M, Trunce-Morales S, Pizarro-Aranguiz N, Durán-Agüero S. Association between cheese consumption but not other dairy products and lower obesity risk in adults. PLoS One 2025; 20:e0320633. [PMID: 40299899 PMCID: PMC12040181 DOI: 10.1371/journal.pone.0320633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 02/21/2025] [Indexed: 05/01/2025] Open
Abstract
INTRODUCTION Some studies have associated dairy consumption with a lower risk of obesity. However, these studies are concentrated in developed countries with high dairy consumption. In developing countries, the evidence is scarce. This study aimed to evaluate the association between the consumption of different types of dairy products and obesity in Chilean adults. MATERIALS AND METHODS A cross-sectional study, stratified by sex and age, was carried out using a validated online survey to assess the consumption of dairy products among adults living in Chile. Dairy product consumption was then classified into tertiles. Obesity was determined based on self-reported body mass index (BMI) ≥ 30 kg/m2. Logistic regression models were used to assess the association between dairy consumption and obesity, adjusting for several confounding variables. RESULTS In total, 2008 participants were included in the analyses. Forty-seven percent, 39% and 14% belonged to the <35 years, 35-60 years, and ≥60-year groups, respectively. 55% were female, 86% had a low-medium socioeconomic level. Cow-derived cheese, milk, and yogurt were the most commonly consumed dairy products. Obese participants had a lower total consumption of dairy products (17.1%) than normal-weight subjects (25.7%, p<0.05). Higher cheese intake was significantly associated with a lower obesity risk (ORadj: 0.70; 95%CI 0.51-0.96, p<0.05). Other types of dairy products and total consumption of dairy products were not significantly associated. DISCUSSION AND CONCLUSIONS Habitual cheese consumption, but not other dairy products, was associated with a lower risk of obesity in this sample of Chilean adults.
Collapse
Affiliation(s)
- Gladys Morales
- Departamento de Salud Pública, Facultad de Medicina, Universidad de La Frontera, Temuco, Chile
- Centro de Investigación en Epidemiología Cardiovascular y Nutricional (EPICYN), Universidad de La Frontera, Temuco, Chile
| | - Claudia Bugueño
- Departamento de clínica, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Rodrigo Valenzuela
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile. Avenida Independencia. Independencia, Santiago, Chile
| | - Rodrigo Chamorro
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile. Avenida Independencia. Independencia, Santiago, Chile
| | - Carla Leiva
- Departamento de Nutrición y Dietética, Escuela de Ciencias de la Salud, Facultad de Medicina, Pontificia Universidad Católica de Chile, San Joaquín, Chile
| | - Martin Gotteland
- Departamento de Nutrición, Facultad de Medicina, Universidad de Chile. Avenida Independencia. Independencia, Santiago, Chile
| | - Silvana Trunce-Morales
- Carrera de Nutrición y Dietética, Departamento de Salud, Universidad de Los Lagos, Osorno, Chile
| | | | - Samuel Durán-Agüero
- Escuela de Nutrición y Dietética, Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Los Leones, Chile
| |
Collapse
|
3
|
Kurhaluk N. Palm oil as part of a high-fat diet: advances and challenges, or possible risks of pathology? Nutr Rev 2025; 83:e547-e573. [PMID: 38699959 DOI: 10.1093/nutrit/nuae038] [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: 05/05/2024] Open
Abstract
Nutritional status disorders have the most significant impact on the development of cardiovascular and oncologic diseases; therefore, the interest in the study of palm oil as among the leading components of nutrition has been increasing. The data examined in this review were sourced from the Scopus, SCIE (Web of Science), PubMed and PubMed Central, MEDLINE, CAPlus/SciFinder, and Embase databases; experts in the field; bibliographies; and abstracts from review analyses from the past 15 years. This review summarizes recent research data focusing on the quantitative and qualitative composition of nutrition of modern humans; concepts of the relationship between high-fat diets and disorders of insulin functioning and transport and metabolism of fatty acids; analyses of data regarding the palmitic acid (16:0) to oleic acid (18:1) ratio; and the effect of diet based on palm oil consumption on cardiovascular risk factors and lipid and lipoprotein levels. Several studies suggest a potential vector contributing to the transmission of maternal, high-fat-diet-induced, addictive-like behaviors and obesogenic phenotypes across generations. The relationship between cholesterol accumulation in lysosomes that may lead to lysosome dysfunction and inhibition of the autophagy process is analyzed, as is the progression of inflammatory diseases, atherosclerosis, nonalcoholic liver inflammation, and obesity with associated complications. Data are discussed from analyses of differences between rodent models and human population studies in the investigated different effects of palm oil consumption as a high-fat diet component. A conclusion is reached that the results cannot be generalized in human population studies because no similar effects were observed. Although there are numerous published reports, more studies are necessary to elucidate the complex regulatory mechanisms in digestive and nutrition processes, because there are great differences in lipoprotein profiles between rodents and humans, which makes it difficult to reproduce the pathology of many diseases caused by different types of the high-fat diet.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Department of Animal Physiology, Institute of Biology, Pomeranian University in Słupsk, Słupsk, Poland
| |
Collapse
|
4
|
Karakuła-Juchnowicz H, Banaszek A, Sitarz R, Karakuła K, Wingralek Z, Rząd Z, Stachowska E, Jakubiak N, Urbańska E, Juchnowicz D, Rog J. The role of fatty acids in the emotional well-being of young adults: associations between fatty acid levels and symptoms of depression, anxiety, stress, and sleep disturbances. POSTEPY PSYCHIATRII NEUROLOGII 2024; 33:201-212. [PMID: 40070427 PMCID: PMC11891755 DOI: 10.5114/ppn.2024.144224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 03/14/2025]
Abstract
Purpose Young adults experience high stress levels, leading to mood disorders. This study investigates the associations between specific fatty acid levels, lipid profiles, inflammatory markers, and emotional well-being among young adults. Methods Seventy-two young adults aged 18-35 participated in this study. Participants completed self-assessments of depression severity (PHQ-9), stress (PSS-10), insomnia (ISI), and anxiety (GAD-7). Blood samples were collected and analyzed for plasma fatty acid profiles, lipid profiles, C-reactive protein (CRP) and kynurenine pathway metabolites. Classification and Regression Tree (C&RT) and multivariate stepwise regression analyses were employed to identify potential predictors of mental health outcomes. Results The analyses revealed significant associations between certain fatty acids, lipid markers, and mental health conditions. Lauric acid, myristic acid, and eicosatrienoic acid were identified as potential indicators of mental health issues. Higher levels of palmitoleic acid were linked to increased depressive symptoms, while higher oleic acid levels were associated with reduced depression. Anxiety was influenced by myristoleic acid and docosahexaenoic acid. Stress and sleep disturbances correlated with specific fatty acids. The models explained a significant percentage of variability in mental health outcomes, accounting for 25% in both depressive symptoms and anxiety, 23% in stress, and 43% in sleep disturbances. Conclusions Specific fatty acids, associated with lipid profiles, kynurenic acid, and CRP, significantly impact the mental health of young adults. Monitoring biomarkers may assist in managing mental health disorders. Personalized dietary interventions could improve well-being and sleep quality. Further research is needed to confirm these findings and establish causal relationships.
Collapse
Affiliation(s)
- Hanna Karakuła-Juchnowicz
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Poland
| | - Agnieszka Banaszek
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Poland
| | - Ryszard Sitarz
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Poland
| | - Kaja Karakuła
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Poland
| | - Zuzanna Wingralek
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Poland
| | - Zuzanna Rząd
- I Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Poland
| | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Poland
| | - Natalia Jakubiak
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Poland
| | - Ewa Urbańska
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Poland
| | | | - Joanna Rog
- Laboratory of Human Metabolism Research, Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Science, Poland Abstract
| |
Collapse
|
5
|
Tillander V, Holmer M, Hagström H, Petersson S, Brismar TB, Stål P, Lindqvist C. Associations between dietary fatty acid and plasma fatty acid composition in non-alcoholic fatty liver disease: secondary analysis from a randomised trial with a hypoenergetic low-carbohydrate high-fat and intermittent fasting diet. Br J Nutr 2024; 132:1-13. [PMID: 39290088 PMCID: PMC11499086 DOI: 10.1017/s0007114524001673] [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: 12/22/2023] [Revised: 07/02/2024] [Accepted: 07/24/2024] [Indexed: 09/19/2024]
Abstract
Dietary fatty acids (FA) affect metabolic risk factors. The aim of this study was to explore if changes in dietary fat intake during energy restriction were associated with plasma FA composition. The study also investigated if these changes were associated with changes in liver fat, liver stiffness and plasma lipids among persons with non-alcoholic fatty liver disease. Dietary and plasma FA were investigated in patients with non-alcoholic fatty liver disease (n 48) previously enrolled in a 12-week-long open-label randomised controlled trial comparing two energy-restricted diets: a low-carbohydrate high-fat diet and intermittent fasting diet (5:2), to a control group. Self-reported 3 d food diaries were used for FA intake, and plasma FA composition was analysed using GC. Liver fat content and stiffness were measured by MRI and transient elastography. Changes in intake of total FA (r 0·41; P = 0·005), SFA (r 0·38; P = 0·011) and MUFA (r 0·42; P = 0·004) were associated with changes in liver stiffness. Changes in plasma SFA (r 0·32; P = 0·032) and C16 : 1n-7 (r 0·33; P = 0·028) were positively associated with changes in liver fat, while total n-6 PUFA (r -0·33; P = 0·028) and C20 : 4n-6 (r -0·42; P = 0·005) were inversely associated. Changes in dietary SFA, MUFA, cholesterol and C20:4 were positively associated with plasma total cholesterol and LDL-cholesterol. Modifying the composition of dietary fats during dietary interventions causes changes in the plasma FA profile in patients with non-alcoholic fatty liver disease. These changes are associated with changes in liver fat, stiffness, plasma cholesterol and TAG. Replacing SFA with PUFA may improve metabolic parameters in non-alcoholic fatty liver disease patients during weight loss treatment.
Collapse
Affiliation(s)
- Veronika Tillander
- Division of Clinical Chemistry, Cardio Metabolic Unit, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Holmer
- Unit of Gastroenterology and Hepatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| | - Hannes Hagström
- Unit of Gastroenterology and Hepatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| | - Sven Petersson
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Torkel B. Brismar
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Karolinska University Hospital in Huddinge, Stockholm sE-14186, Sweden
| | - Per Stål
- Unit of Gastroenterology and Hepatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Hepatology, Department of Upper GI, Karolinska University Hospital, Stockholm, Sweden
| | - Catarina Lindqvist
- Unit of Gastroenterology and Hepatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Medical Unit Clinical Nutrition, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
6
|
Yan L, Yan Y, Yang K, Chang Q, Zhang L. Metabolomics reveals dysregulated all-trans retinoic acid and polyunsaturated fatty acid metabolism contribute to PXR-induced hepatic steatosis in mice. Toxicol Lett 2024; 398:150-160. [PMID: 38971454 DOI: 10.1016/j.toxlet.2024.07.003] [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: 08/15/2023] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Activation of pregnane X receptor (PXR) by xenobiotics has been associated with metabolic diseases. This study aimed to reveal the impact of PXR activation on hepatic metabolome and explore novel mechanisms underlying PXR-mediated lipid metabolism disorder in the liver. Wild-type and PXR-deficient male C57BL/6 mice were used as in vivo models, and hepatic steatosis was induced by pregnenolone-16α-carbonitrile, a typical rodent PXR agonist. Metabolomic analysis of liver tissues showed that PXR activation led to significant changes in metabolites involved in multiple metabolic pathways previously reported, including lipid metabolism, energy homeostasis, and amino acid metabolism. Moreover, the level of hepatic all-trans retinoic acid (ATRA), the main active metabolite of vitamin A, was significantly increased by PXR activation, and genes involved in ATRA metabolism exhibited differential expression following PXR activation or deficiency. Consistent with previous research, the expression of downstream target genes of peroxisome proliferator-activated receptor α (PPARα) was decreased. Analysis of fatty acids by Gas Chromatography-Mass Spectrometer further revealed changes in polyunsaturated fatty acid metabolism upon PXR activation, suggesting inhibition of PPARα activity. Taken together, our findings reveal a novel metabolomic signature of hepatic steatosis induced by PXR activation in mice.
Collapse
Affiliation(s)
- Liang Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou Univerisity, Zhengzhou 450052, China.
| | - Yachun Yan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou Univerisity, Zhengzhou 450052, China
| | - Kun Yang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Qi Chang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medicine, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
7
|
Ozdemir VF, Kocyigit R, Yanar M, Aydin R, Diler A, Palangi V, Lackner M. An investigation of slaughter weight and muscle type effects on carcass fatty acid profiles and meat textural characteristics of young Holstein Friesian bulls. Heliyon 2024; 10:e27316. [PMID: 38509920 PMCID: PMC10950803 DOI: 10.1016/j.heliyon.2024.e27316] [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: 08/02/2023] [Revised: 02/18/2024] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
Study objectives included the assessment of carcass fatty acid composition and meat texture characteristics of younger Holstein Friesian bulls. Three experimental groups were formed based on the weights of the 23 young bulls at slaughter: lighter, medium, and heavier. Samples were taken from the Gluteus medius (GM) and Longissimus thoracis muscles 24 h after slaughter. Fatty acid composition, Warner-Bratzler Meat Shear (WBS) measurements, as well as textural profile analysis (TPA) and sensory analysis of the muscle samples were conducted. The fatty acid composition was determined using Thin Layer Chromatography (HPTLC). Polyunsaturated fatty acids and dietary fatty acids give a neutral hypocholesterolemic effect in direct fluorescent antibody (DFA) contents, DFA/OFA (C14:0+C16:0) ratio, hardness, Warner-Bratzler Shear force and also the chews number - which is desirable - before swallowing (NCBS) the meat were significantly decreased with the increasing slaughter weight. Higher slaughter weight resulted in a larger amount of beef with a better panel tenderness score; however, the meat obtained from the LSW group was less healthy considering the fatty acid profile. Additionally, internal fat contained the highest saturated fatty acids concentrations, while subcutaneous fat contained the highest amount of monounsaturated fatty acids. Furthermore, intramuscular fat levels were highest in PUFA and PUFA/SFA ratio. As a result, this study strongly suggests that slaughter weight and anatomical location of fat samples contribute significantly to meat texture characteristics and fatty acid profiles in Holstein Friesian bulls.
Collapse
Affiliation(s)
- Veysel Fatih Ozdemir
- Department of Animal Science, College of Agriculture, Ataturk University, 25240, Erzurum, Türkiye
| | - Ridvan Kocyigit
- Department of Animal Science, College of Agriculture, Ataturk University, 25240, Erzurum, Türkiye
| | - Mete Yanar
- Department of Animal Science, College of Agriculture, Ataturk University, 25240, Erzurum, Türkiye
| | - Recep Aydin
- Department of Animal Science, College of Agriculture, Ataturk University, 25240, Erzurum, Türkiye
| | - Abdulkerim Diler
- Department of Plant and Animal Sciences, Vocational School of Technical Sciences, Ataturk University, 25240, Erzurum, Türkiye
| | - Valiollah Palangi
- Department of Animal Science, Faculty of Agriculture, Ege University, 35100, Izmir, Türkiye
| | - Maximilian Lackner
- Department of Industrial Engineering, University of Applied Sciences Technikum Wien, Hoechstaedtplatz 6, 1200, Vienna, Austria
| |
Collapse
|
8
|
Peng Y, Zhang L, Bao X, Qian X, Dong W, Jiang M. Palmitoleic acid-rich oleaginous yeast Scheffersomyces segobiensis DSM 27193 exerts anti-obesity effects by ameliorating hepatic steatosis and adipose tissue hypertrophy. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:2156-2164. [PMID: 37926439 DOI: 10.1002/jsfa.13100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Yeast biomass, encompassing fatty acids, terpenoids, vitamins, antioxidants, enzymes, and other bioactive compounds have been extensively utilized in food-related fields. The safety and potential bioactivities of Scheffersomyces segobiensis DSM 27193, an oleaginous yeast strain, are unclear. RESULTS Scheffersomyces segobiensis DSM 27193 accumulated large palmitoleic acid (POA) levels (43.4 g kg-1 biomass) according to the results of whole-cell components. We annotated the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and predicted the categories and host of the pathogen-host interactions (PHI) genes in S. segobiensis DSM 27193. However, S. segobiensis DSM 27193 did not exert toxic effects in mice. Administration of S. segobiensis DSM 27193 led to substantial weight reduction by diminishing food intake in an obesity mouse model. Additionally, it reversed hepatic steatosis and adipose tissue hypertrophy, and improved abnormalities in serum biochemical profiles such as triglyceride, total cholesterol, low-density lipoprotein cholesterol, lipopolysaccharide, tumor necrosis factor-α, interleukin-1β, and interleukin-6. CONCLUSION This study is the first to illustrate the safety and effects of S. segobiensis DSM 27193 against obesity and offers a scientific rationale for its application in functional food supplements. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yujia Peng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Lili Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Xinhui Bao
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Xiujuan Qian
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Weiliang Dong
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Min Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University, Nanjing, China
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Dong G, Xu S, Shi S. De Novo Biosynthesis of Free Vaccenic Acid with a Low Content of Oleic Acid in Saccharomyces cerevisiae. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16204-16211. [PMID: 37856078 DOI: 10.1021/acs.jafc.3c04793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Omega-7 (ω-7) fatty acids have potential application in the fields of nutraceutical, agricultural, and food industry. The natural ω-7 fatty acids are currently from plants or vegetable oils, which are unsustainable and limited by the availability of plant sources. Here, we developed an innovative biosynthetic route to produce vaccenic acid (C18:1 ω-7) while minimizing oleic acid (C18:1 ω-9) content in Saccharomyces cerevisiae. We have engineered S. cerevisiaeto produce C18:1 ω-7 by expressing a fatty acid elongase from Rattus norvegicus. To reduce the content of C18:1 ω-9, the endogenous desaturase Ole1 was replaced by the desaturase, which has specific activity on palmitoyl-coenzyme A (C16:0-CoA). Finally, the production of free C18:1 ω-7 was improved by optimizing the source of cytochrome b5 and overexpressing endoplasmic reticulum chaperones. After combining these strategies, the yield of C18:1 ω-7 was increased from 0 to 9.3 mg/g DCW and C18:1 ω-9 was decreased from 25.2 mg/g DCW to 1.6 mg/g DCW. This work shows a de novo synthetic pathway to produce the highest amount of free C18:1 ω-7 with a low content of C18:1 ω-9 in S. cerevisiae.
Collapse
Affiliation(s)
- Genlai Dong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, North Third Ring Road 15, Chaoyang District, Beijing 100029, China
| | - Shijie Xu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, North Third Ring Road 15, Chaoyang District, Beijing 100029, China
| | - Shuobo Shi
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, North Third Ring Road 15, Chaoyang District, Beijing 100029, China
| |
Collapse
|
12
|
Yang C, Veenstra J, Bartz TM, Pahl MC, Hallmark B, Chen YDI, Westra J, Steffen LM, Brown CD, Siscovick D, Tsai MY, Wood AC, Rich SS, Smith CE, O'Connor TD, Mozaffarian D, Grant SFA, Chilton FH, Tintle NL, Lemaitre RN, Manichaikul A. Genome-wide association studies and fine-mapping identify genomic loci for n-3 and n-6 polyunsaturated fatty acids in Hispanic American and African American cohorts. Commun Biol 2023; 6:852. [PMID: 37587153 PMCID: PMC10432561 DOI: 10.1038/s42003-023-05219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023] Open
Abstract
Omega-3 (n-3) and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) play critical roles in human health. Prior genome-wide association studies (GWAS) of n-3 and n-6 PUFAs in European Americans from the CHARGE Consortium have documented strong genetic signals in/near the FADS locus on chromosome 11. We performed a GWAS of four n-3 and four n-6 PUFAs in Hispanic American (n = 1454) and African American (n = 2278) participants from three CHARGE cohorts. Applying a genome-wide significance threshold of P < 5 × 10-8, we confirmed association of the FADS signal and found evidence of two additional signals (in DAGLA and BEST1) within 200 kb of the originally reported FADS signal. Outside of the FADS region, we identified novel signals for arachidonic acid (AA) in Hispanic Americans located in/near genes including TMX2, SLC29A2, ANKRD13D and POLD4, and spanning a > 9 Mb region on chromosome 11 (57.5 Mb ~ 67.1 Mb). Among these novel signals, we found associations unique to Hispanic Americans, including rs28364240, a POLD4 missense variant for AA that is common in CHARGE Hispanic Americans but absent in other race/ancestry groups. Our study sheds light on the genetics of PUFAs and the value of investigating complex trait genetics across diverse ancestry populations.
Collapse
Affiliation(s)
- Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Jenna Veenstra
- Departments of Biology and Statistics, Dordt University, Sioux Center, IA, USA
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Matthew C Pahl
- Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brian Hallmark
- Center for Biomedical Informatics and Biostatistics, University of Arizona, Tucson, AZ, USA
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jason Westra
- Fatty Acid Research Institute, Sioux Falls, SD, USA
| | - Lyn M Steffen
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Christopher D Brown
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Caren E Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Timothy D O'Connor
- Institute for Genome Sciences; Program in Personalized and Genomic Medicine; Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science & Policy, Tufts University, Tufts School of Medicine and Division of Cardiology, Tufts Medical Center, Boston, MA, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Floyd H Chilton
- School of Nutritional Sciences and Wellness and the BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Nathan L Tintle
- Fatty Acid Research Institute, Sioux Falls, SD, USA
- University of Illinois, Chicago, Chicago, IL, USA
| | - Rozenn N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
13
|
Bock M, von Schacky C, Scherr J, Lorenz E, Lechner B, Krannich A, Wachter R, Duvinage A, Edelmann F, Lechner K. De Novo Lipogenesis-Related Monounsaturated Fatty Acids in the Blood Are Associated with Cardiovascular Risk Factors in HFpEF Patients. J Clin Med 2023; 12:4938. [PMID: 37568339 PMCID: PMC10419368 DOI: 10.3390/jcm12154938] [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: 07/10/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
De novo lipogenesis (DNL)-related monounsaturated fatty acids (MUFAs) in the blood are associated with incident heart failure (HF). This observation's biological plausibility may be due to the potential of these MUFAs to induce proinflammatory pathways, endoplasmic reticulum stress, and insulin resistance, which are pathophysiologically relevant in HF. The associations of circulating MUFAs with cardiometabolic phenotypes in patients with heart failure with a preserved ejection fraction (HFpEF) are unknown. In this secondary analysis of the Aldosterone in Diastolic Heart Failure trial, circulating MUFAs were analysed in 404 patients using the HS-Omega-3-Index® methodology. Patients were 67 ± 8 years old, 53% female, NYHA II/III (87/13%). The ejection fraction was ≥50%, E/e' 7.1 ± 1.5, and the median NT-proBNP 158 ng/L (IQR 82-298). Associations of MUFAs with metabolic, functional, and echocardiographic patient characteristics at baseline/12 months follow-up (12 mFU) were analysed using Spearman's correlation coefficients and linear regression analyses, using sex/age as covariates. Circulating levels of C16:1n7 and C18:1n9 were positively associated with BMI/truncal adiposity and associated traits (dysglycemia, atherogenic dyslipidemia, and biomarkers suggestive of non-alcoholic-fatty liver disease). They were furthermore inversely associated with functional capacity at baseline/12 mFU. In contrast, higher levels of C20:1n9 and C24:1n9 were associated with lower cardiometabolic risk and higher exercise capacity at baseline/12 mFU. In patients with HFpEF, circulating levels of individual MUFAs were differentially associated with cardiovascular risk factors. Our findings speak against categorizing FA based on physicochemical properties. Circulating MUFAs may warrant further investigation as prognostic markers in HFpEF.
Collapse
Affiliation(s)
- Matthias Bock
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Munich, Germany
| | | | - Johannes Scherr
- University Center for Prevention and Sports Medicine, Balgrist University Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Elke Lorenz
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Benjamin Lechner
- Department of Internal Medicine IV, Ludwig-Maximilians University, 80336 Munich, Germany
| | | | - Rolf Wachter
- Clinic and Policlinic for Cardiology, University Hospital Leipzig, 04103 Leipzig, Germany
- University Medical Center Göttingen, Department of Cardiology and Pneumology, Georg-August University, 37099 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - André Duvinage
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Department of Prevention, Rehabilitation and Sports Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| | - Frank Edelmann
- Deutsches Herzzentrum der Charité, Department of Cardiology, Angiology and Intensive Care Medicine, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
- Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Katharina Lechner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, Munich, Germany
- Department of Prevention, Rehabilitation and Sports Medicine, School of Medicine, Technical University of Munich, 80992 Munich, Germany
| |
Collapse
|
14
|
Korkus E, Szustak M, Madaj R, Chworos A, Drzazga A, Koziołkiewicz M, Dąbrowski G, Czaplicki S, Konopka I, Gendaszewska-Darmach E. Trans-palmitoleic acid, a dairy fat biomarker, stimulates insulin secretion and activates G protein-coupled receptors with a different mechanism from the cis isomer. Food Funct 2023. [PMID: 37368452 DOI: 10.1039/d2fo03412c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Dietary trans-palmitoleic acid (trans 16:1n-7, tPOA), a biomarker for high-fat dairy product intake, has been associated with a lower risk of type 2 diabetes mellitus (T2DM) in some cross-sectional and prospective epidemiological studies. Here, we investigated the insulin secretion-promoting activity of tPOA and compared them with the effects evoked by the cis-POA isomer (cPOA), an endogenous lipokine biosynthesized in the liver and adipose tissue, and found in some natural food sources. The debate about the positive and negative relationships of those two POA isomers with metabolic risk factors and the underlying mechanisms is still going on. Therefore, we examined the potency of both POA isomers to potentiate insulin secretion in murine and human pancreatic β cell lines. We also investigated whether POA isomers activate G protein-coupled receptors proposed as potential targets for T2DM treatment. We show that tPOA and cPOA augment glucose-stimulated insulin secretion (GSIS) to a similar extent; however, their insulin secretagogue activity is associated with different signaling pathways. We also performed ligand docking and molecular dynamics simulations to predict the preferred orientation of POA isomers and the strength of association between those two fatty acids and GPR40, GPR55, GPR119, and GPR120 receptors. Overall, this study provides insight into the bioactivity of tPOA and cPOA toward selected GPCR functions, indicating them as targets responsible for the insulin secretagogue action of POA isomers. It reveals that both tPOA and cPOA may promote insulin secretion and subsequently regulate glucose homeostasis.
Collapse
Affiliation(s)
- Eliza Korkus
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537 Lodz, Poland.
| | - Marcin Szustak
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537 Lodz, Poland.
| | - Rafal Madaj
- Division of Bioorganic Chemistry Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza, 112, 90-363 Lodz, Poland
- Institute of Evolutionary Biology, Faculty of Biology, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089, Warsaw, Poland
| | - Arkadiusz Chworos
- Division of Bioorganic Chemistry Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza, 112, 90-363 Lodz, Poland
| | - Anna Drzazga
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537 Lodz, Poland.
| | - Maria Koziołkiewicz
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537 Lodz, Poland.
| | - Grzegorz Dąbrowski
- Faculty of Food Sciences, Chair of Plant Food Chemistry and Processing, University of Warmia and Mazury in Olsztyn, Pl. Cieszyński 1, 10-957 Olsztyn, Poland
| | - Sylwester Czaplicki
- Faculty of Food Sciences, Chair of Plant Food Chemistry and Processing, University of Warmia and Mazury in Olsztyn, Pl. Cieszyński 1, 10-957 Olsztyn, Poland
| | - Iwona Konopka
- Faculty of Food Sciences, Chair of Plant Food Chemistry and Processing, University of Warmia and Mazury in Olsztyn, Pl. Cieszyński 1, 10-957 Olsztyn, Poland
| | - Edyta Gendaszewska-Darmach
- Faculty of Biotechnology and Food Sciences, Institute of Molecular and Industrial Biotechnology, Lodz University of Technology, Stefanowskiego 2/22, 90-537 Lodz, Poland.
| |
Collapse
|
15
|
Rasekh P, Kameli A, Khoradmehr A, Baghban N, Mohebbi G, Barmak A, Nabipour I, Azari H, Heidari Y, Daneshi A, Bargahi A, Khodabandeh Z, Zare S, Afshar A, Shirazi R, Almasi-Turk S, Tamadon A. Proliferative Effect of Aqueous Extract of Sea Cucumber ( Holothuria parva) Body Wall on Human Umbilical Cord Mesenchymal Stromal/Stem Cells. Mar Drugs 2023; 21:md21050267. [PMID: 37233461 DOI: 10.3390/md21050267] [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: 11/13/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 05/27/2023] Open
Abstract
Sea cucumber extracts and their bioactive compounds have the potential for stem cell proliferation induction and for their beneficial therapeutic properties. In this study, human umbilical cord mesenchymal stromal/stem cells (hUC-MSCs) were exposed to an aqueous extract of Holothuria parva body walls. Proliferative molecules were detected using gas chromatography-mass spectrometry (GC-MS) analysis in an aqueous extract of H. parva. The aqueous extract concentrations of 5, 10, 20, 40, and 80 µg/mL and 10 and 20 ng/mL of human epidermal growth factor (EGF) as positive controls were treated on hUC-MSCs. MTT, cell count, viability, and cell cycle assays were performed. Using Western blot analysis, the effects of extracts of H. parva and EGF on cell proliferation markers were detected. Computational modeling was done to detect effective proliferative compounds in the aqueous extract of H. parva. A MTT assay showed that the 10, 20, and 40 µg/mL aqueous extract of H. parva had a proliferative effect on hUC-MSCs. The cell count, which was treated with a 20 µg/mL concentration, increased faster and higher than the control group (p < 0.05). This concentration of the extract did not have a significant effect on hUC-MSCs' viability. The cell cycle assay of hUC-MSCs showed that the percentage of cells in the G2 stage of the extract was biologically higher than the control group. Expression of cyclin D1, cyclin D3, cyclin E, HIF-1α, and TERT was increased compared with the control group. Moreover, expression of p21 and PCNA decreased after treating hUC-MSCs with the extract. However, CDC-2/cdk-1 and ERK1/2 had almost the same expression as the control group. The expression of CDK-4 and CDK-6 decreased after treatment. Between the detected compounds, 1-methyl-4-(1-methyl phenyl)-benzene showed better affinity to CDK-4 and p21 than tetradecanoic acid. The H. parva aqueous extract showed proliferative potential on hUC-MSCs.
Collapse
Affiliation(s)
- Poorya Rasekh
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Ali Kameli
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Neda Baghban
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Gholamhossein Mohebbi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Alireza Barmak
- Food Lab, Bushehr University of Medical Sciences, Bushehr 7518759577, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Hossein Azari
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Yaser Heidari
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Adel Daneshi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Afshar Bargahi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Zahra Khodabandeh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Shahrokh Zare
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Alireza Afshar
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Reza Shirazi
- Department of Anatomy, School of Medical Sciences, Medicine, UNSW Sydney, Sydney 3052, Australia
| | - Sahar Almasi-Turk
- Department of Anatomical Sciences, School of Medicine, Bushehr University of Medical Sciences, Bushehr 7514633196, Iran
| | - Amin Tamadon
- PerciaVista R&D Co., Shiraz 7167683745, Iran
- Department for Scientific Work, West Kazakhstan Marat Ospanov Medical University, Aktobe 030012, Kazakhstan
| |
Collapse
|
16
|
Na G, Zhang J, Lv D, Chen P, Song X, Cai F, Zheng S, Wan W, Shan Y. Germinated Brown rice enhanced n-3 PUFA metabolism in type 2 diabetes patients: A randomized controlled trial. Clin Nutr 2023; 42:579-589. [PMID: 36870245 DOI: 10.1016/j.clnu.2023.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND Brown rice (BR) has been considered as a potential strategy in improving T2DM. However, there are a lack of population-based trials on the association of Germinated brown rice (GBR) and diabetes. AIMS We aimed to explore the influence of GBR diet in T2DM patients for 3 months and whether this effect relates to serum fatty acids. METHODS Two hundred and twenty T2DM patients have been enrolled and eligible subjects (n = 112, 61 female, 51 male) were randomly divided into GBR intervention group (n = 56) and control group (n = 56). Except those who lost follow-up and withdrew, final GBR group and control group consisted of 42 and 43 patients, respectively. Participants in GBR group were asked to consume 100 g/d GBR instead of equal refined grain (RG) for 3 months, while control group maintain their usual eating habits. A structured questionnaire was used for demographic information at baseline, and basic indicators were measured both at the beginning and end of the trail to evaluate plasma glucose and lipids levels. RESULTS In GBR group, mean dietary inflammation index (DII) decreased, indicating GBR intervention retarded patient inflammation. Besides, glycolipid related parameters, including FBG, HbA1c, TC and HDL, were all significantly lower than those in control group. Excitingly, fatty acid composition was changed by intake of GBR, especially n-3 PUFA and n-3/n-6 PUFA rate were significantly increased. Moreover, subjects in GBR group had higher levels of n-3 metabolites, such as RVE, MaR1 and PD1, reducing inflammatory effect. In contrast, n-6 metabolites, like LTB4 and PGE2 which could promote inflammatory effect, were lower in GBR group. CONCLUSION We confirmed that diet with 100 g/d GBR for 3 months could really improve T2DM to some extent. This beneficial effect may be related to n-3 metabolites, namely inflammation changes. TRIAL REGISTRATION ChiCRT-IOR-17013999, www.chictr.org.cn.
Collapse
Affiliation(s)
- Guanqiong Na
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Jing Zhang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Dian Lv
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Ping Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Xinyue Song
- School of Chemical and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Fenfen Cai
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Sicong Zheng
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenting Wan
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China
| | - Yujuan Shan
- School of Public Health and Management, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
17
|
Bajahzer MF, Rosqvist F, Fridén M, Iggman D, Pingel R, Marklund M, Risérus U. Contrasting Carbohydrate Quantity and Quality and the Effects on Plasma Saturated and Monounsaturated Fatty Acids in Healthy Adults: A Randomized Controlled Trial. J Nutr 2023; 153:683-690. [PMID: 36797136 DOI: 10.1016/j.tjnut.2023.01.005] [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: 07/13/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND It is unclear whether moderate differences in dietary carbohydrate quantity and quality influence plasma FAs in the lipogenic pathway in healthy adults. OBJECTIVES We investigated the effects of different carbohydrate quantities and quality on plasma palmitate concentrations (primary outcome) and other saturated and MUFAs in the lipogenic pathway. METHODS Twenty healthy participants were randomly assigned, and 18 (50% women; age: 22-72 y; BMI: 18.2-32.7 kg/m2 and BMI was measured in kg/m2) started the cross-over intervention. During each 3-wk period (separated by a 1-wk washout period), 3 diets were consumed (all foods provided) in random order: low-carbohydrate (LC) (38% energy (E) carbohydrates, 25-35 g fiber/d, 0% E added sugars); high-carbohydrate/high-fiber (HCF) (53% E carbohydrates, 25-35 g fiber/d, 0% E added sugars); and high-carbohydrate/high-sugar (HCS) (53% E carbohydrates, 19-21 g fiber/d, 15% E added sugars). Individual FAs were measured proportionally to total FAs by GC in plasma cholesteryl esters, phospholipids, and TGs. False discovery rate-adjusted repeated measures ANOVA [ANOVA-false discovery rate (FDR)] was used to compare outcomes. RESULTS The self-reported intakes of carbohydrates and added- and free sugars were; 30.6% E and 7.4% E in LC, 41.4% E and 6.9% E in HCF, and 45.7% E and 10.3% in HCS. Plasma palmitate did not differ between the diet periods (ANOVA FDR P > 0.43, n = 18). After HCS, myristate concentrations in cholesterol esters and phospholipids were ≥19% higher than LC and ≥22% higher than HCF (P = 0.005). After LC, palmitoleate in TG was 6% lower compared with HCF and 7% compared with HCS (P = 0.041). Body weight differed (≤0.75 kg) between diets before FDR correction. CONCLUSIONS Different carbohydrate quantity and quality do not influence plasma palmitate concentrations after 3 wk in healthy Swedish adults, whereas myristate increased after the moderately higher intake of carbohydrate/high-sugar, but not carbohydrate/high-fiber. Whether plasma myristate is more responsive than palmitate to differences in carbohydrate intake requires further study, especially considering that participants deviated from the planned dietary targets. J Nutr 20XX;xx:xx-xx. This trial was registered at clinicaltrials.gov as NCT03295448.
Collapse
Affiliation(s)
- Mohammed F Bajahzer
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden; Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - David Iggman
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Ronnie Pingel
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Matti Marklund
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden; The George Institute for Global Health, Faculty of Medicine, UNSW Sydney, Sydeny, New South Wales, Australia; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
18
|
Razquin C, Ruiz-Canela M, Wernitz A, Toledo E, Corella D, Alonso-Gómez Á, Fitó M, Gómez-Gracia E, Estruch R, Fiol M, Lapetra J, Serra-Majem L, Ros E, Arós F, Salas-Salvadó J, Schulze MB, Martinez-Gonzalez MA. Effects of Supplemented Mediterranean Diets on Plasma-Phospholipid Fatty Acid Profiles and Risk of Cardiovascular Disease after 1 Year of Intervention in the PREDIMED Trial. Clin Chem 2023; 69:283-294. [PMID: 36683466 DOI: 10.1093/clinchem/hvac221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/09/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Plasma fatty acids (FAs) have been associated with cardiovascular disease (CVD) risk. Diet and endogenous metabolism influence the FA profile of the plasma phospholipid (PL) fraction. In the PREDIMED trial, we examined 1-year changes in the FA profile of plasma PL according to a nutritional intervention with Mediterranean diets, either supplemented with extra-virgin olive oil (MedDiet + EVOO) or mixed nuts (MedDiet + nuts), in a high cardiovascular risk population. We also analyzed if 1-year changes in PL FAs were associated with subsequent cardiovascular risk. METHODS We included 779 participants in our case-cohort study: 185 incident cases and 594 participants in the subcohort (including 31 overlapping cases). The end point was the incidence of CVD. We measured the FAs of plasma PL at baseline and after 1 year of intervention. RESULTS MedDiet + EVOO increased C17:0 and C20:3n9 in linear regression models [β coefficientperSD : 0.215 (95% CI, 0.032-0.399) and 0.271 (0.107-0.434), respectively] and decreased 16:1n7 and C22:4n6 [βperSD: -0.239 (95% CI, -0.416 to -0.061) and -0.287 (95% CI, -0.460 to -0.113), respectively] vs the control group. MedDiet + nuts increased C18:3n3 [βperSD: 0.382 (95% CI, 0.225 - 0.539)], C18:2n6 [βper SD: 0.250 (95% CI, 0.073 - 0.428)], C18:0 [βperSD: 0.268 (95% CI, 0.085-0.452)], and C22:0 [βper SD: 0.216 (95% CI, 0.031-0.402)]; and decreased the sum of six n6 FAs [βper SD: -0.147 (95% CI, -0.268 to -0.027)] vs the control group. The 1-year increase in C18:2n6 was inversely associated with the subsequent CVD risk (HRperSD: 0.64 (95% CI, 0.44-0.92)). CONCLUSIONS MedDiet interventions changed n6 FAs and C16:1n7c; other changes were specific for each group: MedDiet + EVOO increased C17:0 and C20:3n9, and MedDiet + Nuts C18:3n3, C18:2n6, C18:0, and C22:0 FAs.
Collapse
Affiliation(s)
- Cristina Razquin
- Department of Preventive Medicine and Public Health, University of Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Miguel Ruiz-Canela
- Department of Preventive Medicine and Public Health, University of Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Andreas Wernitz
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Estefania Toledo
- Department of Preventive Medicine and Public Health, University of Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Dolores Corella
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine, Universidad de Valencia, Valencia, Spain
| | - Ángel Alonso-Gómez
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, Hospital Universitario de Álava, Vitoria, Spain
| | - Montse Fitó
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Enrique Gómez-Gracia
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology, Virgen de la Victoria Hospital, Biomedical Research Institute of Málaga, University of Málaga, Málaga, Spain
| | - Ramón Estruch
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Clinic, Institut d'Investigacions Biomèdiques Augist Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Miquel Fiol
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Hospital Son Espases, Palma de Mallorca, Spain
| | - José Lapetra
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Family Medicine-Research Unit, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - Lluis Serra-Majem
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigaciones Biomédicas y Sanitarias, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Emilio Ros
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition, Agust Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Fernando Arós
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, Hospital Universitario de Álava, Vitoria, Spain
| | - Jordi Salas-Salvadó
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unitat de Nutrició Humana, Departament de Bioquimica i Biotecnologia, Universitat Rovira i Virgili, Reus, Spain
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- Unit of Cardiovascular Risk and Nutrition, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Miguel A Martinez-Gonzalez
- Department of Preventive Medicine and Public Health, University of Navarra, IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica En Red (CIBER), M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, USA
| |
Collapse
|
19
|
Burzynska-Pedziwiatr I, Dudzik D, Sansone A, Malachowska B, Zieleniak A, Zurawska-Klis M, Ferreri C, Chatgilialoglu C, Cypryk K, Wozniak LA, Markuszewski MJ, Bukowiecka-Matusiak M. Targeted and untargeted metabolomic approach for GDM diagnosis. Front Mol Biosci 2023; 9:997436. [PMID: 36685282 PMCID: PMC9849575 DOI: 10.3389/fmolb.2022.997436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is a disorder which manifests itself for the first time during pregnancy and is mainly connected with glucose metabolism. It is also known that fatty acid profile changes in erythrocyte membranes and plasma could be associated with obesity and insulin resistance. These factors can lead to the development of diabetes. In the reported study, we applied the untargeted analysis of plasma in GDM against standard glucose-tolerant (NGT) women to identify the differences in metabolomic profiles between those groups. We found higher levels of 2-hydroxybutyric and 3-hydroxybutyric acids. Both secondary metabolites are associated with impaired glucose metabolism. However, they are products of different metabolic pathways. Additionally, we applied lipidomic profiling using gas chromatography to examine the fatty acid composition of cholesteryl esters in the plasma of GDM patients. Among the 14 measured fatty acids characterizing the representative plasma lipidomic cluster, myristic, oleic, arachidonic, and α-linoleic acids revealed statistically significant changes. Concentrations of both myristic acid, one of the saturated fatty acids (SFAs), and oleic acid, which belong to monounsaturated fatty acids (MUFAs), tend to decrease in GDM patients. In the case of polyunsaturated fatty acids (PUFAs), some of them tend to increase (e.g., arachidonic), and some of them tend to decrease (e.g., α-linolenic). Based on our results, we postulate the importance of hydroxybutyric acid derivatives, cholesteryl ester composition, and the oleic acid diminution in the pathophysiology of GDM. There are some evidence suggests that the oleic acid can have the protective role in diabetes onset. However, metabolic alterations that lead to the onset of GDM are complex; therefore, further studies are needed to confirm our observations.
Collapse
Affiliation(s)
| | - Danuta Dudzik
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdansk, Gdansk, Poland
| | - Anna Sansone
- Consiglio Nazionale delle Ricerche, Institute for the Organic Synthesis and Photoreactivity, Bologna, Italy
| | - Beata Malachowska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland,Department of Nursing and Obstetrics, Medical University of Lodz, Lodz, Poland,Department of Clinic Nursing, Medical University of Lodz, Lodz, Poland,Department of Diabetology and Metabolic Diseases Lodz, Medical University of Lodz, Lodz, Poland
| | - Andrzej Zieleniak
- Laboratory of Metabolomic Studies, Department of Structural Biology, Medical University of Lodz, Lodz, Poland
| | - Monika Zurawska-Klis
- Department of Radiation Oncology, Einstein College of Medicine, Bronx, NY, United States
| | - Carla Ferreri
- Consiglio Nazionale delle Ricerche, Institute for the Organic Synthesis and Photoreactivity, Bologna, Italy
| | | | - Katarzyna Cypryk
- Department of Radiation Oncology, Einstein College of Medicine, Bronx, NY, United States
| | - Lucyna A. Wozniak
- Laboratory of Metabolomic Studies, Department of Structural Biology, Medical University of Lodz, Lodz, Poland
| | - Michal J. Markuszewski
- Department of Biopharmaceutics and Pharmacodynamics, Medical University of Gdansk, Gdansk, Poland
| | - Malgorzata Bukowiecka-Matusiak
- Laboratory of Metabolomic Studies, Department of Structural Biology, Medical University of Lodz, Lodz, Poland,*Correspondence: Malgorzata Bukowiecka-Matusiak,
| |
Collapse
|
20
|
Jones JL, Sabaté J, Heskey C, Oda K, Miles F, Rajaram S. Macadamia nut effects on cardiometabolic risk factors: a randomised trial. J Nutr Sci 2023; 12:e55. [PMID: 37180485 PMCID: PMC10173088 DOI: 10.1017/jns.2023.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 05/16/2023] Open
Abstract
We sought to examine the effects of daily consumption of macadamia nuts on body weight and composition, plasma lipids and glycaemic parameters in a free-living environment in overweight and obese adults at elevated cardiometabolic risk. Utilising a randomised cross-over design, thirty-five adults with abdominal obesity consumed their usual diet plus macadamia nuts (~15 % of daily calories) for 8 weeks (intervention) and their usual diet without nuts for 8 weeks (control), with a 2-week washout. Body composition was determined by bioelectrical impedance; dietary intake was assessed with 24-h dietary recalls. Consumption of macadamia nuts led to increased total fat and MUFA intake while SFA intake was unaltered. With mixed model regression analysis, no significant changes in mean weight, BMI, waist circumference, percent body fat or glycaemic parameters, and non-significant reductions in plasma total cholesterol of 2⋅1 % (-4⋅3 mg/dl; 95 % CI -14⋅8, 6⋅1) and low-density lipoprotein (LDL-C) of 4 % (-4⋅7 mg/dl; 95 % CI -14⋅3, 4⋅8) were observed. Cholesterol-lowering effects were modified by adiposity: greater lipid lowering occurred in those with overweight v. obesity, and in those with less than the median percent body fat. Daily consumption of macadamia nuts does not lead to gains in weight or body fat under free-living conditions in overweight or obese adults; non-significant cholesterol lowering occurred without altering saturated fat intake of similar magnitude to cholesterol lowering seen with other nuts. Clinical Trial Registry Number and Website: NCT03801837 https://clinicaltrials.gov/ct2/show/NCT03801837?term = macadamia + nut&draw = 2&rank = 1.
Collapse
Key Words
- % Body fat, percent body fat
- Adiposity
- Apo A1, apoprotein A1
- Apo B, apoprotein B
- BFM, body fat mass
- Body composition
- CV, coefficient of variation
- CVD, cardiovascular disease
- Cholesterol
- DLM, dry lean mass
- HOMA2, homeostasis model assessment 2
- IR, insulin resistance
- LBM, lean body mass
- LDL-C
- Mac, macadamia nuts
- Macadamia nuts
- Palmitoleic acid
- SMM, skeletal muscle mass
- TAG, triacylglycerol
- TBW, total body water
- TC, total cholesterol
- TEE, total energy expenditure
- VLDL,, very low-density lipoprotein
- WC, waist circumference
- oxLDL, oxidised LDL
- sdLDL, small dense low-density lipoprotein
- se, standard error
- sem, standard error of the mean
Collapse
Affiliation(s)
- Julie L. Jones
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Joan Sabaté
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Celine Heskey
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Keiji Oda
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Fayth Miles
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - Sujatha Rajaram
- School of Public Health, Loma Linda University, Loma Linda, CA, USA
- Corresponding author: Sujatha Rajaram, email
| |
Collapse
|
21
|
Li KJ, Brouwer-Brolsma EM, Fleuti C, Badertscher R, Vergères G, Feskens EJM, Burton-Pimentel KJ. Associations between dairy fat intake, milk-derived free fatty acids, and cardiometabolic risk in Dutch adults. Eur J Nutr 2023; 62:185-198. [PMID: 35931833 PMCID: PMC9899750 DOI: 10.1007/s00394-022-02974-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 07/25/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Milk-derived free fatty acids (FFAs) may act as both biomarkers of intake and metabolic effect. In this study we explored associations between different types of dairy consumption, a selection of milk-derived free fatty acids, and cardiometabolic disease (CMD) risk factors. METHODS Sixty-seven FFAs were quantified in the plasma of 131 free-living Dutch adults (median 60 years) using gas chromatography-flame ionization detector. Intakes of different dairy foods and groups were assessed using a food frequency questionnaire. Twelve different CMD risk factors were analyzed. Multiple linear regressions were used to evaluate the associations under study. RESULTS Based on the fully adjusted models, 5 long-chain unsaturated FFAs (C18:1 t13 + c6 + c7 + u, C18:2 c9t11 + u, C20:1 c11, C20:3 c8c11c14, and C20:4 c5c8c11c14), 2 medium-chain saturated FFAs (C15, C15 iso), and a trans FFA (C16:1 t9) were positively associated with at least one variable of dairy intake, as well as plasma total and LDL cholesterol, blood pressure, and SCORE (p ≤ 0.05). A long-chain PUFA associated with high-fat fermented dairy intake (C18:2 t9t12), was negatively associated with serum triglyceride levels, and a long-chain saturated FFA associated with cheese intake (C18:1 u1) was negatively associated with plasma LDL cholesterol and serum triglyceride levels. No clear associations were observed between dairy intake and CMD risk factors. CONCLUSION Milk-derived FFAs could act as sensitive biomarkers for dairy intake and metabolism, allowing the association between dairy and CMD risk to be more precisely evaluated.
Collapse
Affiliation(s)
- Katherine J. Li
- grid.4818.50000 0001 0791 5666Division of Human Nutrition and Health, Department of Agrotechnology and Food Science, Wageningen University & Research, Wageningen, The Netherlands ,grid.484687.1 0000 0001 1457 2921Agroscope, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Bern, Switzerland
| | - Elske M. Brouwer-Brolsma
- grid.4818.50000 0001 0791 5666Division of Human Nutrition and Health, Department of Agrotechnology and Food Science, Wageningen University & Research, Wageningen, The Netherlands
| | - Charlotte Fleuti
- grid.484687.1 0000 0001 1457 2921Agroscope, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Bern, Switzerland
| | - René Badertscher
- grid.484687.1 0000 0001 1457 2921Agroscope, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Bern, Switzerland
| | - Guy Vergères
- grid.484687.1 0000 0001 1457 2921Agroscope, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Bern, Switzerland
| | - Edith J. M. Feskens
- grid.4818.50000 0001 0791 5666Division of Human Nutrition and Health, Department of Agrotechnology and Food Science, Wageningen University & Research, Wageningen, The Netherlands
| | - Kathryn J. Burton-Pimentel
- grid.484687.1 0000 0001 1457 2921Agroscope, Federal Department of Economic Affairs, Education and Research (EAER), Federal Office for Agriculture (FOAG), Bern, Switzerland
| |
Collapse
|
22
|
Reiterer M, Gilani A, Lo JC. Pancreatic Islets as a Target of Adipokines. Compr Physiol 2022; 12:4039-4065. [PMID: 35950650 DOI: 10.1002/cphy.c210044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Rising rates of obesity are intricately tied to the type 2 diabetes epidemic. The adipose tissues can play a central role in protection against or triggering metabolic diseases through the secretion of adipokines. Many adipokines may improve peripheral insulin sensitivity through a variety of mechanisms, thereby indirectly reducing the strain on beta cells and thus improving their viability and functionality. Such effects will not be the focus of this article. Rather, we will focus on adipocyte-secreted molecules that have a direct effect on pancreatic islets. By their nature, adipokines represent potential druggable targets that can reach the islets and improve beta-cell function or preserve beta cells in the face of metabolic stress. © 2022 American Physiological Society. Compr Physiol 12:1-27, 2022.
Collapse
Affiliation(s)
- Moritz Reiterer
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - James C Lo
- Division of Cardiology, Department of Medicine, Weill Center for Metabolic Health, Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
23
|
Luo M, Guo J, Lu W, Fang X, Zhang R, Tang M, Luo Q, Liang W, Yu X, Hu C. The mediating role of maternal metabolites between lipids and adverse pregnancy outcomes of gestational diabetes mellitus. Front Med (Lausanne) 2022; 9:925602. [PMID: 36035400 PMCID: PMC9400014 DOI: 10.3389/fmed.2022.925602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common complications of pregnancy, and the demographics of pregnant women have changed in recent decades. GDM is a metabolic disease with short- and long-term adverse effects on both pregnant women and newborns. The metabolic changes and corresponding risk factors should be of great significance in understanding the pathological mechanism of GDM and reducing the incidence of adverse pregnancy outcomes in patients with GDM. The well-known GDM-associated lipids used in clinical tests, such as triglyceride (TG), are thought to play a major role in metabolic changes during GDM, which have a potential causal relationship with abnormal pregnancy outcomes of GDM. Therefore, this study analyzed the relationship between clinical lipid indicators, metabolic profiles, and abnormal pregnancy outcomes in GDM through mediation analysis. By constructing a metabolic atlas of 399 samples from GDM patients in different trimesters, we efficiently detected the key metabolites of adverse pregnancy outcomes and their mediating roles in bridging abnormal lipids and adverse pregnancy outcomes in patients with GDM. Our study confirmed that TG and total cholesterol were independent risk factors for adverse pregnancy outcomes in patients with GDM. Several key metabolites as mediators (e.g., gamma-linolenic acid, heptadecanoic acid, oleic acid, palmitic acid, and palmitoleic acid) have been identified as potential biomarkers for adverse pregnancy outcomes in patients with GDM. These metabolites mainly participate in the biosynthesis of unsaturated fatty acids, which may shed new light on the pathology of GDM and provide insights for further exploration of the molecular mechanisms underlying adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Mingjuan Luo
- Department of Endocrinology and Metabolism, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Jingyi Guo
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenqian Lu
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Xiangnan Fang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
- Department of Endocrinology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mengyang Tang
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
| | - Qiong Luo
- Department of Obstetrics and Gynecology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wei Liang
- Department of Endocrinology and Metabolism, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiangtian Yu
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- *Correspondence: Xiangtian Yu
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to the Southern Medical University, Shanghai, China
- Shanghai Diabetes Institute, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- Cheng Hu
| |
Collapse
|
24
|
Trans-palmitoleic acid prevents weight gain, but does not modify glucose homeostasis in a rodent model of diet-induced obesity. CLINICAL NUTRITION OPEN SCIENCE 2022. [DOI: 10.1016/j.nutos.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
25
|
Roles of Palmitoleic Acid and Its Positional Isomers, Hypogeic and Sapienic Acids, in Inflammation, Metabolic Diseases and Cancer. Cells 2022; 11:cells11142146. [PMID: 35883589 PMCID: PMC9319324 DOI: 10.3390/cells11142146] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
In the last few years, the monounsaturated hexadecenoic fatty acids are being increasingly considered as biomarkers of health with key functions in physiology and pathophysiology. Palmitoleic acid (16:1n-7) and sapienic acid (16:1n-10) are synthesized from palmitic acid by the action of stearoyl-CoA desaturase-1 and fatty acid desaturase 2, respectively. A third positional isomer, hypogeic acid (16:1n-9) is produced from the partial β-oxidation of oleic acid. In this review, we discuss the current knowledge of the effects of palmitoleic acid and, where available, sapienic acid and hypogeic acid, on metabolic diseases such as diabetes, cardiovascular disease, and nonalcoholic fatty liver disease, and cancer. The results have shown diverse effects among studies in cell lines, animal models and humans. Palmitoleic acid was described as a lipokine able to regulate different metabolic processes such as an increase in insulin sensitivity in muscle, β cell proliferation, prevention of endoplasmic reticulum stress and lipogenic activity in white adipocytes. Numerous beneficial effects have been attributed to palmitoleic acid, both in mouse models and in cell lines. However, its role in humans is not fully understood, and is sometimes controversial. Regarding sapienic acid and hypogeic acid, studies on their biological effects are still scarce, but accumulating evidence suggests that they also play important roles in metabolic regulation. The multiplicity of effects reported for palmitoleic acid and the compartmentalized manner in which they often occur, may suggest the overlapping actions of multiple isomers being present at the same or neighboring locations.
Collapse
|
26
|
De Fano M, Bartolini D, Tortoioli C, Vermigli C, Malara M, Galli F, Murdolo G. Adipose Tissue Plasticity in Response to Pathophysiological Cues: A Connecting Link between Obesity and Its Associated Comorbidities. Int J Mol Sci 2022; 23:ijms23105511. [PMID: 35628322 PMCID: PMC9141504 DOI: 10.3390/ijms23105511] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
Adipose tissue (AT) is a remarkably plastic and active organ with functional pleiotropism and high remodeling capacity. Although the expansion of fat mass, by definition, represents the hallmark of obesity, the dysregulation of the adipose organ emerges as the forefront of the link between adiposity and its associated metabolic and cardiovascular complications. The dysfunctional fat displays distinct biological signatures, which include enlarged fat cells, low-grade inflammation, impaired redox homeostasis, and cellular senescence. While these events are orchestrated in a cell-type, context-dependent and temporal manner, the failure of the adipose precursor cells to form new adipocytes appears to be the main instigator of the adipose dysregulation, which, ultimately, poses a deleterious milieu either by promoting ectopic lipid overspill in non-adipose targets (i.e., lipotoxicity) or by inducing an altered secretion of different adipose-derived hormones (i.e., adipokines and lipokines). This “adipocentric view” extends the previous “expandability hypothesis”, which implies a reduced plasticity of the adipose organ at the nexus between unhealthy fat expansion and the development of obesity-associated comorbidities. In this review, we will briefly summarize the potential mechanisms by which adaptive changes to variations of energy balance may impair adipose plasticity and promote fat organ dysfunction. We will also highlight the conundrum with the perturbation of the adipose microenvironment and the development of cardio-metabolic complications by focusing on adipose lipoxidation, inflammation and cellular senescence as a novel triad orchestrating the conspiracy to adipose dysfunction. Finally, we discuss the scientific rationale for proposing adipose organ plasticity as a target to curb/prevent adiposity-linked cardio-metabolic complications.
Collapse
Affiliation(s)
- Michelatonio De Fano
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Desirèe Bartolini
- Department of Pharmaceutical Sciences, Human Anatomy Laboratory, University of Perugia, 06132 Perugia, Italy; (D.B.); (F.G.)
| | - Cristina Tortoioli
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Cristiana Vermigli
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Massimo Malara
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
| | - Francesco Galli
- Department of Pharmaceutical Sciences, Human Anatomy Laboratory, University of Perugia, 06132 Perugia, Italy; (D.B.); (F.G.)
| | - Giuseppe Murdolo
- Department of Internal Medicine, Endocrinology and Metabolism, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, Piazzale Gambuli, 06081 Perugia, Italy; (M.D.F.); (C.T.); (C.V.); (M.M.)
- Correspondence: ; Tel.: +39-(0)75-578-3301; Fax: +39-75-573-0855
| |
Collapse
|
27
|
Maternal and Fetal Metabolites in Gestational Diabetes Mellitus: A Narrative Review. Metabolites 2022; 12:metabo12050383. [PMID: 35629887 PMCID: PMC9143359 DOI: 10.3390/metabo12050383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 02/05/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is a major public health issue of our century due to its increasing prevalence, affecting 5% to 20% of all pregnancies. The pathogenesis of GDM has not been completely elucidated to date. Increasing evidence suggests the association of environmental factors with genetic and epigenetic factors in the development of GDM. So far, several metabolomics studies have investigated metabolic disruptions associated with GDM. The aim of this review is to highlight the usefulness of maternal metabolites as diagnosis markers of GDM as well as the importance of both maternal and fetal metabolites as prognosis biomarkers for GDM and GDM’s transition to type 2 diabetes mellitus T2DM.
Collapse
|
28
|
Chen Y, Li EM, Xu LY. Guide to Metabolomics Analysis: A Bioinformatics Workflow. Metabolites 2022; 12:357. [PMID: 35448542 PMCID: PMC9032224 DOI: 10.3390/metabo12040357] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/12/2022] [Accepted: 04/14/2022] [Indexed: 02/05/2023] Open
Abstract
Metabolomics is an emerging field that quantifies numerous metabolites systematically. The key purpose of metabolomics is to identify the metabolites corresponding to each biological phenotype, and then provide an analysis of the mechanisms involved. Although metabolomics is important to understand the involved biological phenomena, the approach's ability to obtain an exhaustive description of the processes is limited. Thus, an analysis-integrated metabolomics, transcriptomics, proteomics, and other omics approach is recommended. Such integration of different omics data requires specialized statistical and bioinformatics software. This review focuses on the steps involved in metabolomics research and summarizes several main tools for metabolomics analyses. We also outline the most abnormal metabolic pathways in several cancers and diseases, and discuss the importance of multi-omics integration algorithms. Overall, our goal is to summarize the current metabolomics analysis workflow and its main analysis software to provide useful insights for researchers to establish a preferable pipeline of metabolomics or multi-omics analysis.
Collapse
Affiliation(s)
- Yang Chen
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041,
| |
Collapse
|
29
|
Chen G, Guo L, Zhao X, Ren Y, Chen H, Liu J, Jiang J, Liu P, Liu X, Hu B, Wang N, Peng H, Xu G, Tao H. Serum Metabonomics Reveals Risk Factors in Different Periods of Cerebral Infarction in Humans. Front Mol Biosci 2022; 8:784288. [PMID: 35242810 PMCID: PMC8887861 DOI: 10.3389/fmolb.2021.784288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/31/2021] [Indexed: 12/26/2022] Open
Abstract
Studies of key metabolite variations and their biological mechanisms in cerebral infarction (CI) have increased our understanding of the pathophysiology of the disease. However, how metabolite variations in different periods of CI influence these biological processes and whether key metabolites from different periods may better predict disease progression are still unknown. We performed a systematic investigation using the metabonomics method. Various metabolites in different pathways were investigated by serum metabolic profiling of 143 patients diagnosed with CI and 59 healthy controls. Phe-Phe, carnitine C18:1, palmitic acid, cis-8,11,14-eicosatrienoic acid, palmitoleic acid, 1-linoleoyl-rac-glycerol, MAG 18:1, MAG 20:3, phosphoric acid, 5α-dihydrotestosterone, Ca, K, and GGT were the major components in the early period of CI. GCDCA, glycocholate, PC 36:5, LPC 18:2, and PA showed obvious changes in the intermediate time. In contrast, trans-vaccenic acid, linolenic acid, linoleic acid, all-cis-4,7,10,13,16-docosapentaenoic acid, arachidonic acid, DHA, FFA 18:1, FFA 18:2, FFA 18:3, FFA 20:4, FFA 22:6, PC 34:1, PC 36:3, PC 38:4, ALP, and Crea displayed changes in the later time. More importantly, we found that phenylalanine metabolism, medium-chain acylcarnitines, long-chain acylcarnitines, choline, DHEA, LPC 18:0, LPC 18:1, FFA 18:0, FFA 22:4, TG, ALB, IDBIL, and DBIL played vital roles in the development of different periods of CI. Increased phenylacetyl-L-glutamine was detected and may be a biomarker for CI. It was of great significance that we identified key metabolic pathways and risk metabolites in different periods of CI different from those previously reported. Specific data are detailed in the Conclusion section. In addition, we also explored metabolite differences of CI patients complicated with high blood glucose compared with healthy controls. Further work in this area may inform personalized treatment approaches in clinical practice for CI by experimentally elucidating the pathophysiological mechanisms.
Collapse
Affiliation(s)
- Guoyou Chen
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Li Guo
- Department of Anesthesia, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Xinjie Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yachao Ren
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Hongyang Chen
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Jincheng Liu
- Academic Affairs Office, Harbin Medical University-Daqing, Daqing, China
| | - Jiaqi Jiang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Peijia Liu
- Department of Clinical Laboratory, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoying Liu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Bo Hu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Na Wang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Haisheng Peng
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Guowang Xu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Haiquan Tao
- Department of Neurosurgery, Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Cerebrovascular Diseases Department, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
30
|
Yang C, Wei J, Cao G, Cai Z. Lipid metabolism dysfunction and toxicity of BDE-47 exposure in white adipose tissue revealed by the integration of lipidomics and metabolomics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150350. [PMID: 34555606 DOI: 10.1016/j.scitotenv.2021.150350] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 06/13/2023]
Abstract
2,2',4,4'-Tetrabromodiphenyl ether (BDE-47) is one of the main and toxic congeners of polybrominated diphenyl ether (PBDE) family and considered to be associated with the development of obesity. However, little is known about its direct metabolic alterations on white adipose tissue (WAT). In this study, we evaluated the impacts of BDE-47 exposure on WAT dysfunctions in mice fed with low-fat diet (LFD) or high-fat diet (HFD) by the integration analysis of mass spectrometry-based metabolomics and lipidomics. The results showed that BDE-47 exposure together with HFD intervention induced adipocyte hypertrophy and accelerated the weight gain of WAT, whereas no obvious effects were observed in mice fed with LFD. The combination of BDE-47 and HFD induced intolerable levels of metabolites in purine and glutathione metabolism pathways, sufficient to increase oxidative stress in WAT. Importantly, continuous exposure of BDE-47 in HFD -fed mice caused lipid metabolism dysfunction by promoting fatty acid uptake and de novo synthesis and suppressing β-oxidation, ultimately leading to the accumulation of saturated fatty acids, triglycerides in WAT. At the same time, BDE-47 increased inflammatory infiltration into WAT, consequently promoting the productions of cytokines, TNFα and IL-6, in HFD fed mice. It is found that dysfunction of lipid metabolism and increasing inflammation led to lipotoxicity in WAT and severe obesity in HFD mice. Taken together, our findings deepen the understanding of the obesogenic effect of BDE-47 and help identify new potential strategies for clarifying the molecular and metabolic mechanisms.
Collapse
Affiliation(s)
- Chunxue Yang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Juntong Wei
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Guodong Cao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
31
|
Betz IR, Qaiyumi SJ, Goeritzer M, Thiele A, Brix S, Beyhoff N, Grune J, Klopfleisch R, Greulich F, Uhlenhaut NH, Kintscher U, Foryst-Ludwig A. Cardioprotective Effects of Palmitoleic Acid (C16:1n7) in a Mouse Model of Catecholamine-Induced Cardiac Damage Are Mediated by PPAR Activation. Int J Mol Sci 2021; 22:ijms222312695. [PMID: 34884498 PMCID: PMC8657733 DOI: 10.3390/ijms222312695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 01/25/2023] Open
Abstract
Palmitoleic acid (C16:1n7) has been identified as a regulator of physiological cardiac hypertrophy. In the present study, we aimed to investigate the molecular pathways involved in C16:1n7 responses in primary murine cardiomyocytes (PCM) and a mouse model of isoproterenol (ISO)-induced cardiac damage. PCMs were stimulated with C16:1n7 or a vehicle. Afterwards, RNA sequencing was performed using an Illumina HiSeq sequencer. Confirmatory analysis was performed in PCMs and HL-1 cardiomyocytes. For an in vivo study, 129 sv mice were orally treated with a vehicle or C16:1n7 for 22 days. After 5 days of pre-treatment, the mice were injected with ISO (25 mg/kg/d s. c.) for 4 consecutive days. Cardiac phenotyping was performed using echocardiography. In total, 129 genes were differentially expressed in PCMs stimulated with C16:1n7, including Angiopoietin-like factor 4 (Angptl4) and Pyruvate Dehydrogenase Kinase 4 (Pdk4). Both Angptl4 and Pdk4 are proxisome proliferator-activated receptor α/δ (PPARα/δ) target genes. Our in vivo results indicated cardioprotective and anti-fibrotic effects of C16:1n7 application in mice. This was associated with the C16:1n7-dependent regulation of the cardiac PPAR-specific signaling pathways. In conclusion, our experiments demonstrated that C16:1n7 might have protective effects on cardiac fibrosis and inflammation. Our study may help to develop future lipid-based therapies for catecholamine-induced cardiac damage.
Collapse
Affiliation(s)
- Iris Rosa Betz
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- Berlin Institute of Health, Emergency Department Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Sarah Julia Qaiyumi
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
| | - Madeleine Goeritzer
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Arne Thiele
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Sarah Brix
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Niklas Beyhoff
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Jana Grune
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany;
| | - Franziska Greulich
- German Center for Environmental Health GmbH, Institute for Diabetes and Cancer (IDC), 85764 Munich, Germany; (F.G.); (N.H.U.)
- Metabolic Programming, School of Life Sciences Weihenstephan, Technische Universitaet Muenchen (TUM), 85354 Freising, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, 13125 Berlin, Germany
| | - Nina Henriette Uhlenhaut
- German Center for Environmental Health GmbH, Institute for Diabetes and Cancer (IDC), 85764 Munich, Germany; (F.G.); (N.H.U.)
- Metabolic Programming, School of Life Sciences Weihenstephan, Technische Universitaet Muenchen (TUM), 85354 Freising, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, 13125 Berlin, Germany
| | - Ulrich Kintscher
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
| | - Anna Foryst-Ludwig
- Center for Cardiovascular Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10115 Berlin, Germany; (I.R.B.); (S.J.Q.); (M.G.); (A.T.); (S.B.); (N.B.); (J.G.); (U.K.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10785 Berlin, Germany
- Correspondence:
| |
Collapse
|
32
|
Smith CC, Sheedy DL, McEwen HP, Don AS, Kril JJ, Sutherland GT. Lipidome changes in alcohol-related brain damage. J Neurochem 2021; 160:271-282. [PMID: 34699608 DOI: 10.1111/jnc.15530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 10/13/2021] [Accepted: 10/13/2021] [Indexed: 11/30/2022]
Abstract
Alcohol-related brain injury is characterized by cognitive deficits and brain atrophy with the prefrontal cortex particularly susceptible. White matter in the human brain is lipid rich and a major target of damage from chronic alcohol abuse; yet, there is sparse information on how these lipids are affected. Here, we used untargeted lipidomics as a discovery tool to describe these changes in the prefrontal, middle temporal, and visual cortices of human subjects with alcohol use disorder and controls. Significant changes to the lipidome, predominantly in the prefrontal and visual cortices, and differences between the white and grey matter of each brain region were identified. These effects include broad decreases to phospholipids and ceramide, decreased polyunsaturated fatty acids, decreased sphingadiene backbones, and selective decreases in cholesteryl ester fatty acid chains. Our findings show that chronic alcohol abuse results in selective changes to the neurolipidome, which likely reflects both the directs effects on the brain and concurrent effects on the liver.
Collapse
Affiliation(s)
- Caine C Smith
- Faculty of Medicine and Health, School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Donna L Sheedy
- Faculty of Medicine and Health, School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Holly P McEwen
- Centenery Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony S Don
- Faculty of Medicine and Health, School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Centenery Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Jillian J Kril
- Faculty of Medicine and Health, School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Greg T Sutherland
- Faculty of Medicine and Health, School of Medical Sciences and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
33
|
Castor K, Dawlaty J, Arakaki X, Gross N, Woldeamanuel YW, Harrington MG, Cowan RP, Fonteh AN. Plasma Lipolysis and Changes in Plasma and Cerebrospinal Fluid Signaling Lipids Reveal Abnormal Lipid Metabolism in Chronic Migraine. Front Mol Neurosci 2021; 14:691733. [PMID: 34531722 PMCID: PMC8438335 DOI: 10.3389/fnmol.2021.691733] [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: 04/07/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Background Lipids are a primary storage form of energy and the source of inflammatory and pain signaling molecules, yet knowledge of their importance in chronic migraine (CM) pathology is incomplete. We aim to determine if plasma and cerebrospinal fluid (CSF) lipid metabolism are associated with CM pathology. Methods We obtained plasma and CSF from healthy controls (CT, n = 10) or CM subjects (n = 15) diagnosed using the International Headache Society criteria. We measured unesterified fatty acid (UFA) and esterified fatty acids (EFAs) using gas chromatography-mass spectrometry. Glycerophospholipids (GP) and sphingolipid (SP) levels were determined using LC-MS/MS, and phospholipase A2 (PLA2) activity was determined using fluorescent substrates. Results Unesterified fatty acid levels were significantly higher in CM plasma but not in CSF. Unesterified levels of five saturated fatty acids (SAFAs), eight monounsaturated fatty acids (MUFAs), five ω-3 polyunsaturated fatty acids (PUFAs), and five ω-6 PUFAs are higher in CM plasma. Esterified levels of three SAFAs, eight MUFAs, five ω-3 PUFAs, and three ω-6 PUFAs, are higher in CM plasma. The ratios C20:4n-6/homo-γ-C20:3n-6 representative of delta-5-desaturases (D5D) and the elongase ratio are lower in esterified and unesterified CM plasma, respectively. In the CSF, the esterified D5D index is lower in CM. While PLA2 activity was similar, the plasma UFA to EFA ratio is higher in CM. Of all plasma GP/SPs detected, only ceramide levels are lower (p = 0.0003) in CM (0.26 ± 0.07%) compared to CT (0.48 ± 0.06%). The GP/SP proportion of platelet-activating factor (PAF) is significantly lower in CM CSF. Conclusions Plasma and CSF lipid changes are consistent with abnormal lipid metabolism in CM. Since plasma UFAs correspond to diet or adipose tissue levels, higher plasma fatty acids and UFA/EFA ratios suggest enhanced adipose lipolysis in CM. Differences in plasma and CSF desaturases and elongases suggest altered lipid metabolism in CM. A lower plasma ceramide level suggests reduced de novo synthesis or reduced sphingomyelin hydrolysis. Changes in CSF PAF suggest differences in brain lipid signaling pathways in CM. Together, this pilot study shows lipid metabolic abnormality in CM corresponding to altered energy homeostasis. We propose that controlling plasma lipolysis, desaturases, elongases, and lipid signaling pathways may relieve CM symptoms.
Collapse
Affiliation(s)
- Katherine Castor
- Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States
| | - Jessica Dawlaty
- Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States
| | - Xianghong Arakaki
- Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Noah Gross
- Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States
| | | | - Michael G Harrington
- Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
| | - Robert P Cowan
- Pain Center, Department of Neurology, Stanford University, Stanford, CA, United States
| | - Alfred N Fonteh
- Department of Neurosciences, Huntington Medical Research Institutes, Pasadena, CA, United States.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
34
|
Huang NK, Bůžková P, Matthan NR, Djoussé L, Kizer JR, Mukamal KJ, Polak JF, Lichtenstein AH. Serum Non-Esterified Fatty Acids, Carotid Artery Intima-Media Thickness and Flow-Mediated Dilation in Older Adults: The Cardiovascular Health Study (CHS). Nutrients 2021; 13:nu13093052. [PMID: 34578930 PMCID: PMC8465602 DOI: 10.3390/nu13093052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 12/24/2022] Open
Abstract
Backgrounds and aims: Elevated common carotid artery intima-media thickness (carotid IMT) and diminished flow-mediated dilation (FMD) are early subclinical indicators of atherosclerosis. Serum total non-esterified fatty acid (NEFA) concentrations have been positively associated with subclinical atherosclerosis. The relations between individual NEFA, carotid IMT and FMD have as yet to be assessed. Methods: We investigated the associations between fasting serum individual NEFA, carotid IMT and FMD among Cardiovascular Health Study (CHS) participants with (n = 255 for carotid IMT, 301 for FMD) or without (n = 1314 for carotid IMT, 1462 for FMD) known atherosclerotic cardiovascular disease (ASCVD). Using archived samples (fasting) collected from 1996-1997 (baseline), 35 individual NEFAs were measured using gas chromatography. Carotid IMT and estimated plaque thickness (mean of maximum internal carotid IMT) were determined in 1998-1999. FMD was measured in 1997-1998. Linear regression adjusted by the Holm-Bonferroni method was used to assess relations between individual NEFA, carotid IMT and FMD. Results: In multivariable adjusted linear regression models per SD increment, the non-esterified trans fatty acid conjugated linoleic acid (trans-18:2 CLA) was positively associated with carotid IMT [β (95% CI): 44.8 (19.2, 70.4), p = 0.025] among participants with, but not without, ASCVD [2.16 (-6.74, 11.5), p = 1.000]. Non-esterified cis-palmitoleic acid (16:1n-7c) was positively associated with FMD [19.7 (8.34, 31.0), p = 0.024] among participants without, but not with ASCVD. No significant associations between NEFAs and estimated plaque thickness were observed. Conclusions: In older adults, serum non-esterified CLA and palmitoleic acid were positively associated with carotid IMT and FMD, respectively, suggesting potential modifiable biomarkers for arteriopathy.
Collapse
Grants
- HHSN268201200036C, HHSN268200800007C, HHSN268201800001C, N01HC55222, N01HC85079, N01HC85080, N01HC85081, N01HC85082, N01HC85083, N01HC85086, and grants U01HL080295 and U01HL130114 NHLBI NIH HHS
- R01AG023629, R01AG053325, and K24AG065525 NIA NIH HHS
Collapse
Affiliation(s)
- Neil K. Huang
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (N.K.H.); (N.R.M.)
| | - Petra Bůžková
- Department of Biostatistics, University of Washington, Seattle, WA 98115, USA;
| | - Nirupa R. Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (N.K.H.); (N.R.M.)
| | - Luc Djoussé
- Division of Aging, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Jorge R. Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA;
- Department of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kenneth J. Mukamal
- Beth Israel Deaconess Medical Center, Division of General Medicine, Boston, MA 02215, USA;
| | - Joseph F. Polak
- Department of Radiology, Tufts Medical Center, Boston, MA 02111, USA;
| | - Alice H. Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA; (N.K.H.); (N.R.M.)
- Correspondence: ; Tel.: +1-(617)-556-3127
| |
Collapse
|
35
|
Wang QY, You LH, Xiang LL, Zhu YT, Zeng Y. Current progress in metabolomics of gestational diabetes mellitus. World J Diabetes 2021; 12:1164-1186. [PMID: 34512885 PMCID: PMC8394228 DOI: 10.4239/wjd.v12.i8.1164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/20/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is one of the most common metabolic disorders of pregnancy and can cause short- and long-term adverse effects in both pregnant women and their offspring. However, the etiology and pathogenesis of GDM are still unclear. As a metabolic disease, GDM is well suited to metabolomics study, which can monitor the changes in small molecular metabolites induced by maternal stimuli or perturbations in real time. The application of metabolomics in GDM can be used to discover diagnostic biomarkers, evaluate the prognosis of the disease, guide the application of diet or drugs, evaluate the curative effect, and explore the mechanism. This review provides comprehensive documentation of metabolomics research methods and techniques as well as the current progress in GDM research. We anticipate that the review will contribute to identifying gaps in the current knowledge or metabolomics technology, provide evidence-based information, and inform future research directions in GDM.
Collapse
Affiliation(s)
- Qian-Yi Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 21000, Jiangsu Province, China
| | - Liang-Hui You
- Nanjing Maternity and Child Health Care Institute, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Lan-Lan Xiang
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Yi-Tian Zhu
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| | - Yu Zeng
- Department of Clinical Laboratory, Women’s Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 21000, Jiangsu Province, China
| |
Collapse
|
36
|
Akazawa Y, Morisaki T, Fukuda H, Norimatsu K, Shiota J, Hashiguchi K, Tabuchi M, Kitayama M, Matsushima K, Yamaguchi N, Kondo H, Fujita F, Takeshita H, Nakao K, Takeshima F. Significance of serum palmitoleic acid levels in inflammatory bowel disease. Sci Rep 2021; 11:16260. [PMID: 34376800 PMCID: PMC8355139 DOI: 10.1038/s41598-021-95923-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/28/2021] [Indexed: 01/22/2023] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis (UC) and Crohn’s disease (CD), are chronic intestinal diseases of unknown etiology that present with variable disease extents and outcomes. The use of biomarkers for the diagnosis and management of IBDs is considered beneficial. Palmitoleic acid (PO) is an adipose tissue-derived mono-unsaturated free fatty acid that potentially serves as a lipokine in metabolic and inflammatory diseases. The aim of this study was to investigate the significance of PO levels in the serum of patients with UC and CD. The study included patients with UC (n = 22), patients with CD (n = 35), and controls (n = 22). The levels of serum PO were analyzed using gas chromatography. The association of serum PO levels with the clinical features and disease outcomes in IBD was examined. Serum PO levels were significantly higher in patients with CD than in controls, whereas no difference in these levels was observed between patients with UC and controls. Serum PO levels were significantly associated with the CD activity index. Additionally, high serum PO levels were associated with an increased risk of surgical intervention requirement during follow-up. In a pilot study with a few patients, high PO levels were observed in the mesenteric tissue in the active disease site of patients with CD (n = 7) compared with those with colon cancer (n = 6). Elevated serum PO levels might serve as a marker for local inflammation and prognosis in patients with CD.
Collapse
Affiliation(s)
- Yuko Akazawa
- Tissue and Histopathology Section, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan.
| | | | | | - Kiyuu Norimatsu
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Junya Shiota
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Keiichi Hashiguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Maiko Tabuchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Moto Kitayama
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kayoko Matsushima
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoyuki Yamaguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hisayoshi Kondo
- Biostatistics Section, Division of Scientific Data Registry, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | | | | | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Fuminao Takeshima
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Nagasaki Goto Chuoh Hospital, Goto, Japan
| |
Collapse
|
37
|
Dilworth L, Facey A, Omoruyi F. Diabetes Mellitus and Its Metabolic Complications: The Role of Adipose Tissues. Int J Mol Sci 2021; 22:ijms22147644. [PMID: 34299261 PMCID: PMC8305176 DOI: 10.3390/ijms22147644] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Many approaches have been used in the effective management of type 2 diabetes mellitus. A recent paradigm shift has focused on the role of adipose tissues in the development and treatment of the disease. Brown adipose tissues (BAT) and white adipose tissues (WAT) are the two main types of adipose tissues with beige subsets more recently identified. They play key roles in communication and insulin sensitivity. However, WAT has been shown to contribute significantly to endocrine function. WAT produces hormones and cytokines, collectively called adipocytokines, such as leptin and adiponectin. These adipocytokines have been proven to vary in conditions, such as metabolic dysfunction, type 2 diabetes, or inflammation. The regulation of fat storage, energy metabolism, satiety, and insulin release are all features of adipose tissues. As such, they are indicators that may provide insights on the development of metabolic dysfunction or type 2 diabetes and can be considered routes for therapeutic considerations. The essential roles of adipocytokines vis-a-vis satiety, appetite, regulation of fat storage and energy, glucose tolerance, and insulin release, solidifies adipose tissue role in the development and pathogenesis of diabetes mellitus and the complications associated with the disease.
Collapse
Affiliation(s)
- Lowell Dilworth
- Department of Pathology, Mona Campus, University of the West Indies, Kingston 7, Jamaica;
| | - Aldeam Facey
- Mona Academy of Sport, Mona Campus, University of the West Indies, Kingston 7, Jamaica;
| | - Felix Omoruyi
- Department of Life Sciences, Texas A&M University, Corpus Christi, TX 78412, USA
- Correspondence:
| |
Collapse
|
38
|
Tang J, Yang B, Yan Y, Tong W, Zhou R, Zhang J, Mi J, Li D. Palmitoleic Acid Protects against Hypertension by Inhibiting NF-κB-Mediated Inflammation. Mol Nutr Food Res 2021; 65:e2001025. [PMID: 33865240 DOI: 10.1002/mnfr.202001025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/25/2021] [Indexed: 11/08/2022]
Abstract
SCOPE The role of palmitoleic acid (POA) in hypertension or blood pressure remains uncertain. This study aims to investigate the epidemiological association between circulating POA and primary hypertension in humans, and subsequently evaluate the effects of exogenous POA on blood pressure and aortic remodeling in spontaneously hypertensive rats (SHRs). METHODS AND RESULTS A case-control study of 349 hypertensive and 1396 normotensive children and adolescents is conducted, and found hypertensive cases show significant lower erythrocyte phospholipid POA than normotensive controls (p < 0.001). In conditional logistic regression model, participants in the top quartile of POA have a lower prevalence of primary hypertension than those in the bottom (multivariate-adjusted OR: 0.47, 95% CI: 0.25-0.89). In animal study, 24 SHRs are randomly assigned to n-3 PUFAs (500 mg kg-1 ), POA (500 mg kg-1 ), or vehicle (olive oil) for 8 weeks. At the end of intervention, as compared to SHRs treated with vehicle, SHRs treated with POA shows significantly decreased systolic blood pressure (SBP), improved aortic remodeling, and also decreased aortic expressions of NF-κB and its downstream proinflammatory cytokines. CONCLUSIONS Circulating POA is inversely associated with risk of primary hypertension, and exogenous POA supplementation can decrease SBP and improve aortic remodeling by inhibiting NF-κB-mediated inflammation.
Collapse
Affiliation(s)
- Jun Tang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Institute of Lipids Medicines, Wenzhou Medical University, Wenzhou, China
| | - Yinkun Yan
- Department of Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenfeng Tong
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Renke Zhou
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Junhui Zhang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Jie Mi
- Department of Non-communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Duo Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
- Institute of Nutrition & Health, Qingdao University, Qingdao, China
| |
Collapse
|
39
|
Trans-palmitoleic acid reduces adiposity via increased lipolysis in a rodent model of diet-induced obesity. Br J Nutr 2021; 127:801-809. [PMID: 33958011 DOI: 10.1017/s0007114521001501] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Obesity is defined as increased adiposity, which leads to metabolic disease. The growth of adipose tissue depends on its capacity to expand through hyperplasia or hypertrophy, in order to buffer energy surplus. Also, during the establishment of obesity, adipose tissue expansion reflects adipose lipid metabolism (lipogenesis and/or lipolysis). It is well known that dietary factors can modify lipid metabolism promoting or preventing the development of metabolic abnormalities that concur with obesity. Trans-palmitoleic acid (TP), a biomarker of dairy consumption, has been associated with reduced adiposity in clinical studies. Thus, we aimed to evaluate the effect of TP over adiposity and lipid metabolism-related genes in a rodent model of diet-induced obesity (DIO). To fulfil this aim, we fed C57BL/6 mice with a Control or a High-Fat diet, added with or without TP (3 g/kg diet), during 11 weeks. Body weight and food intake were monitored, fat pads were weighted, histology of visceral adipose tissue was analysed and lipid metabolism-related gene expression was explored by qPCR. Results show that TP consumption prevented weight gain induced by high-fat diet, reduced visceral adipose tissue weight and adipocyte size, while increasing the expression of lipolytic molecules. In conclusion, we show for the first time that TP influences adipose tissue metabolism, specifically lipolysis, resulting in decreased adiposity and reduced adipocyte size in a DIO mice model.
Collapse
|
40
|
Bourebaba L, Kornicka-Garbowska K, Al Naem M, Röcken M, Łyczko J, Marycz K. MSI-1436 improves EMS adipose derived progenitor stem cells in the course of adipogenic differentiation through modulation of ER stress, apoptosis, and oxidative stress. Stem Cell Res Ther 2021; 12:97. [PMID: 33536069 PMCID: PMC7860037 DOI: 10.1186/s13287-020-02102-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Protein tyrosine phosphatase 1B (PTP1B) is one of the major negative regulators of leptin and insulin signaling, and has been strongly implicated in insulin resistance development in the course of obesity and metabolic syndrome conditions; however, its exact role in controlling adipose tissue biogenesis is still poorly understood. OBJECTIVES This investigation aimed to elucidate whether selective inhibition of PTP1B using MSI-1436 compound may improve and restore the defective adipogenicity of ASCs isolated from EMS-affected horses. METHODS Equine ASC EMS cells were cultured under adipogenic conditions in the presence of PTP1B inhibitor and were subsequently tested for expression of the main adipogenic-related genes using RT-qPCR, changes in free fatty acid profiles by means of GC-MS technique, and for mitochondrial dynamics improvement through the analysis of mitochondrial transmembrane potential and oxidative stress. RESULTS Selective inhibition of PTP1B in equine ASC EMS cells improved substantially adipogenic differentiation by promoting cellular proliferation and normalizing expression of C/EBPalpha, PPARγ, and Adipoq markers that are critical for proper adipogenesis. Levels of secreted adiponectin and PPARγ were also shown to be increased in MSI-1436-conditioned cells, while total leptin levels markedly dropped under the same conditions. Moreover, MSI-1436 treatment enabled the regulation of metabolic-related transcripts that are crosslink to adipogenesis, namely Akt1, Akt2, and SHBG. The obtained results demonstrated also an obvious reduction in intracellular accumulated ROS and NO, as well as mitigated ER stress through the downregulation of Chop, Perk, Atf6, Ire1, and Xbp1 transcripts upon PTP1B inhibition. Furthermore, general fluctuations in FFA composition of all differentiated groups have been highlighted, where palmitic acid, palmitoleic acid, stearic acid, and linolelaidic acid that are known to be associated with the development of metabolic disorders were found to be normalized upon PTP1B inhibition during adipogenic differentiation. CONCLUSION The presented data provides the evidence that the use of PTP1B inhibitor may be successful in controlling and enhancing adipogenic differentiation of impaired equine ASCs affected by metabolic syndrome, and thus offers new insights for the management of obesity through the regulation of adipose tissue dynamics.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Norwida 27B Street, A7 Building, 50-375, Wrocław, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kornicka-Garbowska
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Norwida 27B Street, A7 Building, 50-375, Wrocław, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland
| | - Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany
| | - Jacek Łyczko
- Department of Chemistry, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Norwida 27B Street, A7 Building, 50-375, Wrocław, Poland.
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany.
| |
Collapse
|
41
|
Huang WW, Hong BH, Bai KK, Tan R, Yang T, Sun JP, Yi RZ, Wu H. Cis- and Trans-Palmitoleic Acid Isomers Regulate Cholesterol Metabolism in Different Ways. Front Pharmacol 2020; 11:602115. [PMID: 33363473 PMCID: PMC7753117 DOI: 10.3389/fphar.2020.602115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022] Open
Abstract
Hypercholesterolemia is a preventable risk factor for atherosclerosis and cardiovascular disease. However, the mechanisms whereby cis-palmitoleic acid (cPOA) and trans-palmitoleic acid (tPOA) promote cholesterol homeostasis and ameliorate hypercholesterolemia remain elusive. To investigate the effects of cPOA and tPOA on cholesterol metabolism and its mechanisms, we induced hypercholesterolemia in mice using a high-fat diet and then intragastrically administered cPOA or tPOA once daily for 4 weeks. tPOA administration reduced serum cholesterol, low-density lipoprotein, high-density lipoprotein, and hepatic free cholesterol and total bile acids (TBAs). Conversely, cPOA had no effect on these parameters except for TBAs. Histological examination of the liver, however, revealed that cPOA ameliorated hepatic steatosis more effectively than tPOA. tPOA significantly reduced the expression of 3-hydroxy-3-methyl glutaryl coenzyme reductase (HMGCR), LXRα, and intestinal Niemann-Pick C1-Like 1 (NPC1L1) and increased cholesterol 7-alpha hydroxylase (CYP7A1) in the liver, whereas cPOA reduced the expression of HMGCR and CYP7A1 in the liver and had no effect on intestinal NPC1L1. In summary, our results suggest that cPOA and tPOA reduce cholesterol synthesis by decreasing HMGCR levels. Furthermore, tPOA, but not cPOA, inhibited intestinal cholesterol absorption by downregulating NPC1L1. Both high-dose tPOA and cPOA may promote the conversion of cholesterol into bile acids by upregulating CYP7A1. tPOA and cPOA prevent hypercholesterolemia via distinct mechanisms.
Collapse
Affiliation(s)
- Wen-Wen Huang
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bi-Hong Hong
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Kai-Kai Bai
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Ran Tan
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Ting Yang
- Fujian Provincial Key Laboratory on Hematology, Department of Hematology, Fujian Medical University Union Hospital, Fujian Institute of Hematology, Fuzhou, China
| | - Ji-Peng Sun
- Zhejiang Marine Development Research Institute, Zhoushan, China
| | - Rui-Zao Yi
- Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China
| | - Hao Wu
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
42
|
Chao YS, Wu CJ, Wu HC, Hsu HT, Tsao LC, Cheng YP, Lai YC, Chen WC. Opportunities and Challenges From Leading Trends in a Biomonitoring Project: Canadian Health Measures Survey 2007-2017. Front Public Health 2020; 8:460. [PMID: 33014969 PMCID: PMC7509036 DOI: 10.3389/fpubh.2020.00460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/22/2020] [Indexed: 12/31/2022] Open
Abstract
Background: Biomonitoring can be conducted by assessing the levels of chemicals in human bodies and their surroundings, for example, as was done in the Canadian Health Measures Survey (CHMS). This study aims to report the leading increasing or decreasing biomarker trends and determine their significance. Methods: We implemented a trend analysis for all variables from CHMS biomonitoring data cycles 1–5 conducted between 2007 and 2017. The associations between time and obesity were determined with linear regressions using the CHMS cycles and body mass index (BMI) as predictors. Results: There were 997 unique biomarkers identified and 86 biomarkers with significant trends across cycles. Nine of the 10 leading biomarkers with the largest decreases were environmental chemicals. The levels of 1,2,3-trimethyl benzene, dodecane, palmitoleic acid, and o-xylene decreased by more than 60%. All of the 10 chemicals with the largest increases were environmental chemicals, and the levels of 1,2,4-trimethylbenzene, nonanal, and 4-methyl-2-pentanone increased by more than 200%. None of the 20 biomarkers with the largest increases or decreases between cycles were associated with BMI. Conclusions: The CHMS provides the opportunity for researchers to determine associations between biomarkers and time or BMI. However, the unknown causes of trends with large magnitudes of increase or decrease and their unclear impact on Canadians' health present challenges. We recommend that the CHMS plan future cycles on leading trends and measure chemicals with both human and environmental samples.
Collapse
Affiliation(s)
| | - Chao-Jung Wu
- Département d'informatique, Université du Québec à Montréal, Montreal, QC, Canada
| | - Hsing-Chien Wu
- Taipei Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | | | | | | | - Yi-Chun Lai
- National Yang-Ming University Hospital, Yilan, Taiwan
| | - Wei-Chih Chen
- Attending Physician, Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Emergency and Critical Care Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
43
|
Weir NL, Steffen BT, Guan W, Johnson LM, Djousse L, Mukamal KJ, Tsai MY. Circulating omega-7 fatty acids are differentially related to metabolic dysfunction and incident type II diabetes: The Multi-Ethnic Study of Atherosclerosis (MESA). DIABETES & METABOLISM 2020; 46:319-325. [PMID: 31706030 PMCID: PMC7200281 DOI: 10.1016/j.diabet.2019.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 12/16/2022]
Abstract
AIM Determine whether plasma omega-7 vaccenic acid and palmitoleic acid levels are related to homeostasis model of insulin resistance scores and incident type II diabetes, and whether race/ethnicity modifies these associations. METHODS Plasma phospholipid fatty acids were measured by gas chromatography with flame-ionization detection in Multi-Ethnic Study of Atherosclerosis participants. Linear regression determined associations of vaccenic acid and palmitoleic acid with log-transformed homeostasis model of insulin resistance scores (n=5689), and Cox regression determined associations with incident type II diabetes (n=5413, 660 cases). Race-interactions were tested. RESULTS Adjusting for typical risk factors, higher levels of plasma vaccenic acid were found to be inversely associated with insulin resistance scores across all four race/ethnicities, and a significant race-interaction was observed between Hispanics and Caucasians (P for interaction=0.03). Vaccenic acid was related to 17%, 32%, and 39% lower risks of incident type II diabetes in Black, Hispanic, and Chinese American participants, respectively. Differences in associations between races were detected (P for interactions<0.05). By contrast, higher levels of plasma palmitoleic acid were related to greater insulin resistance scores in Blacks (P<0.001) and Hispanics (P<0.001); significant race-based differences between associations were detected (P for interactions<0.05). Palmitoleic acid was correspondingly related to a 21% greater risk of incident type II diabetes in Black individuals. CONCLUSIONS Results suggest that plasma vaccenic acid and palmitoleic acid are markers of metabolic health and dysfunction, respectively. Coupled with previous evidence and the significant race-interactions, our findings have implications for future studies of the race-based differences in omega-7 fatty acids and their regulation in the context of deteriorating metabolic health.
Collapse
Affiliation(s)
- Natalie L. Weir
- University of Minnesota, Department of Laboratory Medicine & Pathology, Minneapolis, MN 55455
| | - Brian T. Steffen
- University of Minnesota, Department of Laboratory Medicine & Pathology, Minneapolis, MN 55455
| | - Weihua Guan
- University of Minnesota, School of Public Health Biostatistics Division, Minneapolis, MN 55455
| | - Lisa M. Johnson
- University of Utah, Department of Pathology, Salt Lake City, UT 84108
| | - Luc Djousse
- Brigham and Women’s Hospital, Department of Medicine Division of Aging, Boston, MA 02115
| | - Kenneth J. Mukamal
- Beth Israel Deaconess Medical Center, General Medicine, Brookline, MA 02446
| | - Michael Y. Tsai
- University of Minnesota, Department of Laboratory Medicine & Pathology, Minneapolis, MN 55455
| |
Collapse
|
44
|
Shramko VS, Polonskaya YV, Kashtanova EV, Stakhneva EM, Ragino YI. The Short Overview on the Relevance of Fatty Acids for Human Cardiovascular Disorders. Biomolecules 2020; 10:E1127. [PMID: 32751513 PMCID: PMC7464661 DOI: 10.3390/biom10081127] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
This review presents existing evidence of the influence of saturated and unsaturated fatty acids on cardiovascular diseases (CVD). Data are discussed regarding the roles of the most relevant fatty acids, such as myristic (C14:0), palmitic (C16:0), stearic (C18:0), palmitoleic (C16:1), oleic (C18:1), linoleic (C18:2), α-linolenic (C18:3, ω-3), γ-linolenic (C18:3, ω-6), arachidonic (C20:4), eicosapentaenoic (C20:5), docosahexaenoic (C22:6), and docosapentaenoic (C22:5) acid. The accumulated knowledge has expanded the understanding of the involvement of fatty acids in metabolic processes, thereby enabling the transition from basic exploratory studies to practical issues of application of these biomolecules to CVD treatment. In the future, these findings are expected to facilitate the interpretation and prognosis of changes in metabolic lipid aberrations in CVD.
Collapse
Affiliation(s)
| | | | | | - Ekaterina M. Stakhneva
- Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Research Institute of Internal and Preventive Medicine, 630089 Novosibirsk, Russia; (V.S.S.); (Y.V.P.); (E.V.K.); (Y.I.R.)
| | | |
Collapse
|
45
|
Chen G, Li Y, Zeng F, Deng G, Liang J, Wang J, Su Y, Chen Y, Mao L, Liu Z, Bao W, Zhang Z. Biomarkers of fatty acids and risk of type 2 diabetes: a systematic review and meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 2020; 61:2705-2718. [PMID: 32598176 DOI: 10.1080/10408398.2020.1784839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We aimed to quantify and generate comprehensive evidence on the associations of different fatty acids (FAs) with the risk of type 2 diabetes mellitus (T2DM). PubMed, MEDLINE, and Embase were searched for prospective cohort studies that examined the association between FA biomarkers and the risk of T2DM published before 18 May 2020. Random-effects meta-analyses of the effects of FA concentration on T2DM were performed. Thirty-three studies involving 95,810 adults (19,904 cases) were included. Divergent associations of different types of FAs with type 2 diabetes were observed. The pooled relative risk (RRs) of T2DM comparing the top versus the bottom tertile of saturated FAs (C14:0, C16:0, C18:0, total saturated FAs), monounsaturated FA (C16:1 n-7), polyunsaturated FA (C20:3 n-6, C22:4 n-6), and Δ-6-desaturase activities ranged from 1.19 to 1.80. Interestingly, unlike previous studies, we found a negative correlation between odd-chain saturated FAs (C15:0, C17:0), trans-FAs (trans-C16:1 n-7), total n-6, Δ-5-desaturase activities and risk of T2DM. The pooled RRs of T2DM comparing the top versus the bottom tertile of these FAs ranged from 0.62 to 0.78. No associations with T2DM were observed for the other FAs. Considerable heterogeneity was observed in our study, and no definitive conclusions can be made until further investigation has been carried out.
Collapse
Affiliation(s)
- Gengdong Chen
- Department of Obstetrics, Foshan Institute of Fetal Medicine, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Yan Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Fangfang Zeng
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou, China
| | - Guifang Deng
- Department of Clinical Nutrition, Nanshan People's Hospital, Shenzhen, China
| | - Jingjing Liang
- Department of Child Health Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jue Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yixiang Su
- Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuming Chen
- Guangdong Provincial Key Laboratory of Food, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Limei Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhengping Liu
- Department of Obstetrics, Foshan Institute of Fetal Medicine, Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Wei Bao
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, Iowa, USA.,Obesity Research and Education Initiative, University of Iowa, Iowa City, Iowa, USA.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, Iowa, USA
| | - Zheqing Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
46
|
Huang NK, Matthan NR, Galluccio JM, Shi P, Lichtenstein AH, Mozaffarian D. Supplementation with Seabuckthorn Oil Augmented in 16:1n-7t Increases Serum Trans-Palmitoleic Acid in Metabolically Healthy Adults: A Randomized Crossover Dose-Escalation Study. J Nutr 2020; 150:1388-1396. [PMID: 32140719 PMCID: PMC7269729 DOI: 10.1093/jn/nxaa060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/20/2019] [Accepted: 02/24/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In animal models cis-palmitoleic acid (9-hexadecenoic acid; 16:1n-7c), a lipokine, improves insulin sensitivity, inflammation, and lipoprotein profiles; in humans trans-palmitoleic acid (16:1n-7t) has been associated with lower incidence of type 2 diabetes. The response to dose-escalation of supplements containing cis- and trans-palmitoleic acid has not been evaluated. OBJECTIVES We examined dose-escalation effects of oral supplementation with seabuckthorn oil and seabuckthorn oil augmented in 16:1n-7t on serum phospholipid fatty acids (PLFAs). METHODS Thirteen participants (7 women and 6 men; age 48 ± 16 y, BMI 30.4 ± 3.7 kg/m2) participated in a randomized, double-blind, crossover, dose-escalation trial of unmodified seabuckthorn oils relatively high in 16:1n-7c (380, 760, and 1520 mg 16:1n-7c/d) and seabuckthorn oils augmented in 16:1n-7t (120, 240, and 480 mg 16:1n-7t/d). Each of the 3 escalation doses was provided for 3 wk, with a 4-wk washout period between the 2 supplements. At the end of each dose period, fasting blood samples were used to determine the primary outcomes (serum concentrations of the PLFAs 16:1n-7t and 16:1n-7c) and the secondary outcomes (glucose homeostasis, serum lipids, and clinical measures). Trends across doses were evaluated using linear regression. RESULTS Compared with baseline, supplementation with seabuckthorn oil augmented in 16:1n-7t increased phospholipid 16:1n-7t by 26.6% at the highest dose (P = 0.0343). Supplementation with unmodified seabuckthorn oil resulted in a positive trend across the dose-escalations (P-trend = 0.0199). No significant effects of either supplement were identified on blood glucose, insulin, lipids, or other clinical measures, although this dosing study was not powered to detect such effects. No carryover or adverse effects were observed. CONCLUSIONS Supplementation with seabuckthorn oil augmented in 16:1n-7t and unmodified seabuckthorn oil moderately increased concentrations of their corresponding PLFAs in metabolically healthy adults, supporting the use of supplementation with these fatty acids to test potential clinical effects in humans.This trial was registered at clinicaltrials.gov as NCT02311790.
Collapse
Affiliation(s)
- Neil K Huang
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Nirupa R Matthan
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA,Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| | - Jean M Galluccio
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA
| | - Peilin Shi
- Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| | - Alice H Lichtenstein
- Jean Mayer USDA Human Nutrition Research Center on Aging, Boston, MA, USA,Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA,Address correspondence to AHL (e-mail: )
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA, USA
| |
Collapse
|
47
|
Fatty Acid Profile and Desaturase Activities in 7-10-Year-Old Children Attending Primary School in Verona South District: Association between Palmitoleic Acid, SCD-16, Indices of Adiposity, and Blood Pressure. Int J Mol Sci 2020; 21:ijms21113899. [PMID: 32486144 PMCID: PMC7312303 DOI: 10.3390/ijms21113899] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
In previous studies, dietary and circulating fatty acids (FA) and desaturases activity (delta-5 desaturase [D5D], delta-6 desaturase [D6D], and stearoyl-CoA desaturase [SCD-16]) involved in their metabolism were associated with metabolic and cardiovascular disorders. The aim of the study was to assess the association between different FAs and desaturases activity (estimated as product:precursor ratios) with individual cardiovascular risk factors (in particular, anthropometric measurements and blood pressure [BP]) in children. The FA profile was determined on a whole-blood drop in 243 children (age: 8.6 ± 0.72 years) participating in a school-based cross-sectional study. Docosahexaenoic acid (DHA) inversely correlated with indices of adiposity, glucose, and triglycerides. Palmitoleic acid and SCD-16 were directly associated with markers of adiposity and BP, even after adjustment for main confounders. D6D correlated directly with the waist/height ratio. Children with excess weight (>85th percentile; that is overweight plus obese ones) showed higher palmitic acid, palmitoleic acid, and higher SCD-16 activity as compared to normal-weight children. Most of the associations were confirmed in the excess-weight group. Omega-3 FAs, particularly DHA, but not omega-6 FA, showed a potentially beneficial association with metabolic parameters, whereas palmitoleic acid and SCD-16 showed a potentially harmful association with indices of adiposity and BP, especially in obese children.
Collapse
|
48
|
Inhibition of Protein-tyrosine Phosphatase PTP1B and LMPTP Promotes Palmitate/Oleate-challenged HepG2 Cell Survival by Reducing Lipoapoptosis, Improving Mitochondrial Dynamics and Mitigating Oxidative and Endoplasmic Reticulum Stress. J Clin Med 2020; 9:jcm9051294. [PMID: 32369900 PMCID: PMC7288314 DOI: 10.3390/jcm9051294] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives: Non-alcoholic fatty liver disease (NAFLD) is considered a well-known pathology that is determined without using alcohol and has emerged as a growing public health problem. Lipotoxicity is known to promote hepatocyte death, which, in the context of NAFLD, is termed lipoapoptosis. The severity of NAFLD correlates with the degree of hepatocyte lipoapoptosis. Protein–tyrosine phosphatases (PTP) including PTP1B and Low molecular weight PTP (LMPTP), are negative regulators of the insulin signaling pathway and are considered a promising therapeutic target in the treatment of diabetes. In this study, we hypothesized that the inhibition of PTP1B and LMPTP may potentially prevent hepatocyte apoptosis, mitochondrial dysfunction and endoplasmic reticulum (ER) stress onset, following lipotoxicity induced using a free fatty acid (FFA) mixture. Methods: HepG2 cells were cultured in the presence or absence of two PTP inhibitors, namely MSI-1436 and Compound 23, prior to palmitate/oleate overloading. Apoptosis, ER stress, oxidative stress, and mitochondrial dynamics were then evaluated by either MUSE or RT-qPCR analysis. Results: The obtained data demonstrate that the inhibition of PTP1B and LMPTP prevents apoptosis induced by palmitate and oleate in the HepG2 cell line. Moreover, mitochondrial dynamics were positively improved following inhibition of the enzyme, with concomitant oxidative stress reduction and ER stress abrogation. Conclusion: In conclusion, PTP’s inhibitory properties may be a promising therapeutic strategy for the treatment of FFA-induced lipotoxicity in the liver and ultimately in the management of the NAFLD condition.
Collapse
|
49
|
Geng C, Guo Y, Wang C, Liao D, Han W, Zhang J, Jiang P. Systematic impacts of chronic unpredictable mild stress on metabolomics in rats. Sci Rep 2020; 10:700. [PMID: 31959868 PMCID: PMC6971284 DOI: 10.1038/s41598-020-57566-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/02/2020] [Indexed: 01/23/2023] Open
Abstract
Depression is the most common disabling psychiatric disease, with a high prevalence and mortality. Chronic unpredictable mild stress (CUMS) is a well-accepted method used to mimic clinical depression. Recent evidence has consistently suggested that the cumulative effects of CUMS could lead to allostatic overload in the body, thereby inducing systemic disorders; however, there are no previous systematic metabonomics studies on the main stress-targeted tissues associated with depression. A non-targeted gas chromatography–mass spectrometry (GC–MS) approach was used to identify metabolic biomarkers in the main stress-targeted tissues (serum, heart, liver, brain, and kidney) in a CUMS model of depression. Male Sprague–Dawley rats were randomly allocated to the CUMS group (n = 8) or a control group (n = 8). Multivariate analysis was performed to identify the metabolites that were differentially expressed between the two groups. There were 10, 10, 9, 4, and 7 differentially expressed metabolites in the serum, heart, liver, brain and kidney tissues, respectively, between the control and CUMS groups. These were linked to nine different pathways related to the metabolism of amino acids, lipids, and energy. In summary, we provided a comprehensive understanding of metabolic alterations in the main stress-targeted tissues, helping to understand the potential mechanisms underlying depression.
Collapse
Affiliation(s)
- Chunmei Geng
- Jining First People's Hospital, Jining Medical University, Jining, 272000, China
| | - Yujin Guo
- Jining First People's Hospital, Jining Medical University, Jining, 272000, China
| | - Changshui Wang
- Department of Clinical Translational Medicine, Jining Life Science Center, Jining, 272000, China
| | - Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, Central South University, Changsha, 410011, China
| | - Wenxiu Han
- Jining First People's Hospital, Jining Medical University, Jining, 272000, China
| | - Jing Zhang
- Department of Medical Engineering, Jining Medical University, Jining, 272000, China
| | - Pei Jiang
- Jining First People's Hospital, Jining Medical University, Jining, 272000, China.
| |
Collapse
|
50
|
Yuan S, Larsson SC. Association of genetic variants related to plasma fatty acids with type 2 diabetes mellitus and glycaemic traits: a Mendelian randomisation study. Diabetologia 2020; 63:116-123. [PMID: 31690987 PMCID: PMC6890658 DOI: 10.1007/s00125-019-05019-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/28/2019] [Indexed: 01/22/2023]
Abstract
AIMS/HYPOTHESIS Epidemiological data on the associations of circulating fatty acid levels with type 2 diabetes are inconsistent. We conducted a two-sample Mendelian randomisation study to explore the causal associations of plasma levels of ten fatty acids with type 2 diabetes and glycaemic traits. METHODS Thirteen SNPs associated with circulating levels of ten individual fatty acids at the genome-wide significance level (p < 5 × 10-8) were selected as instrumental variables for the exposures. For the outcomes, summary-level data were obtained from the DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) consortium for type 2 diabetes (898,130 individuals) and from the Meta-Analyses of Glucose and Insulin-related traits Consortium (MAGIC) for the glycaemic traits (up to 46,186 non-diabetic individuals). The inverse-variance weighted method was used for analyses. RESULTS Genetic predisposition to higher plasma levels of eight of the ten fatty acids were statistically significantly associated with lower or higher odds of type 2 diabetes. The OR per one SD increment of each fatty acid was 0.93 (95% CI 0.90, 0.96; p = 2.21 × 10-5) for α-linolenic acid, 0.96 (95% CI 0.94, 0.98; p = 1.85 × 10-4) for linoleic acid, 0.86 (95% CI 0.81, 0.91; p = 6.68 × 10-7) for palmitoleic acid, 0.87 (95% CI 0.81, 0.93; p = 2.21 × 10-5) for oleic acid, 1.08 (95% CI 1.03, 1.12; p = 0.002) for eicosapentaenoic acid, 1.04 (95% CI 1.02, 1.07; p = 0.001) for docosapentaenoic acid, 1.03 (95% CI 1.02, 1.05; p = 2.51 × 10-5) for arachidonic acid and 1.09 (95% CI 1.03, 1.15; p = 0.003) for stearic acid. The same eight fatty acids were also associated with fasting glucose levels and HOMA-B. The associations, except that for palmitoleic acid, were driven by variants in FADS1/2. CONCLUSIONS/INTERPRETATION Genetic predisposition to higher circulating levels of eight out of ten fatty acids was associated with type 2 diabetes, fasting glucose and islet beta cell function. However, the associations, except that for palmitoleic acid, were driven by variants in FADS1/2, which encode enzymes with a key role in fatty acid metabolism.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Nobelsväg 13, 17177, Stockholm, Sweden
| | - Susanna C Larsson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Nobelsväg 13, 17177, Stockholm, Sweden.
| |
Collapse
|