1
|
Rodrigues AMDS, Martins LB, Fagundes GBP, Tibaes JRB, Amaral MHA, Vieira ÉLM, Oliveira MC, Correia MITD, Ferreira AVM. Acute inflammatory and metabolic effect of high fructose intake in normal-weight women: A randomized, double-masked, crossover trial. Nutrition 2025; 129:112602. [PMID: 39442382 DOI: 10.1016/j.nut.2024.112602] [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: 06/05/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVES We aimed to evaluate the acute effect of a fructose-rich single meal on metabolic and inflammatory biomarkers RESEARCH METHODS AND PROCEDURES: This single-center, double-masked, randomized crossover trial recruited females aged 20 to 47 with a normal body mass index and was conducted at Hospital das Clínicas (Belo Horizonte, MG, Brazil). Participants received a standardized meal with either sucrose, glucose, or a fructose overload. Blood samples were collected after overnight fasting (baseline) and at 30, 60, 120, and 240 minutes postprandial. Serum levels of glucose, triglycerides (primary outcome), total cholesterol, alanine aminotransferase, aspartate aminotransferase, adiponectin, leptin, resistin, interleukin (IL)-2, IL-4, IL-5, IL-6, IL-10, IL-17, interferon-gamma, tumor necrosis factor, eotaxin, and total blood leukocytes were measured. RESULTS This trial was completed with 25 enrolled participants, and three dropped out. The per-protocol analysis included 22 participants. As expected, postprandial glycemia increased 30 minutes after consuming meals rich in sucrose (P = 0.045) or glucose (P < 0.001). Triglyceride and leucocyte concentrations increased only at 240 minutes after consuming a high-fructose meal (P < 0.05). Regardless of the type of carbohydrate overload, leptin concentrations decreased postprandially compared to baseline at all time points (P < 0.05). Four participants reported adverse events after consuming the standardized meal with glucose or fructose, including nausea and malaise. CONCLUSIONS Our findings indicate that a fructose-rich single meal leads to a more significant increase in triglyceride and leukocyte concentrations compared to glucose and sucrose in healthy women. These findings support concerns regarding the potential inflammatory and metabolic dysfunction associated with frequent consumption of high-fructose meals.
Collapse
Affiliation(s)
- Ana Maria Dos S Rodrigues
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laís B Martins
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela B P Fagundes
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jenneffer R B Tibaes
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Matheus H A Amaral
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Érica Leandro M Vieira
- Department of Internal Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina C Oliveira
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria Isabel T D Correia
- Department of Surgery, Medicine School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adaliene V M Ferreira
- Immunometabolism, Department of Nutrition, Nursing School, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Miller KC, Geyer B, Alexopoulos AS, Moylan CA, Pagidipati N. Disparities in Metabolic Dysfunction-Associated Steatotic Liver Disease Prevalence, Diagnosis, Treatment, and Outcomes: A Narrative Review. Dig Dis Sci 2024:10.1007/s10620-024-08722-0. [PMID: 39560808 DOI: 10.1007/s10620-024-08722-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/26/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is a leading cause of morbidity and mortality, and health disparities have been shown to influence disease burden. AIM In this review, we aim to characterize disparities in prevalence, diagnosis, treatment, and outcomes of MASLD, and to make recommendations for next steps to minimize these disparities. METHODS Literature search on PubMed and Scopus databases was conducted to identify relevant articles published before September 2, 2024. RESULTS Relative to women and White populations, MASLD is more common in men and Hispanic populations and less common in Black populations. It is also more prevalent among those with lower SES. Noninvasive clinical scores may perform differently across groups, and screening practices vary both for initial disease and for progression to metabolic dysfunctionassociated steatohepatitis (MASH), formerly called non-alcoholic steatohepatitis (NASH). Women and Black and Hispanic patients suffer worse outcomes including rates of progression to MASH and mortality. CONCLUSIONS Health disparities related to race, ethnicity, gender, and socioeconomic factors impact multiple stages of care for patients with MASLD.
Collapse
|
3
|
Srnic N, Dearlove D, Johnson E, MacLeod C, Krupa A, McGonnell A, Frazer-Morris C, O'Rourke P, Parry S, Hodson L. Greater oxidation of dietary linoleate compared to palmitate in humans following an acute high-carbohydrate diet. Clin Nutr 2024; 43:2305-2315. [PMID: 39226718 DOI: 10.1016/j.clnu.2024.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND We have previously demonstrated that dietary saturated fatty acids (SFA), when compared to polyunsaturated fatty acids (PUFA), are preferentially partitioned into oxidation pathways. However, it remains unclear if this preferential handling is maintained when hepatocellular metabolism is shifted toward fatty acid (FA) esterification and away from oxidation, such as when hepatic de novo lipogenesis (DNL) is upregulated. AIM To investigate whether an acute upregulation of hepatic DNL influences dietary FA partitioning into oxidation pathways. METHODS 20 healthy volunteers (11 females) underwent a fasting baseline visit followed by two study days, 2-weeks apart. Prior to each study day, participants consumed an isocaloric high-carbohydrate diet (to upregulate hepatic DNL) for 3-days. On the two study days, participants consumed an identical standardised test meal that contained either [U13C]palmitate or [U13C]linoleate, in random order, to trace the fate of dietary FA. Blood and breath samples were collected over a 6h postprandial period and 13C enrichment in breath CO2 and plasma lipid fractions were measured using gas-chromatography-combustion-isotope ratio mass spectrometry. RESULTS Compared to the baseline visit, fasting plasma triglyceride concentrations and markers of hepatic DNL, the lipogenic and stearyl-CoA desaturase indices, were significantly (p < 0.05) increased after consumption of the high-carbohydrate diet. Appearance of 13C in expired CO2 and tracer recovery were significantly (p < 0.05) higher after consumption of the meal containing [U13C]linoleate compared to [U13C]palmitate (5.1 ± 0.5% vs. 3.7 ± 0.4%), respectively. Incorporation of 13C into the plasma triglyceride and non-esterified fatty acid pool was significantly (p < 0.001) greater for [U13C]palmitate compared to [U13C]linoleate. CONCLUSION Dietary PUFA compared to SFA appear to be preferentially partitioned into oxidation pathways during an acute upregulation of hepatic DNL, thus consumption of a PUFA-enriched diet may help mitigate intrahepatic triglyceride accumulation in individuals at risk of cardiometabolic disease.
Collapse
Affiliation(s)
- Nikola Srnic
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - David Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Elspeth Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Cameron MacLeod
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Antoni Krupa
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Alice McGonnell
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Charlotte Frazer-Morris
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Paige O'Rourke
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Sion Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Aston Medical School, Aston University, Birmingham B4 7ET, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
4
|
Westcott FA, Nagarajan SR, Parry SA, Savic D, Green CJ, Marjot T, Johnson E, Cornfield T, Mózes FE, O’Rourke P, Mendall J, Dearlove D, Fielding B, Smith K, Tomlinson JW, Hodson L. Dissociation between liver fat content and fasting metabolic markers of selective hepatic insulin resistance in humans. Eur J Endocrinol 2024; 191:463-472. [PMID: 39353069 PMCID: PMC11497584 DOI: 10.1093/ejendo/lvae123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE Fasting hyperglycemia and hypertriglyceridemia are characteristic of insulin resistance (IR) and rodent work has suggested this may be due to selective hepatic IR, defined by increased hepatic gluconeogenesis and de novo lipogenesis (DNL), but this has not been shown in humans. DESIGN Cross-sectional study in men and women across a range of adiposity. METHODS Medication-free participants (n = 177) were classified as normoinsulinemic (NI) or hyperinsulinemic (HI) and as having low (LF) or high (HF) liver fat content measured by magnetic resonance spectroscopy. Fractional gluconeogenesis (frGNG) and hepatic DNL were measured using stable isotope tracer methodology following an overnight fast. RESULTS Although HI and HF groups had higher fasting plasma glucose and triglyceride concentrations when compared to NI and LF groups respectively, there was no difference in frGNG. However, HF participants tended to have lower frGNG than LF participants. HI participants had higher DNL compared to NI participants but there was no difference observed between liver fat groups. CONCLUSIONS Taken together, we found no metabolic signature of selective hepatic IR in fasting humans. DNL may contribute to hypertriglyceridemia in individuals with HI but not those with HF. Glycogenolysis and systemic glucose clearance may have a larger contribution to fasting hyperglycemia than gluconeogenesis, especially in those with HF, and these pathways should be considered for therapeutic targeting.
Collapse
Affiliation(s)
- Felix A Westcott
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Shilpa R Nagarajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Sion A Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- Aston Medical School, Aston University, Birmingham, B4 7ET, United Kingdom
| | - Dragana Savic
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Elspeth Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Thomas Cornfield
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Ferenc E Mózes
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Paige O’Rourke
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Jessica Mendall
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - David Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Barbara Fielding
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, United Kingdom
| | - Kieran Smith
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- OCDEM, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, OX3 7LE, United Kingdom
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LE, United Kingdom
- OCDEM, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, OX3 7LE, United Kingdom
| |
Collapse
|
5
|
Gladding M, Shen X, Snyder MP, Havel PJ, Adams SH. Interindividual Variability in Postprandial Plasma Fructose Patterns in Adults. Nutrients 2024; 16:3079. [PMID: 39339679 PMCID: PMC11435096 DOI: 10.3390/nu16183079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
High fructose consumption is associated with an increased risk of cardiometabolic disease, and fructose feeding dose-dependently induces markers reflective of poor metabolic health. However, unlike glucose, surprisingly little is known about person-to-person differences in postprandial plasma fructose patterns. Herein, we performed post hoc analyses of two published studies to address this question. In the first cohort, 16 participants' all-day plasma fructose concentration patterns (08:00-23:30) were determined (8 women and 8 men) while consuming mixed meals (breakfast, lunch, and dinner) with a fructose-sweetened beverage at each meal (30% of calories). Individually plotted results demonstrate remarkably disparate fructose patterns with respect to peak concentration and timing. A secondary study confirmed substantial interindividual variability in plasma fructose patterns over 240 min in 16 adults consuming Ensure®, a commercially available mixed macronutrient drink containing a low dose of fructose. The health ramifications of interindividual variations in postprandial fructose metabolism and the underlying physiological mechanisms driving differences in post-meal blood patterns remain to be explored. Future research is warranted to determine if interindividual variability in fructose digestion, metabolism, and postprandial blood concentration patterns is associated with cardiometabolic health phenotypes and disease risk.
Collapse
Affiliation(s)
- Mia Gladding
- Department of Food Science and Nutrition, California Polytechnic University, San Luis Obispo, CA 93407, USA
| | - Xiaotao Shen
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94306, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, CA 94306, USA
| | - Michael P Snyder
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94306, USA
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, CA 94306, USA
| | - Peter J Havel
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Sean H Adams
- Department of Surgery, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Center for Alimentary and Metabolic Science, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
6
|
Weng X, Xu J, Yang S. Association between the arm circumference and non-alcoholic fatty liver disease in American children and adolescence: a population-based analysis. Front Public Health 2024; 12:1323795. [PMID: 38859898 PMCID: PMC11163100 DOI: 10.3389/fpubh.2024.1323795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 05/14/2024] [Indexed: 06/12/2024] Open
Abstract
Background The arm circumference (AC) has been used as an important tool to access the risk of non-alcoholic fatty liver disease (NAFLD) in adults. However, the association between AC and NAFLD in children and adolescence remains unclear. This study aims to explore the relationship between AC and NAFLD in American children and adolescence. Methods 2017-2020 National Health and Nutrition Examination Survey (NHANES) was used to carry out the cross-sectional study. The association between AC and the risk of NAFLD, and liver steatosis was analyzed using weighted multivariable logistic regression and multivariate linear regression. Additionally, a two-part linear regression model was used to identify threshold effects in this study. Subgroup analysis, interaction tests and receiver operating characteristic (ROC) curve analysis were also carried out. Results A total of 1,559 children and adolescence aged 12-18 years old were included, and the prevalence of NAFLD was 27.3%. AC was positively correlated with the risk of NAFLD (OR = 1.25, 95% CI: 1.19, 1.32) and liver steatosis (β = 4.41, 95% CI: 3.72, 5.09). Subgroup analysis stratified by age and race showed a consistent positive correlation. A non-linear relationship and saturation effect between AC and NAFLD risk were identified, with an S shaped curve and an inflection point at 34.5 cm. Area under the ROC of AC to NAFLD was 0.812, with the sensitivity of 67.6%, the specificity of 83.8% and the cutoff value of 31.7 cm. Conclusion Our study shows that AC is independently correlated with an increased risk of NAFLD and the severity of liver steatosis in American children and adolescence.
Collapse
Affiliation(s)
- Xiaolu Weng
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Xu
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shouxing Yang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Pontifex MG, Vauzour D, Muller M. Sexual dimorphism in the context of nutrition and health. Proc Nutr Soc 2024; 83:109-119. [PMID: 37665115 DOI: 10.1017/s0029665123003610] [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: 09/05/2023]
Abstract
Diets and dietary constituents that we consume have a considerable impact on disease risk. Intriguingly these effects may be modulated to some extent by sex. Lack of female representation in nutritional studies as well as a lack of stratification by sex has and continues to limit our understanding of these sex × diet interactions. Here we provide an overview of the current and available literature describing how exposure to certain dietary patterns (Western-style diet, Mediterranean diet, vegetarian/vegan, ketogenic diet) and dietary constituents (dietary fibre, PUFA and plant bioactive) influences disease risk in a sex-specific manner. Interestingly, these sex differences appear to be highly disease-specific. The identification of such sex differences in response to diet stresses the importance of sex stratification in nutritional research.
Collapse
Affiliation(s)
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Michael Muller
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| |
Collapse
|
8
|
Ibrahim KG, Chivandi E, Erlwanger KH, Brooksbank RL. Neonatal administration of fenofibrate had no developmental programming effect on the lipid profile and relative leucocyte telomere lengths of adolescent rats fed a high-fructose diet postnatally. Can J Physiol Pharmacol 2023; 101:565-573. [PMID: 37433224 DOI: 10.1139/cjpp-2022-0528] [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: 07/13/2023]
Abstract
Telomere length, a marker of ageing, is susceptible to developmental programming that may cause its accelerated attrition. Metabolic syndrome triggers telomere attrition. Fenofibrate, a peroxisome proliferator-activated receptor-alpha agonist, is protective against telomere attrition. We investigated the impact of fenofibrate administered during suckling on the lipid profile and leucocyte telomere lengths of rats fed a high-fructose diet post-weaning. Suckling Sprague-Dawley pups (n = 119) were allocated to four groups and gavaged with either 10 mL·kg-1 body mass 0.5% dimethyl sulfoxide, 100 mg·kg-1 body mass fenofibrate, fructose (20%, w / v), or a combination of fenofibrate and fructose for 15 days. Upon weaning, each of the initial groups was split into two subgroups: one had plain water while the other had fructose solution (20%, w / v) to drink for 6 weeks. Blood was collected for DNA extraction and relative leucocyte telomere length determination by real-time PCR. Plasma triglycerides and cholesterol were also quantified. The treatments had no effect (p > 0.05) on body mass, cholesterol concentration, and relative leucocyte telomere lengths in both sexes. Post-weaning fructose increased triglyceride concentrations (p < 0.05) in female rats. Fenofibrate administered during suckling did not affect ageing nor did it prevent high fructose-induced hypertriglyceridaemia in female rats.
Collapse
Affiliation(s)
- Kasimu Ghandi Ibrahim
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, P.O. Box 2000, Zarqa 13110, Jordan
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, Johannesburg, South Africa
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, P.M.B. 2254, Sokoto, Nigeria
| | - Eliton Chivandi
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, Johannesburg, South Africa
| | - Kennedy Honey Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, Johannesburg, South Africa
| | - Richard Leslie Brooksbank
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, Johannesburg, South Africa
| |
Collapse
|
9
|
Fridén M, Mora AM, Lind L, Risérus U, Kullberg J, Rosqvist F. Diet composition, nutrient substitutions and circulating fatty acids in relation to ectopic and visceral fat depots. Clin Nutr 2023; 42:1922-1931. [PMID: 37633021 DOI: 10.1016/j.clnu.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/29/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND & AIMS Short-term randomized trials have demonstrated that replacing saturated fat (SFA) with polyunsaturated fat (PUFA) causes a reduction or prevention of liver fat accumulation, but population-based studies on diet and body fat distribution are limited. We investigated cross-sectional associations between diet, circulating fatty acids and liver fat, visceral adipose tissue (VAT), intermuscular adipose tissue (IMAT) and other fat depots using different energy-adjustment models. METHODS Sex-stratified analyses of n = 9119 (for serum fatty acids) to 13 849 (for nutrients) participants in UK Biobank were conducted. Fat depots were assessed by MRI, circulating fatty acids by NMR spectroscopy and diet by repeated 24-h recalls. Liver fat, VAT and IMAT were primary outcomes; total adipose tissue (TAT) and abdominal subcutaneous adipose tissue (ASAT) were secondary outcomes. Three a priori defined models were constructed: the all-components model, standard model and leave-one-out model (main model including specified nutrient substitutions). Imiomics (MRI-derived) was used to confirm and visualize associations. RESULTS In women, substituting carbohydrates and free sugars with saturated fat (SFA) was positively associated with liver fat (β (95% CI) = 0.19 (0.02, 0.36) and β (95% CI) = 0.20 (0.05-0.35), respectively) and IMAT (β (95% CI) = 0.07 (0.00, 0.14) and β (95% CI) = 0.08 (0.02, 0.13), respectively), whereas substituting animal fat with plant fat was inversely associated with IMAT, ASAT and TAT. In the all-components and standard models, SFA and animal fat were positively associated with liver fat, IMAT and VAT whereas plant fat was inversely associated with IMAT in women. Few associations were observed in men. Circulating polyunsaturated fatty acids (PUFA) were inversely associated with liver fat, IMAT and VAT in both men and women, whereas SFA and monounsaturated fatty acids were positively associated. CONCLUSIONS Type of dietary fat may be an important determinant of ectopic fat in humans consuming their habitual diet. Plant fat and PUFA should be preferred over animal fat and SFA. This is corroborated by circulating fatty acids and overall consistent through different energy adjustment models. TWITTER SUMMARY In UK Biobank, intake of saturated- and animal fat were positively whereas biomarkers of polyunsaturated fat were inversely associated with liver-, visceral- and intermuscular fat. Type of dietary fat may be a determinant of ectopic fat, a risk factor for cardiometabolic disease.
Collapse
Affiliation(s)
- Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Andrés Martínez Mora
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden.
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden.
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Joel Kullberg
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden; Antaros Medical AB, Mölndal, Sweden.
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Nagarajan SR, Livingstone EJ, Monfeuga T, Lewis LC, Ali SHL, Chandran A, Dearlove DJ, Neville MJ, Chen L, Maroteau C, Ruby MA, Hodson L. MLX plays a key role in lipid and glucose metabolism in humans: Evidence from in vitro and in vivo studies. Metabolism 2023; 144:155563. [PMID: 37088121 PMCID: PMC10687193 DOI: 10.1016/j.metabol.2023.155563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND AND AIM Enhanced hepatic de novo lipogenesis (DNL) has been proposed as an underlying mechanism for the development of NAFLD and insulin resistance. Max-like protein factor X (MLX) acts as a heterodimer binding partner for glucose sensing transcription factors and inhibition of MLX or downstream targets has been shown to alleviate intrahepatic triglyceride (IHTG) accumulation in mice. However, its effect on insulin sensitivity remains unclear. As human data is lacking, the aim of the present work was to investigate the role of MLX in regulating lipid and glucose metabolism in primary human hepatocytes (PHH) and in healthy participants with and without MLX polymorphisms. METHODS PHH were transfected with non-targeting or MLX siRNA to assess the effect of MLX knockdown on lipid and glucose metabolism, insulin signalling and the hepatocellular transcriptome. A targeted association analysis on imputed genotype data for MLX on healthy individuals was undertaken to assess associations between specific MLX SNPs (rs665268, rs632758 and rs1474040), plasma biochemistry, IHTG content, DNL and gluconeogenesis. RESULTS MLX knockdown in PHH altered lipid metabolism (decreased DNL (p < 0.05), increased fatty acid oxidation and ketogenesis (p < 0.05), and reduced lipid accumulation (p < 0.001)). Additionally, MLX knockdown increased glycolysis, lactate secretion and glucose production (p < 0.001) and insulin-stimulated pAKT levels (p < 0.01) as assessed by transcriptomic, steady-state and dynamic measurements. Consistent with the in vitro data, individuals with the rs1474040-A and rs632758-C variants had lower fasting plasma insulin (p < 0.05 and p < 0.01, respectively) and TG (p < 0.05 and p < 0.01, respectively). Although there was no difference in IHTG or gluconeogenesis, individuals with rs632758 SNP had notably lower hepatic DNL (p < 0.01). CONCLUSION We have demonstrated using human in vitro and in vivo models that MLX inhibition favored lipid catabolism over anabolism and increased glucose production, despite increased glycolysis and phosphorylation of Akt, suggesting a metabolic mechanism that involves futile cycling.
Collapse
Affiliation(s)
- Shilpa R Nagarajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | | | - Thomas Monfeuga
- Novo Nordisk Research Centre Oxford, Innovation Building, Oxford, UK
| | - Lara C Lewis
- Novo Nordisk Research Centre Oxford, Innovation Building, Oxford, UK
| | | | | | - David J Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, UK
| | - Lingyan Chen
- Novo Nordisk Research Centre Oxford, Innovation Building, Oxford, UK
| | - Cyrielle Maroteau
- Novo Nordisk Research Centre Oxford, Innovation Building, Oxford, UK
| | - Maxwell A Ruby
- Novo Nordisk Research Centre Oxford, Innovation Building, Oxford, UK.
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, UK.
| |
Collapse
|
11
|
Brouwers MCGJ. Fructose 1-phosphate, an evolutionary signaling molecule of abundancy. Trends Endocrinol Metab 2022; 33:680-689. [PMID: 35995682 DOI: 10.1016/j.tem.2022.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022]
Abstract
Evidence is accumulating that specifically fructose exerts adverse cardiometabolic effects in humans. Recent experimental studies have shown that fructose not only serves as a substrate for, among others, intrahepatic lipid formation, but also has a signaling function. It is postulated that fructose 1-phosphate (F1-P) has evolved as a signaling molecule of abundancy that stimulates nutrient absorption, lipid storage, and reproduction. Such a role would provide an explanation for why fructose contributes to the pathogenesis of evolutionary mismatch diseases, including nonalcoholic fatty liver disease (NAFLD), cardiovascular disease, polycystic ovary syndrome (PCOS), and colorectal cancer, in the current era of nutritional abundance. It is anticipated that reducing F1-P, by either pharmacological inhibition of ketohexokinase (KHK) or societal measures, will mitigate the risk of these diseases.
Collapse
Affiliation(s)
- Martijn C G J Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Centre, Maastricht, The Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
12
|
High-fructose feeding suppresses cold-stimulated brown adipose tissue glucose uptake independently of changes in thermogenesis and the gut microbiome. Cell Rep Med 2022; 3:100742. [PMID: 36130480 PMCID: PMC9512695 DOI: 10.1016/j.xcrm.2022.100742] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/14/2022] [Accepted: 08/23/2022] [Indexed: 12/30/2022]
Abstract
Diets rich in added sugars are associated with metabolic diseases, and studies have shown a link between these pathologies and changes in the microbiome. Given the reported associations in animal models between the microbiome and brown adipose tissue (BAT) function, and the alterations in the microbiome induced by high-glucose or high-fructose diets, we investigated the potential causal link between high-glucose or -fructose diets and BAT dysfunction in humans. Primary outcomes are changes in BAT cold-induced thermogenesis and the fecal microbiome (clinicaltrials.gov, NCT03188835). We show that BAT glucose uptake, but not thermogenesis, is impaired by a high-fructose but not high-glucose diet, in the absence of changes in the gastrointestinal microbiome. We conclude that decreased BAT glucose metabolism occurs earlier than other pathophysiological abnormalities during fructose overconsumption in humans. This is a potential confounding factor for studies relying on 18F-FDG to assess BAT thermogenesis. Fructose overfeeding decreases brown adipose tissue glucose metabolism These changes occur independently of oxidative metabolism No change is observed with glucose overfeeding The gut microbiome is not affected by fructose/glucose overfeeding
Collapse
|
13
|
ERα-Dependent Regulation of Adropin Predicts Sex Differences in Liver Homeostasis during High-Fat Diet. Nutrients 2022; 14:nu14163262. [PMID: 36014766 PMCID: PMC9416503 DOI: 10.3390/nu14163262] [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: 07/28/2022] [Revised: 08/07/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a public health issue, due to its prevalence and association with other cardiometabolic diseases. Growing evidence suggests that NAFLD alters the production of hepatokines, which, in turn, influence several metabolic processes. Despite accumulating evidence on the major role of estrogen signaling in the sexually dimorphic nature of NAFLD, dependency of hepatokine expression on sex and estrogens has been poorly investigated. Through in vitro and in vivo analysis, we determined the extent to which hepatokines, known to be altered in NAFLD, can be regulated, in a sex-specific fashion, under different hormonal and nutritional conditions. Our study identified four hepatokines that better recapitulate sex and estrogen dependency. Among them, adropin resulted as one that displays a sex-specific and estrogen receptor alpha (ERα)-dependent regulation in the liver of mice under an excess of dietary lipids (high-fat diet, HFD). Under HFD conditions, the hepatic induction of adropin negatively correlates with the expression of lipogenic genes and with fatty liver in female mice, an effect that depends upon hepatic ERα. Our findings support the idea that ERα-mediated induction of adropin might represent a potential approach to limit or prevent NAFLD.
Collapse
|
14
|
Yan RR, Chan CB, Louie JCY. Current WHO recommendation to reduce free sugar intake from all sources to below 10% of daily energy intake for supporting overall health is not well supported by available evidence. Am J Clin Nutr 2022; 116:15-39. [PMID: 35380611 PMCID: PMC9307988 DOI: 10.1093/ajcn/nqac084] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 04/01/2022] [Indexed: 01/15/2023] Open
Abstract
Sugar is widely consumed over the world. Although the mainstream view is that high added or free sugar consumption leads to obesity and related metabolic diseases, controversies exist. This narrative review aims to highlight important findings and identify major limitations and gaps in the current body of evidence in relation to the effect of high sugar intakes on health. Previous animal studies have shown that high sucrose or fructose consumption causes insulin resistance in the liver and skeletal muscle and consequent hyperglycemia, mainly because of fructose-induced de novo hepatic lipogenesis. However, evidence from human observational studies and clinical trials has been inconsistent, where most if not all studies linking high sugar intake to obesity focused on sugar-sweetened beverages (SSBs), and studies focusing on sugars from solid foods yielded null findings. In our opinion, the substantial limitations in the current body of evidence, such as short study durations, use of supraphysiological doses of sugar or fructose alone in animal studies, and a lack of direct comparisons of the effects of solid compared with liquid sugars on health outcomes, as well as the lack of appropriate controls, seriously curtail the translatability of the findings to real-world situations. It is quite possible that "high" sugar consumption at normal dietary doses (e.g., 25% daily energy intake) per se-that is, the unique effect of sugar, especially in the solid form-may indeed not pose a health risk for individuals apart from the potential to reduce the overall dietary nutrient density, although newer evidence suggests "low" sugar intake (<5% daily energy intake) is just as likely to be associated with nutrient dilution. We argue the current public health recommendations to encourage the reduction of both solid and liquid forms of free sugar intake (e.g., sugar reformulation programs) should be revised due to the overextrapolation of results from SSBs studies.
Collapse
Affiliation(s)
- Rina Ruolin Yan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | |
Collapse
|
15
|
Fridén M, Kullberg J, Ahlström H, Lind L, Rosqvist F. Intake of Ultra-Processed Food and Ectopic-, Visceral- and Other Fat Depots: A Cross-Sectional Study. Front Nutr 2022; 9:774718. [PMID: 35445063 PMCID: PMC9013765 DOI: 10.3389/fnut.2022.774718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction The purpose of this study was to investigate associations between intake of ultra-processed food (UPF) and liver fat, pancreas fat and visceral adipose tissue (VAT) but also subcutaneous adipose tissue (SAT), VAT/SAT ratio and total fat mass. Materials and Methods Cross-sectional analysis of n = 286 50-year old men and women. Energy percentage (%E) from UPF was calculated from a semi-quantitative food frequency questionnaire. Food items were categorized according to the NOVA-classification system and fat depots were assessed using magnetic resonance imaging (MRI) and bioelectrical impedance analysis (BIA). Associations were analyzed using linear regression, adjusted for sex, education, physical activity, smoking, dietary factors and BMI. Results Mean intake of UPF was 37.8 ± 10.2 %E and the three largest contributors to this were crisp- and wholegrain breads and spreads, indicating overall healthy food choices. Consumption of UPF was associated with higher intake of energy, carbohydrates and fiber and lower intake of protein and polyunsaturated fat but no differences were observed for total fat, saturated fat (SFA), monounsaturated fat, sugar or alcohol between tertiles of UPF. Intake of UPF was positively associated with liver- and pancreas fat, VAT, VAT/SAT and inversely associated with total fat mass in crude models. The association for VAT remained after full adjustment (β = 0.01 (95% CI: 0.002, 0.02), P = 0.02) and was driven by women. Conclusion Energy intake from UPF is not associated with ectopic fat, SAT or total fat after adjustment for multiple confounders in this population having overall healthy food habits. However, a positive association between UPF and VAT was observed which was driven by women.
Collapse
Affiliation(s)
- Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Joel Kullberg
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden
- Antaros Medical AB, BioVenture Hub, Mölndal, Sweden
| | - Håkan Ahlström
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden
- Antaros Medical AB, BioVenture Hub, Mölndal, Sweden
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
- *Correspondence: Fredrik Rosqvist
| |
Collapse
|
16
|
Zhou X, Lin X, Chen J, Pu J, Wu W, Wu Z, Lin H, Huang K, Zhang L, Dai Y, Ni Y, Dong G, Fu J. Clinical spectrum transition and prediction model of nonalcoholic fatty liver disease in children with obesity. Front Endocrinol (Lausanne) 2022; 13:986841. [PMID: 36120457 PMCID: PMC9471666 DOI: 10.3389/fendo.2022.986841] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE This study aims to outline the clinical characteristics of pediatric NAFLD, as well as establish and validate a prediction model for the disease. MATERIALS AND METHODS The retrospective study enrolled 3216 children with obesity from January 2003 to May 2021. They were divided into obese without NAFLD, nonalcoholic fatty liver (NAFL), and nonalcoholic steatohepatitis (NASH) groups. Clinical data were retrieved, and gender and chronologic characteristics were compared between groups. Data from the training set (3036) were assessed using univariate analyses and stepwise multivariate logistic regression, by which a nomogram was developed to estimate the probability of NAFLD. Another 180 cases received additional liver hydrogen proton magnetic resonance spectroscopy (1H-MRS) as a validation set. RESULTS The prevalence of NAFLD was higher in males than in females and has increased over the last 19 years. In total, 1915 cases were NAFLD, and the peak onset age was 10-12 years old. Hyperuricemia ranked first in childhood NAFLD comorbidities, followed by dyslipidemia, hypertension, metabolic syndrome (MetS), and dysglycemia. The AUROC of the eight-parameter nomogram, including waist-to-height ratio (WHtR), hip circumference (HC), triglyceride glucose-waist circumference (TyG-WC), alanine aminotransferase (ALT), high-density lipoprotein cholesterol (HDL-C), apolipoprotein A1(ApoA1), insulin sensitivity index [ISI (composite)], and gender, for predicting NAFLD was 0.913 (sensitivity 80.70%, specificity 90.10%). Calibration curves demonstrated a great calibration ability of the model. CONCLUSION AND RELEVANCE NAFLD is the most common complication in children with obesity. The nomogram based on anthropometric and laboratory indicators performed well in predicting NAFLD. This can be used as a quick screening tool to assess pediatric NAFLD in children with obesity.
Collapse
|
17
|
Chi Y, Youn DY, Xiaoli AM, Liu L, Qiu Y, Kurland IJ, Pessin JB, Yang F, Pessin JE. Comparative impact of dietary carbohydrates on the liver transcriptome in two strains of mice. Physiol Genomics 2021; 53:456-472. [PMID: 34643091 PMCID: PMC8616594 DOI: 10.1152/physiolgenomics.00053.2021] [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: 05/17/2021] [Revised: 09/02/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
Excessive long-term consumption of dietary carbohydrates, including glucose, sucrose, or fructose, has been shown to have significant impact on genome-wide gene expression, which likely results from changes in metabolic substrate flux. However, there has been no comprehensive study on the acute effects of individual sugars on the genome-wide gene expression that may reveal the genetic changes altering signaling pathways, subsequent metabolic processes, and ultimately physiological/pathological responses. Considering that gene expressions in response to acute carbohydrate ingestion might be different in nutrient sensitive and insensitive mammals, we conducted comparative studies of genome-wide gene expression by deep mRNA sequencing of the liver in nutrient sensitive C57BL/6J and nutrient insensitive BALB/cJ mice. Furthermore, to determine the temporal responses, we compared livers from mice in the fasted state and following ingestion of standard laboratory mouse chow supplemented with plain drinking water or water containing 20% glucose, sucrose, or fructose. Supplementation with these carbohydrates induced unique extents and temporal changes in gene expressions in a strain specific manner. Fructose and sucrose stimulated gene changes peaked at 3 h postprandial, whereas glucose effects peaked at 12 h and 6 h postprandial in C57BL/6J and BABL/cJ mice, respectively. Network analyses revealed that fructose changed genes were primarily involved in lipid metabolism and were more complex in C57BL/6J than in BALB/cJ mice. These data demonstrate that there are qualitative and antitative differences in the normal physiological responses of the liver between these two strains of mice and C57BL/6J is more sensitive to sugar intake than BALB/cJ.
Collapse
Affiliation(s)
- Yuling Chi
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- The Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
| | - Dou Yeon Youn
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- The Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
| | - Alus M Xiaoli
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- The Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Li Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- The Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
| | - Yunping Qiu
- Einstein Stable Isotope and Metabolomics Core, Albert Einstein College of Medicine, Bronx, New York
| | - Irwin J Kurland
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Einstein Stable Isotope and Metabolomics Core, Albert Einstein College of Medicine, Bronx, New York
| | - Jacob B Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Fajun Yang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- The Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- The Fleischer Institute of Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
18
|
Della Torre S. Beyond the X Factor: Relevance of Sex Hormones in NAFLD Pathophysiology. Cells 2021; 10:2502. [PMID: 34572151 PMCID: PMC8470830 DOI: 10.3390/cells10092502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, being frequently associated with obesity, unbalanced dietary regimens, and reduced physical activity. Despite their greater adiposity and reduced physical activity, women show a lower risk of developing NAFLD in comparison to men, likely a consequence of a sex-specific regulation of liver metabolism. In the liver, sex differences in the uptake, synthesis, oxidation, deposition, and mobilization of lipids, as well as in the regulation of inflammation, are associated with differences in NAFLD prevalence and progression between men and women. Given the major role of sex hormones in driving hepatic sexual dimorphism, this review will focus on the role of sex hormones and their signaling in the regulation of hepatic metabolism and in the molecular mechanisms triggering NAFLD development and progression.
Collapse
Affiliation(s)
- Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
19
|
Hashemi L, Zhang Q, Getahun D, Jasuja GK, McCracken C, Pisegna J, Roblin D, Silverberg MJ, Tangpricha V, Vupputuri S, Goodman M. Longitudinal Changes in Liver Enzyme Levels Among Transgender People Receiving Gender Affirming Hormone Therapy. J Sex Med 2021; 18:1662-1675. [PMID: 37057433 PMCID: PMC8444147 DOI: 10.1016/j.jsxm.2021.06.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/10/2021] [Accepted: 06/18/2021] [Indexed: 01/12/2023]
Abstract
Background: The effect of gender affirming hormone therapy (GAHT) on clinical laboratory parameters, including levels of liver enzymes alanine aminotransferase (ALT) and aspartate transaminase (AST), is an area of uncertainty in transgender health. Aim: We sought to estimate the distribution parameters of liver enzyme levels among transmasculine (TM) and transfeminine (TF) persons receiving GAHT relative to the corresponding measures in cisgender reference groups, and to evaluate longitudinal changes in these laboratory measures following GAHT initiation. Methods: The data for this longitudinal study included 624 TF and 438 transmasculine (TM) people as well as 4,090 cisgender males and 4,797 cisgender females enrolled in 3 integrated health systems. Time under observation was divided into 2 intervals: from the first blood test to the date of the first filled GAHT prescription and from GAHT initiation to the most recent ALT or AST measurement. Linear mixed models were used to compare changes in log-transformed ALT and AST values among transgender cohort members before and after GAHT initiation, and relative to the reference groups. The results were expressed as relative differences (in %) and the ratios of these differences (ratios-of-ratios) along with the 95% confidence intervals (CIs). Outcomes: Changes in ALT and AST levels among transgender people over time and relative to the corresponding changes in cisgender referents. Results: Among TM study participants, the post GAHT ratios-of-ratios for AST were 1.61 (95% CI: 1.13, 2.31) and 1.57 (95% CI: 1.06, 2.31) relative to cisgender males and females respectively. For ALT, the corresponding comparisons yielded the ratios-of-ratios (95% CIs) of 2.06 (1.67, 2.54) and 1.90 (1.50, 2.40). No statistically significant changes were observed among TF participants. Other factors associated with higher liver enzyme levels included alcohol use/abuse and obesity. Clinical Implications: TM persons may experience modest increases in ALT and AST concentrations following testosterone initiation; however, clinical significance of the observed association remains unclear and requires further investigation. By contrast, feminizing GAHT is unlikely to induce appreciable changes in liver enzyme levels. Strength and Limitations: The strengths of this study are the longitudinal design and the ability to assemble an unselected cohort nested within large health systems. The main limitations include the lack of information on hormone levels and the inability to take into account GAHT doses and routes of administration. Conclusion: The influence of long-term GAHT on ALT and AST levels appears modest and not likely to reflect clinically meaningful changes in liver function.
Collapse
Affiliation(s)
- Leila Hashemi
- VA Greater Los Angeles Healthcare System, Department of General Internal Medicine, Los Angeles, CA, USA; David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Qi Zhang
- Rollins School of Public Health, Emory University, Atlanta GA, USA
| | - Darios Getahun
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Guneet K Jasuja
- Center for Healthcare Organization and Implementation Research (CHOIR), Edith Nourse Memorial Veterans Hospital, Bedford VA Medical Center, Bedford, MA, USA; Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, MA, USA
| | - Courtney McCracken
- Center for Research and Evaluation, Kaiser Permanente Georgia, Atlanta, GA, USA
| | - Joseph Pisegna
- VA Greater Los Angeles Healthcare System, Department of General Internal Medicine, Los Angeles, CA, USA; David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Douglas Roblin
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente Mid-Atlanta States, Rockville, MD, USA
| | | | - Vin Tangpricha
- Department of Endocrinology, Emory University, School of Medicine, Atlanta, GA, USA; The Atlanta VA Medical Center, Atlanta, GA, USA
| | - Suma Vupputuri
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente Mid-Atlanta States, Rockville, MD, USA
| | - Michael Goodman
- Rollins School of Public Health, Emory University, Atlanta GA, USA
| |
Collapse
|
20
|
Burra P, Bizzaro D, Gonta A, Shalaby S, Gambato M, Morelli MC, Trapani S, Floreani A, Marra F, Brunetto MR, Taliani G, Villa E. Clinical impact of sexual dimorphism in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Liver Int 2021; 41:1713-1733. [PMID: 33982400 DOI: 10.1111/liv.14943] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022]
Abstract
NAFLD/NASH is a sex-dimorphic disease, with a general higher prevalence in men. Women are at reduced risk of NAFLD compared to men in fertile age, whereas after menopause women have a comparable prevalence of NAFLD as men. Indeed, sexual category, sex hormones and gender habits interact with numerous NAFLD factors including cytokines, stress and environmental factors and alter the risk profiles and phenotypes of NAFLD. In the present review, we summarized the last findings about the influence of sex on epidemiology, pathogenesis, progression in cirrhosis, indication for liver transplantation and alternative therapies, including lifestyle modification and pharmacological strategies. We are confident that an appropriate consideration of sex, age, hormonal status and sociocultural gender differences will lead to a better understanding of sex differences in NAFLD risk, therapeutic targets and treatment responses and will aid in achieving sex-specific personalized therapies.
Collapse
Affiliation(s)
- Patrizia Burra
- Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padua, Padua, Italy
| | - Debora Bizzaro
- Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padua, Padua, Italy
| | - Anna Gonta
- Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padua, Padua, Italy
| | - Sarah Shalaby
- Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padua, Padua, Italy
| | - Martina Gambato
- Multivisceral Transplant Unit, Department of Surgery, Oncology and Gastroenterology, University Hospital of Padua, Padua, Italy
| | | | - Silvia Trapani
- Italian National Transplant Center, Italian National Institute of Health, Rome, Italy
| | - Annarosa Floreani
- University of Padova, Padua, Italy.,IRCCS Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - Fabio Marra
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maurizia Rossana Brunetto
- Hepatology and Liver Physiopathology Laboratory and Internal Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gloria Taliani
- Infectious Diseases Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Erica Villa
- Gastroenterology Unit, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | | |
Collapse
|
21
|
Dietary fructose intake is correlated with fat distribution in the Newfoundland population. Nutrition 2021; 93:111434. [PMID: 34492622 DOI: 10.1016/j.nut.2021.111434] [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: 03/08/2021] [Revised: 07/08/2021] [Accepted: 07/18/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Increased dietary fructose intake is associated with elevated body weight and body mass index. Few studies are available regarding the relationship between fat distribution and dietary fructose intake. The aim of this study was to investigate the association between dietary fructose intake and fat distribution in adults in a large Newfoundland cohort. METHODS We analyzed 2298 adults from CODING (Complex Diseases in the New found land Population: Environment and Genetics) study. Intake of dietary fructose was evaluated from the Willett food frequency questionnaire. Fat distribution was estimated by dual-energy x-ray absorptiometry. Partial correlation analysis was used to determine the correlations of dietary fructose intake with fat distribution adjusted for major confounding factors. RESULTS Daily dietary fructose intake was negatively associated with arm fat in postmenopausal women (r = -0.080, P < 0.05), but positively associated with arm fat in premenopausal women after adjusting for age, total calorie intake, and physical activity levels (r = 0.079, P < 0.05). Dietary fructose intake was negatively correlated with both arm fat (r = -0.131, P < 0.05) and visceral fat (r = -0.124 measured in mass, r = -0.124 measured in volume respectively; P < 0.05) in men <45 y of age, not in men ≥45 y. CONCLUSION This study demonstrated that dietary fructose intake is significantly correlated with arm fat in both women and men, and visceral fat in men in the Newfoundland free-living population. The correlations are sex- and menopause-status dependent.
Collapse
|
22
|
Annandale M, Daniels LJ, Li X, Neale JPH, Chau AHL, Ambalawanar HA, James SL, Koutsifeli P, Delbridge LMD, Mellor KM. Fructose Metabolism and Cardiac Metabolic Stress. Front Pharmacol 2021; 12:695486. [PMID: 34267663 PMCID: PMC8277231 DOI: 10.3389/fphar.2021.695486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease is one of the leading causes of mortality in diabetes. High fructose consumption has been linked with the development of diabetes and cardiovascular disease. Serum and cardiac tissue fructose levels are elevated in diabetic patients, and cardiac production of fructose via the intracellular polyol pathway is upregulated. The question of whether direct myocardial fructose exposure and upregulated fructose metabolism have potential to induce cardiac fructose toxicity in metabolic stress settings arises. Unlike tightly-regulated glucose metabolism, fructose bypasses the rate-limiting glycolytic enzyme, phosphofructokinase, and proceeds through glycolysis in an unregulated manner. In vivo rodent studies have shown that high dietary fructose induces cardiac metabolic stress and functional disturbance. In vitro, studies have demonstrated that cardiomyocytes cultured in high fructose exhibit lipid accumulation, inflammation, hypertrophy and low viability. Intracellular fructose mediates post-translational modification of proteins, and this activity provides an important mechanistic pathway for fructose-related cardiomyocyte signaling and functional effect. Additionally, fructose has been shown to provide a fuel source for the stressed myocardium. Elucidating the mechanisms of fructose toxicity in the heart may have important implications for understanding cardiac pathology in metabolic stress settings.
Collapse
Affiliation(s)
- M Annandale
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - L J Daniels
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - X Li
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - J P H Neale
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - A H L Chau
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - H A Ambalawanar
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - S L James
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - P Koutsifeli
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - L M D Delbridge
- Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - K M Mellor
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Department of Physiology, School of Biomedical Sciences, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
23
|
Bhat SF, Pinney SE, Kennedy KM, McCourt CR, Mundy MA, Surette MG, Sloboda DM, Simmons RA. Exposure to high fructose corn syrup during adolescence in the mouse alters hepatic metabolism and the microbiome in a sex-specific manner. J Physiol 2021; 599:1487-1511. [PMID: 33450094 DOI: 10.1113/jp280034] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
KEY POINTS The prevalence of obesity and non-alcoholic fatty liver disease in children is dramatically increasing at the same time as consumption of foods with a high sugar content. Intake of high fructose corn syrup (HFCS) is a possible aetiology as it is thought to be more lipogenic than glucose. In a mouse model, HFCS intake during adolescence increased fat mass and hepatic lipid levels in male and female mice. However, only males showed impaired glucose tolerance. Multiple metabolites including lipids, bile acids, carbohydrates and amino acids were altered in liver in a sex-specific manner at 6 weeks of age. Some of these changes were also present in adulthood even though HFCS exposure ended at 6 weeks. HFCS significantly altered the gut microbiome, which was associated with changes in key microbial metabolites. These results suggest that HFCS intake during adolescence has profound metabolic changes that are linked to changes in the microbiome and these changes are sex-specific. ABSTRACT The rapid increase in obesity, diabetes and fatty liver disease in children over the past 20 years has been linked to increased consumption of high fructose corn syrup (HFCS), making it essential to determine the short- and long-term effects of HFCS during this vulnerable developmental window. We hypothesized that HFCS exposure during adolescence significantly impairs hepatic metabolic signalling pathways and alters gut microbial composition, contributing to changes in energy metabolism with sex-specific effects. C57bl/6J mice with free access to HFCS during adolescence (3-6 weeks of age) underwent glucose tolerance and body composition testing and hepatic metabolomics, gene expression and triglyceride content analysis at 6 and 30 weeks of age (n = 6-8 per sex). At 6 weeks HFCS-exposed mice had significant increases in fat mass, glucose intolerance, hepatic triglycerides (females) and de novo lipogenesis gene expression (ACC, DGAT, FAS, ChREBP, SCD, SREBP, CPT and PPARα) with sex-specific effects. At 30 weeks, HFCS-exposed mice also had abnormalities in glucose tolerance (males) and fat mass (females). HFCS exposure enriched carbohydrate, amino acid, long chain fatty acid and secondary bile acid metabolism at 6 weeks with changes in secondary bile metabolism at 6 and 30 weeks. Microbiome studies performed immediately before and after HFCS exposure identified profound shifts of microbial species in male mice only. In summary, short-term HFCS exposure during adolescence induces fatty liver, alters important metabolic pathways, some of which continue to be altered in adulthood, and changes the microbiome in a sex-specific manner.
Collapse
Affiliation(s)
- Shazia F Bhat
- Department of Pediatrics, Christiana Care Health System, Newark, DE, USA
| | - Sara E Pinney
- Division of Endocrinology and Diabetes, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katherine M Kennedy
- Department of Biochemistry & Biomedical Sciences, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Cole R McCourt
- School of Arts and Sciences, University of Pennsylvania, PA, USA
| | | | - Michael G Surette
- Department of Biochemistry & Biomedical Sciences, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Deborah M Sloboda
- Department of Biochemistry & Biomedical Sciences, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Rebecca A Simmons
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, PA, USA
| |
Collapse
|
24
|
Roumans KH, Basset Sagarminaga J, Peters HP, Schrauwen P, Schrauwen-Hinderling VB. Liver fat storage pathways: methodologies and dietary effects. Curr Opin Lipidol 2021; 32:9-15. [PMID: 33234776 PMCID: PMC7810416 DOI: 10.1097/mol.0000000000000720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Nonalcoholic fatty liver is the result of an imbalance between lipid storage [from meal, de novo lipogenesis (DNL) and fatty acid (FA) uptake] and disposal (oxidation and VLDL output). Knowledge on the contribution of each of these pathways to liver fat content in humans is essential to develop tailored strategies to prevent and treat nonalcoholic fatty liver. Here, we review the techniques available to study the different storage pathways and review dietary modulation of these pathways. RECENT FINDINGS The type of carbohydrate and fat could be of importance in modulating DNL, as complex carbohydrates and omega-3 FAs have been shown to reduce DNL. No effects were found on the other pathways, however studies investigating this are scarce. SUMMARY Techniques used to assess storage pathways are predominantly stable isotope techniques, which require specific expertise and are costly. Validated biomarkers are often lacking. These methodological limitations also translate into a limited number of studies investigating to what extent storage pathways can be modulated by diet. Further research is needed to elucidate in more detail the impact that fat and carbohydrate type can have on liver fat storage pathways and content.
Collapse
Affiliation(s)
- Kay H.M. Roumans
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht
| | | | | | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht
| | - Vera B. Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
25
|
Novelle MG, Bravo SB, Deshons M, Iglesias C, García-Vence M, Annells R, da Silva Lima N, Nogueiras R, Fernández-Rojo MA, Diéguez C, Romero-Picó A. Impact of liver-specific GLUT8 silencing on fructose-induced inflammation and omega oxidation. iScience 2021; 24:102071. [PMID: 33554072 PMCID: PMC7856473 DOI: 10.1016/j.isci.2021.102071] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/14/2020] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
Excessive consumption of high-fructose diets is associated with insulin resistance, obesity, and non-alcoholic fatty liver disease (NAFLD). However, fructose differentially affects hepatic regulation of lipogenesis in males and females. Hence, additional studies are necessary in order to find strategies taking gender disparities in fructose-induced liver damage into consideration. Although the eighth member of facilitated glucose transporters (GLUT8) has been linked to fructose-induced macrosteatosis in female mice, its contribution to the inflammatory state of NAFLD remains to be elucidated. Combining pharmacological, biochemical, and proteomic approaches, we evaluated the preventive effect of targeted liver GLUT8 silencing on liver injury in a mice female fructose-induced non-alcoholic steatohepatitis female mouse model. Liver GLUT8-knockdown attenuated fructose-induced ER stress, recovered liver inflammation, and dramatically reduced fatty acid content, in part, via the omega oxidation. Therefore, this study links GLUT8 with liver inflammatory response and suggests GLUT8 as a potential target for the prevention of NAFLD.
Collapse
Affiliation(s)
- Marta G Novelle
- Functional Obeosomics and Molecular Metabolism laboratories, Centro singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidad de Santiago de Compostela, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Av. Barcelona s/n 15782, A Coruña, Santiago de Compostela, Spain.,Hepatic Regenerative Medicine Laboratory, Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, Madrid, E28049, Spain
| | - Susana Belén Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Maxime Deshons
- Laboratoire de Toxicologie, Faculté de Pharmacie, Université Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Cristina Iglesias
- Functional Obeosomics and Molecular Metabolism laboratories, Centro singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidad de Santiago de Compostela, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Av. Barcelona s/n 15782, A Coruña, Santiago de Compostela, Spain
| | - María García-Vence
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Rebecca Annells
- Department of Physiology, Anatomy and Genetics, University of Oxford, OX1 3PT, Oxford, UK
| | - Natália da Silva Lima
- Functional Obeosomics and Molecular Metabolism laboratories, Centro singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidad de Santiago de Compostela, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Av. Barcelona s/n 15782, A Coruña, Santiago de Compostela, Spain
| | - Rubén Nogueiras
- Functional Obeosomics and Molecular Metabolism laboratories, Centro singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidad de Santiago de Compostela, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Av. Barcelona s/n 15782, A Coruña, Santiago de Compostela, Spain
| | - Manuel Alejandro Fernández-Rojo
- Hepatic Regenerative Medicine Laboratory, Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, Madrid, E28049, Spain.,School of Medicine, The University of Queensland, Herston, 4006, Brisbane, Australia
| | - Carlos Diéguez
- Functional Obeosomics and Molecular Metabolism laboratories, Centro singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidad de Santiago de Compostela, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Av. Barcelona s/n 15782, A Coruña, Santiago de Compostela, Spain
| | - Amparo Romero-Picó
- Functional Obeosomics and Molecular Metabolism laboratories, Centro singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Universidad de Santiago de Compostela, CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Av. Barcelona s/n 15782, A Coruña, Santiago de Compostela, Spain
| |
Collapse
|
26
|
Della Torre S. Non-alcoholic Fatty Liver Disease as a Canonical Example of Metabolic Inflammatory-Based Liver Disease Showing a Sex-Specific Prevalence: Relevance of Estrogen Signaling. Front Endocrinol (Lausanne) 2020; 11:572490. [PMID: 33071979 PMCID: PMC7531579 DOI: 10.3389/fendo.2020.572490] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
There is extensive evidence supporting the interplay between metabolism and immune response, that have evolved in close relationship, sharing regulatory molecules and signaling systems, to support biological functions. Nowadays, the disruption of this interaction in the context of obesity and overnutrition underlies the increasing incidence of many inflammatory-based metabolic diseases, even in a sex-specific fashion. During evolution, the interplay between metabolism and reproduction has reached a degree of complexity particularly high in female mammals, likely to ensure reproduction only under favorable conditions. Several factors may account for differences in the incidence and progression of inflammatory-based metabolic diseases between females and males, thus contributing to age-related disease development and difference in life expectancy between the two sexes. Among these factors, estrogens, acting mainly through Estrogen Receptors (ERs), have been reported to regulate several metabolic pathways and inflammatory processes particularly in the liver, the metabolic organ showing the highest degree of sexual dimorphism. This review aims to investigate on the interaction between metabolism and inflammation in the liver, focusing on the relevance of estrogen signaling in counteracting the development and progression of non-alcoholic fatty liver disease (NAFLD), a canonical example of metabolic inflammatory-based liver disease showing a sex-specific prevalence. Understanding the role of estrogens/ERs in the regulation of hepatic metabolism and inflammation may provide the basis for the development of sex-specific therapeutic strategies for the management of such an inflammatory-based metabolic disease and its cardio-metabolic consequences.
Collapse
Affiliation(s)
- Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
27
|
Ministrini S, Montecucco F, Sahebkar A, Carbone F. Macrophages in the pathophysiology of NAFLD: The role of sex differences. Eur J Clin Invest 2020; 50:e13236. [PMID: 32294235 DOI: 10.1111/eci.13236] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 03/14/2020] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a multifactorial pathological condition, which recognizes a certain sexual dimorphism. Experimental and clinical studies provided evidence for a critical role of macrophages in NAFLD development and progression. Especially, liver-resident macrophages (also known as Kupffer cells) are likely the common final pathway of several pro-steatosic signals. A huge amount of danger-associated molecular patterns recognized by Kupffer cells is provided within the liver by lipid and glucose toxicity. Other pro-inflammatory signals come from surrounding tissues into the portal vein, directly to the liver: they come from dysfunctional adipocytes, adipose tissue macrophages and gut dysbiosis. These complex crosstalks are differently represented across sexes, as sexual hormones control many of these processes. Sexual dimorphism then modulates metabolic and inflammatory cascades driving the liver from a simple steatosis to NAFLD and beyond. Here, metabolic and inflammatory mechanisms underlying NALFD pathophysiology will be updated. A special attention will be paid to describe sex-related differences that could provide insights for patient stratification and more tailored therapeutic approaches.
Collapse
Affiliation(s)
- Stefano Ministrini
- Internal Medicine Department, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Federico Carbone
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy.,First Clinic of Internal Medicine Department of Internal Medicine, University of Genoa, Genoa, Italy
| |
Collapse
|
28
|
Fappi A, Mittendorfer B. Different physiological mechanisms underlie an adverse cardiovascular disease risk profile in men and women. Proc Nutr Soc 2020; 79:210-218. [PMID: 31340878 PMCID: PMC7583670 DOI: 10.1017/s0029665119001022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CVD affect about one-third of the population and are the leading cause of mortality. The prevalence of CVD is closely linked to the prevalence of obesity because obesity is commonly associated with metabolic abnormalities that are important risk factors for CVD, including insulin resistance, pre-diabetes, and type-2 diabetes, atherosclerotic dyslipidaemia, endothelial dysfunction and hypertension. Women have a more beneficial traditional CVD risk profile (lower fasting plasma glucose, less atherogenic lipid profile) and a lower absolute risk for CVD than men. However, the relative risk for CVD associated with hyperglycaemia and dyslipidaemia is several-fold higher in women than in men. The reasons for the sex differences in CVD risk associated with metabolic abnormalities are unclear but could be related to differences in the mechanisms that cause hyperglycaemia and dyslipidaemia in men and women, which could influence the pathogenic processes involved in CVD. In the present paper, we review the influence of a person's sex on key aspects of metabolism involved in the cardiometabolic disease process, including insulin action on endogenous glucose production, tissue glucose disposal, and adipose tissue lipolysis, insulin secretion and insulin plasma clearance, postprandial glucose, fatty acid, and triglyceride kinetics, hepatic lipid metabolism and myocardial substrate use. We conclude that there are marked differences in many aspects of metabolism in men and women that are not all attributable to differences in the sex hormone milieu. The mechanisms responsible for these differences and the clinical implications of these observations are unclear and require further investigation.
Collapse
Affiliation(s)
- Alan Fappi
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Wallace M, Metallo CM. Tracing insights into de novo lipogenesis in liver and adipose tissues. Semin Cell Dev Biol 2020; 108:65-71. [PMID: 32201132 DOI: 10.1016/j.semcdb.2020.02.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Lipids play important roles in biology that include structural compartmentation as membranes, energy storage, and regulatory functions as signaling molecules. These molecules can be obtained via the surrounding environment (e.g. diet) or synthesized de novo. Fatty acid synthesis is an energetically demanding process and must therefore be tightly regulated to balance fatty acid availability with the functional and energetic needs of cells and tissues. Here we review key aspects of de novo lipogenesis (DNL) in mammalian systems. We highlight key nodes in the pathway that are used for quantitation of lipogenic fluxes and regulation of fatty acid diversity across tissues. Next, we discuss key aspects of DNL function in the major lipogenic tissues of mammals: liver, white adipose tissue (WAT), and brown adipose tissue (BAT), highlighting recent molecular discoveries that suggest potential roles for tissue specific DNL. Finally, we propose critical questions that will be important to address using the advanced approaches for DNL quantitation described herein.
Collapse
Affiliation(s)
- Martina Wallace
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Christian M Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
30
|
Rodgers M, Heineman B, Dushay J. Increased fructose consumption has sex-specific effects on fibroblast growth factor 21 levels in humans. Obes Sci Pract 2019; 5:503-510. [PMID: 31687174 PMCID: PMC6819978 DOI: 10.1002/osp4.360] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/26/2019] [Accepted: 06/30/2019] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE Fibroblast growth factor 21 (FGF21), a primarily hepatic hormone with pleotropic metabolic effects, is regulated by fructose in humans. Recent work has established that 75 g of oral fructose robustly stimulates FGF21 levels in humans with peak levels occurring 2 h following ingestion; this has been termed an oral fructose tolerance test (OFTT). It is unknown whether prolonged high-fructose consumption influences the FGF21 response to acute fructose or whether biological sex influences FGF21-fructose dynamics. METHODS Thirty-nine healthy adults underwent baseline OFTT following an overnight fast. For the high-fructose exposure protocol, 20 subjects ingested 75 g of fructose daily for 14 ± 3 d, followed by repeat OFTT. For the control group, an OFTT was repeated following 14 ± 3 d of ad lib diet. For all subjects, FGF21 levels, glucose, insulin, non-esterified fatty acids and triglyceride levels were measured at baseline and 2 h following OFTT. All subjects maintained 3-d food logs prior to OFTT testing. RESULTS Women demonstrated significantly higher baseline and peak stimulated total and intact FGF21 levels compared with men both before and after high-fructose exposure. Baseline total and intact FGF21 levels decreased following ongoing fructose exposure, maintaining a stable ratio. This decrease was sex specific, with only women demonstrating decreased baseline FGF21 levels. There were no changes in metabolic or anthropometric parameters following the high-fructose exposure. CONCLUSIONS Daily ingestion of 75 g of fructose for 2 weeks results in a sex-specific decrease in baseline FGF21 levels without change in body weight or biochemical evidence of metabolic injury. There were also sex-specific differences in peak fructose-stimulated FGF21 levels, which do not change with high-fructose consumption. The role of FGF21 in the development of metabolic disease caused by fructose consumption may differ based on biological sex. Future long-term studies should consider sex differences in FGF21-fructose dynamics.
Collapse
Affiliation(s)
- M. Rodgers
- Division of Endocrinology and MetabolismBeth Israel Deaconess Medical CenterBostonMAUSA
| | - B. Heineman
- Division of Endocrinology and MetabolismBeth Israel Deaconess Medical CenterBostonMAUSA
| | - J. Dushay
- Division of Endocrinology and MetabolismBeth Israel Deaconess Medical CenterBostonMAUSA
| |
Collapse
|
31
|
Hodson L, Karpe F. Hyperinsulinaemia: does it tip the balance toward intrahepatic fat accumulation? Endocr Connect 2019; 8:R157-R168. [PMID: 31581129 PMCID: PMC6826170 DOI: 10.1530/ec-19-0350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022]
Abstract
In health, the liver is metabolically flexible over the course of the day, as it undertakes a multitude of physiological processes including the regulation of intrahepatic and systemic glucose and lipid levels. The liver is the first organ to receive insulin and through a cascade of complex metabolic processes, insulin not only plays a key role in the intrahepatic regulation of glucose and lipid metabolism, but also in the regulation of systemic glucose and lipid concentrations. Thus, when intrahepatic insulin signalling becomes aberrant then this may lead to perturbations in intrahepatic metabolic processes that have the potential to impact on metabolic health. For example, obesity is associated with intrahepatic fat accumulation (known as nonalcoholic liver disease (NAFLD)) and hyperinsulinaemia, the latter as a result of insulin hypersecretion or impaired hepatic insulin extraction. Although insulin signalling directly alters intra- and extrahepatic metabolism, the regulation of hepatic glucose and fatty acid metabolism is also indirectly driven by substrate availability. Here we discuss the direct and indirect effects of insulin on intrahepatic processes such as the synthesis of fatty acids and peripherally regulating the flux of fatty acids to the liver; processes that may play a role in the development of insulin resistance and/or intrahepatocellular triacylglycerol (IHTAG) accumulation in humans.
Collapse
Affiliation(s)
- Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford University Hospital Trusts, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Oxford University Hospital Trusts, Oxford, UK
| |
Collapse
|
32
|
Payolla TB, Teixeira CJ, Sato FT, Murata GM, Zonta GA, Sodré FS, Campos CV, Mesquita FN, Anhê GF, Bordin S. In Utero Dexamethasone Exposure Exacerbates Hepatic Steatosis in Rats That Consume Fructose During Adulthood. Nutrients 2019; 11:nu11092114. [PMID: 31491968 PMCID: PMC6770256 DOI: 10.3390/nu11092114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/10/2019] [Accepted: 08/21/2019] [Indexed: 12/12/2022] Open
Abstract
Distinct environmental insults might interact with fructose consumption and contribute to the development of metabolic disorders. To address whether in utero glucocorticoid exposure and fructose intake modulate metabolic responses, adult female Wistar rats were exposed to dexamethasone (DEX) during pregnancy, and the offspring were administered fructose at a later time. Briefly, dams received DEX during the third period of pregnancy, while control dams remained untreated. Offspring born to control and DEX-treated mothers were defined as CTL-off and DEX-off, respectively, while untreated animals were designated CTL-off-CTL and DEX-off-CTL. CLT-off and DEX-off treated with 10% fructose in the drinking water for 8 weeks are referred to as CTL-off-FRU and DEX-off-FRU. We found that fructose promoted glucose intolerance and whole-body gluconeogenesis in both CTL-off-FRU and DEX-off-FRU animals. On the other hand, hepatic lipid accumulation was significantly stimulated in DEX-off-FRU rats when compared to the CTL-off-FRU group. The DEX-off-FRU group also displayed impaired very-low-density lipoprotein (VLDL) production and reduced hepatic expression of apoB, mttp, and sec22b. DEX-off-FRU has lower hepatic levels of autophagy markers. Taken together, our results support the unprecedented notion that in utero glucocorticoid exposure exacerbates hepatic steatosis caused by fructose consumption later in life.
Collapse
Affiliation(s)
- Tanyara B Payolla
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Caio J Teixeira
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-887, Brazil
| | - Fabio T Sato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Gilson M Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Gizela A Zonta
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Frhancielly S Sodré
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Carolina V Campos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Filiphe N Mesquita
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-887, Brazil
| | - Gabriel F Anhê
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas, Sao Paulo 13083-887, Brazil
| | - Silvana Bordin
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
33
|
Gonzalez JT. Symposium overview: fructose in physiology. J Physiol 2019; 597:3523-3525. [DOI: 10.1113/jp278267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
34
|
Hernández-Díazcouder A, Romero-Nava R, Carbó R, Sánchez-Lozada LG, Sánchez-Muñoz F. High Fructose Intake and Adipogenesis. Int J Mol Sci 2019; 20:E2787. [PMID: 31181590 PMCID: PMC6600229 DOI: 10.3390/ijms20112787] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
In modern societies, high fructose intake from sugar-sweetened beverages has contributed to obesity development. In the diet, sucrose and high fructose corn syrup are the main sources of fructose and can be metabolized in the intestine and transported into the systemic circulation. The liver can metabolize around 70% of fructose intake, while the remaining is metabolized by other tissues. Several tissues including adipose tissue express the main fructose transporter GLUT5. In vivo, chronic fructose intake promotes white adipose tissue accumulation through activating adipogenesis. In vitro experiments have also demonstrated that fructose alone induces adipogenesis by several mechanisms, including (1) triglycerides and very-low-density lipoprotein (VLDL) production by fructose metabolism, (2) the stimulation of glucocorticoid activation by increasing 11β-HSD1 activity, and (3) the promotion of reactive oxygen species (ROS) production through uric acid, NOX and XOR expression, mTORC1 signaling and Ang II induction. Moreover, it has been observed that fructose induces adipogenesis through increased ACE2 expression, which promotes high Ang-(1-7) levels, and through the inhibition of the thermogenic program by regulating Sirt1 and UCP1. Finally, microRNAs may also be involved in regulating adipogenesis in high fructose intake conditions. In this paper, we propose further directions for research in fructose participation in adipogenesis.
Collapse
Affiliation(s)
- Adrián Hernández-Díazcouder
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
- Departamento de Ciencias de la Salud, Área de Investigación Médica, Universidad Autónoma Metropolitana Iztapalapa, Mexico city 09340, Mexico.
| | - Rodrigo Romero-Nava
- Departamento de Ciencias de la Salud, Área de Investigación Médica, Universidad Autónoma Metropolitana Iztapalapa, Mexico city 09340, Mexico.
- Laboratorio de investigación en Farmacología, Hospital Infantil de México Federico Gómez, Mexico city 06720, Mexico.
- Sección de Postgraduados, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico.
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
| | - L Gabriela Sánchez-Lozada
- Laboratorio de Fisiopatología Renal, Departamento de Nefrología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
| | - Fausto Sánchez-Muñoz
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
- Sección de Postgraduados, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico.
| |
Collapse
|
35
|
Pinnick KE, Hodson L. Challenging metabolic tissues with fructose: tissue-specific and sex-specific responses. J Physiol 2019; 597:3527-3537. [PMID: 30883738 DOI: 10.1113/jp277115] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/25/2019] [Indexed: 12/16/2022] Open
Abstract
Excessive consumption of free sugars (which typically includes a composite of glucose and fructose) is associated with an increased risk of developing chronic metabolic diseases including obesity, non-alcoholic fatty liver disease (NAFLD), type 2 diabetes and cardiovascular disease. Determining the utilisation, storage and fate of dietary sugars in metabolically relevant tissues is fundamental to understanding their contribution to metabolic disease risk. To date, the study of fructose metabolism has primarily focused on the liver, where it has been implicated in impaired insulin sensitivity, increased fat accumulation and dyslipidaemia. Yet we still have only a limited understanding of the mechanisms by which consumption of fructose, as part of a mixed meal, may alter hepatic fatty acid synthesis and partitioning. Moreover, surprisingly little is known about the metabolism of fructose within other organs, specifically subcutaneous adipose tissue, which is the largest metabolically active organ in the human body and is consistently exposed to nutrient fluxes. This review summarises what is known about fructose metabolism in the liver and adipose tissue and examines evidence for tissue-specific and sex-specific responses to fructose.
Collapse
Affiliation(s)
- Katherine E Pinnick
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Affiliation(s)
- Amedeo Lonardo
- Azienda Ospedalero-Universitaria di Modena, Operating Unit of Internal Medicine, Ospedale Civile di Baggiovara, Modena, Italy
| | - Ayako Suzuki
- Division of Gastroenterology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
37
|
Lin WT, Lee CY, Tsai S, Huang HL, Wu PW, Chin YT, Seal DW, Chen T, Chao YY, Lee CH. Clustering of Metabolic Risk Components and Associated Lifestyle Factors: A Nationwide Adolescent Study in Taiwan. Nutrients 2019; 11:nu11030584. [PMID: 30857325 PMCID: PMC6471895 DOI: 10.3390/nu11030584] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
Clustering of metabolic syndrome (MetS) risk components in childhood has been linked to a higher risk of diabetes and cardiovascular diseases in adulthood. By using data from the 2010⁻2011 Nutrition and Health Survey in Taiwan, this study investigated epidemic patterns and correlates for the clustering of MetS risk components. A total of 1920 adolescents aged 12⁻18 years were included in this study. The MetS diagnostic criteria defined by the Taiwan Pediatric Association (TPA) and International Diabetes Federation (IDF) for adolescents and the criteria defined by the Joint Interim Statement for adults (JIS-Adult) were used to evaluate MetS and its abnormal components. The prevalence of TPA-, IDF-, and JIS-Adult-defined MetS was 4.1%, 3.0%, and 4.0%, with 22.1%, 19.3%, and 17.7%⁻18.1% of adolescents having high fasting glucose, low high-density lipoprotein cholesterol, and central obesity, respectively. A 0.4-to-0.5-fold decreased risk of having ≥2 MetS abnormal components was detected among adolescents who consumed ≥1 serving/week of dairy products and fresh fruits. Boys who consumed ≥7 drinks/week of soda and girls who consumed ≥7 drinks/week of tea had a 4.6- and 5.2-fold risk of MetS, respectively. In conclusion, our findings revealed significant dimensions of adolescent MetS, including detecting population-specific prevalent patterns for MetS risk components and their clustering, and emphasized on health promotion activities that reduce sugar-sweetened beverage intake.
Collapse
Affiliation(s)
- Wei-Ting Lin
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Global Community Health and Behavioral Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Chun-Ying Lee
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Sharon Tsai
- Department of Laboratory Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung 812, Taiwan.
| | - Hsiao-Ling Huang
- Department of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Pei-Wen Wu
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yu-Ting Chin
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - David W Seal
- Department of Global Community Health and Behavioral Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Ted Chen
- Department of Global Community Health and Behavioral Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA.
| | - Yu-Ying Chao
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Hung Lee
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|