1
|
Pafili K, Zaharia OP, Strassburger K, Knebel B, Herder C, Huttasch M, Karusheva Y, Kabisch S, Strom A, Nowotny B, Szendroedi J, Roden M. PNPLA3 gene variation modulates diet-induced improvement in liver lipid content in type 2 diabetes. Clin Nutr 2025; 48:6-15. [PMID: 40090039 DOI: 10.1016/j.clnu.2025.02.032] [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: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND&AIMS Lifestyle-induced weight reduction remains crucial for managing type 2 diabetes and steatotic liver disease, but its effectiveness varies. We postulated that the G allele in the rs738409 single nucleotide polymorphism within patatin-like phospholipase domain-containing protein 3 (PNPLA3), which associates with metabolic dysfunction-associated steatotic liver disease, also modulates diet-related metabolic effects. METHODS Participants with type 2 diabetes were randomized to 8-week hypocaloric diets (energy intake: -1,256 kJ/d of, <30 kcal% fat): high in cereal fiber and coffee excluding red meat (HF-RM + C; n = 16), or low in cereal fiber, devoid of coffee, but high in red meat (LF + RM-C; n = 15). Whole-body insulin sensitivity (M value) was assessed using [2H]glucose and hyperinsulinemic-normoglycemic clamps, hepatic lipid content (HCL) and body fat volumes by magnetic resonance spectroscopy/imaging before and after intervention. RESULTS Despite comparable weight loss, HCL decreased more in non-carriers (-65 %) than in G-allele carriers (-36 %) upon HF-RM + C diet (both p < 0.05 vs baseline and between groups), but only among non-carriers (-46 %, p < 0.05 vs baseline) upon LF + RM-C. Upon HF-RM + C diet, increase in insulin sensitivity was not different between carriers (+27 % p = 0.051 from baseline) and non-carriers (+21 %, p = 0.032 from baseline), p > 0.05 for between-group comparison. Upon LF + RM-C diet, both groups equally improved their whole-body insulin sensitivity (+42 % for non-carriers and +37 % for carriers, p < 0.05 vs baseline). Upon HF-RM + C diet, non-carriers decreased circulating interleukin-18 from baseline by -31 %, whereas, upon LF + RM-C diet, non-carriers decreased circulating anti-inflammatory interleukin-1 receptor antagonist levels by 14 % (both p < 0.05 vs baseline). CONCLUSIONS Humans with the PNPLA3 G-allele show modified dietary-induced effects on steatotic liver disease in type 2 diabetes despite body weight reduction. Registration at Clinicaltrials.gov, Identifier number: NCT01409330.
Collapse
Affiliation(s)
- Kalliopi Pafili
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Oana-Patricia Zaharia
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Klaus Strassburger
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Birgit Knebel
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Christian Herder
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Maximilian Huttasch
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Yanislava Karusheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany
| | - Stefan Kabisch
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Department of Endocrinology and Metabolic Medicine, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Bettina Nowotny
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; Bayer AG, Research and Development Pharmaceuticals, Aprather Weg 42113 Wuppertal, Germany
| | - Julia Szendroedi
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; Department for Internal Medicine I, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| |
Collapse
|
2
|
Wanders L, Gijbels A, Hul GB, Feskens EJM, Afman LA, Blaak EE, Hopman MTE, Goossens GH, Thijssen DHJ. Impact of a 12-week personalized dietary intervention on vascular function and cardiovascular risk factors. Diabetes Obes Metab 2025; 27:2601-2612. [PMID: 40013435 PMCID: PMC11965023 DOI: 10.1111/dom.16261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/28/2025]
Abstract
AIMS Individuals with liver insulin-resistant (LIR) or muscle insulin-resistant (MIR) phenotypes may respond differently to dietary interventions. Given the interaction between insulin resistance and cardiovascular risk, this sub-analysis of the PERSON study examined whether a personalized diet according to MIR or LIR phenotypes improves vascular function and cardiovascular disease risk factors. MATERIALS AND METHODS We randomized 119 participants to a 12-week low-fat, high-protein, high-fibre diet (LFHP; may be optimal for LIR) or Mediterranean diet (high in monounsaturated fat, HMUFA; may be optimal for MIR). Randomization linked the insulin-resistant (IR) phenotype to the proposed optimal diet, leading to PhenoDiet A (MIR-HMUFA and LIR-LFHP) and PhenoDiet B (MIR-LFHP and LIR-HMUFA). Before and after the intervention, vascular function (carotid artery reactivity) and cardiovascular risk factors (blood pressure, total cholesterol, HDL-cholesterol and Framingham risk score) were examined. A 7-point oral glucose tolerance test was performed to determine insulin resistance (Matsuda index and HOMA-IR) and disposition index. RESULTS Following drop-out (n = 18), 101 participants finished the intervention (54 women, 61 ± 7 years, 27.6 [26.4;30.0] kg/m2), with n = 80 available for the primary outcome of vascular function. Overall, the dietary interventions significantly decreased blood pressure, total cholesterol, HDL-cholesterol and the Framingham risk score (all p < 0.05), while vascular function was not affected (p = 0.485). Insulin resistance (p ≤ 0.001), but not disposition index (p = 0.362), was significantly improved after intervention. The Matsuda index (p = 0.078) tended to increase more and total cholesterol (p = 0.052) tended to decrease more in PhenoDiet group B than A, but other changes in outcome parameters were not significantly different between PhenoDiet groups. The LFHP diet resulted in more pronounced improvements in cholesterol, diastolic blood pressure (DBP) and insulin resistance compared with the HMUFA diet (all p < 0.05). CONCLUSION A 12-week diet improves metabolic and cardiovascular outcomes, but not vascular function in insulin-resistant adults with overweight or obesity. Whilst the LFHP diet resulted in greater improvements in cardiometabolic risk markers than the HMUFA diet, we found no significant differences between the PhenoDiet groups.
Collapse
Affiliation(s)
- Lisa Wanders
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
- TiFNWageningenThe Netherlands
| | - Anouk Gijbels
- TiFNWageningenThe Netherlands
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Gabby B. Hul
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Edith J. M. Feskens
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Lydia A. Afman
- Division of Human Nutrition and HealthWageningen UniversityWageningenThe Netherlands
| | - Ellen E. Blaak
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Maria T. E. Hopman
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
| | - Gijs H. Goossens
- Department of Human BiologyInstitute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical CenterMaastrichtThe Netherlands
| | - Dick H. J. Thijssen
- Radboud Institute for Health Sciences, Department of PhysiologyRadboud university medical centerNijmegenThe Netherlands
- Research Institute for Sport and Exercise SciencesLiverpool John Moores UniversityLiverpoolUK
| |
Collapse
|
3
|
Chen X, Li J, Shan Y, Wang Q, Xu P, Qian H, Wu Y. Comparative Transcriptome Analysis Reveals the Effects of a High-Protein Diet on Silkworm Midgut. INSECTS 2025; 16:337. [PMID: 40332774 PMCID: PMC12027703 DOI: 10.3390/insects16040337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/02/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025]
Abstract
The silkworm is a species within the order Lepidoptera and an economic insect. The nutrients are obtained from the leaf and utilized by the silkworm larvae for body growth, development, and cocoon formation. Protein plays a significant functional role in the diet of silkworms. To investigate the impact of the high-protein diet (HPD 6%) on silkworm growth and development, transcriptomic analysis was conducted on the silkworm midgut, and 1724 differentially expressed genes (DEGs) were identified, comprising 803 up-regulated genes and 921 down-regulated genes. The up-regulated genes exhibited the majority pathway of mitochondrial oxidative phosphorylation, ribosome, and ribosome biogenesis in eukaryotes. The down-regulated genes of DEGs were mostly annotated in ABC transporters, lysosome, endocytosis, and sphingolipid metabolism pathways. The comprehensive analysis of DEGs indicated that substantial modifications were observed in various pathways associated with crucial biological processes. HPD 6% decreased oxidative stress and increased mitochondrial activity, ribosomal activity, and DNA repair capacity. Additionally, the ATP levels were increased in the midgut, malpighian tubule, middle silk gland, and posterior silk gland of the HPD 6% group. Moreover, the activities of SOD and NADH were enhanced in the midgut of the HPD 6% group. Our findings provide valuable insights into the wide-ranging effects of an HPD treatment in insects such as silkworms.
Collapse
Affiliation(s)
- Xinyi Chen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jiahao Li
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yuxi Shan
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Qiaoling Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Pingzhen Xu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Heying Qian
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yangchun Wu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (X.C.); (J.L.); (Y.S.); (Q.W.); (P.X.); (H.Q.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
4
|
Whelehan G, Dirks ML, West S, Abdelrahman DR, Murton AJ, Finnigan TJA, Wall BT, Stephens FB. High-protein vegan and omnivorous diets improve peripheral insulin sensitivity to a similar extent in people with type 2 diabetes. Diabetes Obes Metab 2025; 27:1143-1152. [PMID: 39604044 PMCID: PMC11802395 DOI: 10.1111/dom.16100] [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: 10/10/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND High-protein diets have been recognized as a potential strategy in the nutritional management of type 2 diabetes (T2D). Mycoprotein is a high-fibre, high-protein food ingredient previously shown to improve acute glycaemic control. We determined whether incorporating mycoprotein into a high-protein vegan diet would improve glycaemic control to a greater extent than an isonitrogenous omnivorous diet in people with T2D. METHODS Seventeen adults (f = 5, age = 58.3 ± 8.3 years, BMI = 32.9 ± 4.7 kg∙m-2, HbA1c = 60 ± 15 mmol∙mol-1) with T2D were randomly allocated to a 5-week eucaloric high-protein (30% energy from protein) diet, either an omnivorous diet (OMNI; 70% protein from omnivorous sources) or an isonitrogenous, mycoprotein-rich, vegan diet (VEG; 50% protein from mycoprotein). Glycaemic control was assessed using a two-step hyperinsulinaemic-euglycaemic clamp (HEC) with D-[6,6-2H2] glucose infusion, a mixed-meal tolerance test (MMTT) and continuous glucose monitoring. RESULTS The rate of glucose disappearance (RdT), glucose disposal rate and endogenous glucose production, as well as postprandial time-course of blood glucose, serum insulin and C-peptide were assessed during the HEC and MMTT, respectively. Both groups had improved peripheral insulin sensitivity (intervention effect, p = 0.006; increased RdT/Insulin of 1.0 ± 0.6 and 1.0 ± 0.3 mg kg-1 min-1 in OMNI and VEG, respectively), HbA1c (intervention; p = 0.001) and glycaemic variability (intervention; p = 0.040; increased time in-range of 11.8 ± 9.3% and 23.3 ± 12.9% in OMNI and VEG). There were no improvements in hepatic insulin sensitivity or in postprandial blood glucose and serum C-peptide (p > 0.05) during the MMTT. CONCLUSIONS High-protein diets, whether predicated on vegan or omnivorous proteins, can improve glycaemic control by increasing peripheral insulin sensitivity in people with T2D.
Collapse
Affiliation(s)
- Gráinne Whelehan
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Marlou L. Dirks
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
- Human and Animal PhysiologyWageningen UniversityWageningenThe Netherlands
| | - Sam West
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Doaa R. Abdelrahman
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexasUSA
- Sealy Center of AgingUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Andrew J. Murton
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTexasUSA
- Sealy Center of AgingUniversity of Texas Medical BranchGalvestonTexasUSA
| | | | - Benjamin T. Wall
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Francis B. Stephens
- Department of Public Health and Sport Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| |
Collapse
|
5
|
Mohammadi S, Ashtary-Larky D, Mehrbod M, Kouhi Sough N, Salehi Omran H, Dolatshahi S, Amirani N, Asbaghi O. Impacts of supplementation with milk proteins on inflammation: a systematic review and meta-analysis. Inflammopharmacology 2025; 33:1061-1083. [PMID: 39775243 DOI: 10.1007/s10787-024-01615-8] [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: 10/03/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Impacts of milk proteins (MPs) on inflammation are uncertain. The current systematic review and dose-response meta-analysis of randomized controlled trials (RCTs) evaluated the effects of whey protein (WP), casein protein (CP), or MP supplementation on serum levels of cytokines and adipokines in adults. METHODS A comprehensive search of various online databases was conducted to find appropriate clinical trials published until September 2024. A random-effect statistical model was implemented. RESULTS The meta-analysis included 53 RCTs. It was indicated that MP supplements had no substantial effects on serum values of C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), adiponectin, and leptin. However, there were statistically significant decreases in serum levels of interleukin-6 (IL-6) following supplementation with MP (weighted mean difference (WMD): - 0.25 pg/mL, 95% CI - 0.48, - 0.03; P = 0.026) in the intervention group compared with the control group. CONCLUSION This study revealed that MP supplementation may not have any considerable impacts on the levels of cytokines and adipokines.
Collapse
Affiliation(s)
- Shooka Mohammadi
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Damoon Ashtary-Larky
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Milad Mehrbod
- School of Medicine, University of Louisville, Louisville, KY, USA
| | | | - Hossein Salehi Omran
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Dolatshahi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niusha Amirani
- Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Omid Asbaghi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Kabisch S, Hajir J, Sukhobaevskaia V, Weickert MO, Pfeiffer AFH. Impact of Dietary Fiber on Inflammation in Humans. Int J Mol Sci 2025; 26:2000. [PMID: 40076626 PMCID: PMC11900212 DOI: 10.3390/ijms26052000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Cohort studies consistently show that a high intake of cereal fiber and whole-grain products is associated with a decreased risk of type 2 diabetes (T2DM), cancer, and cardiovascular diseases. Similar findings are also reported for infectious and chronic inflammatory disorders. All these disorders are at least partially caused by inflammaging, a chronic state of inflammation associated with aging and Metabolic Syndrome. Surprisingly, insoluble (cereal) fiber intake consistently shows stronger protective associations with most long-term health outcomes than soluble fiber. Most humans consume soluble fiber mainly from sweet fruits, which usually come with high levels of sugar, counteracting the potentially beneficial effects of fiber. In both observational and interventional studies, high-fiber diets show a beneficial impact on inflammation, which can be attributed to a variety of nutrients apart from dietary fiber. These confounders need to be considered when evaluating the effects of fiber as part of complex dietary patterns. When assessing specific types of fiber, inulin and resistant starch clearly elicit anti-inflammatory short-term effects, while results for pectins, beta-glucans, or psyllium turn out to be less convincing. For insoluble fiber, promising but sparse data have been published so far. Hypotheses on putative mechanisms of anti-inflammatory fiber effects include a direct impact on immune cells (e.g., for pectin), fermentation to pleiotropic short-chain fatty acids (for fermentable fiber only), modulation of the gut microbiome towards higher levels of diversity, changes in bile acid metabolism, a differential release of gut hormones (such as the glucose-dependent insulinotropic peptide (GIP)), and an improvement of insulin resistance via the mTOR/S6K1 signaling cascade. Moreover, the contribution of phytate-mediated antioxidative and immune-modulatory means of action needs to be considered. In this review, we summarize the present knowledge on the impact of fiber-rich diets and dietary fiber on the human inflammatory system. However, given the huge heterogeneity of study designs, cohorts, interventions, and outcomes, definite conclusions on which fiber to recommend to whom cannot yet be drawn.
Collapse
Affiliation(s)
- Stefan Kabisch
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Jasmin Hajir
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Varvara Sukhobaevskaia
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism; The ARDEN NET Centre, ENETS CoE; University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Centre of Applied Biological & Exercise Sciences (ABES), Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Translational & Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| |
Collapse
|
7
|
Barber TM, Kabisch S, Pfeiffer AFH, Weickert MO. Optimised Skeletal Muscle Mass as a Key Strategy for Obesity Management. Metabolites 2025; 15:85. [PMID: 39997710 PMCID: PMC11857510 DOI: 10.3390/metabo15020085] [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: 01/07/2025] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/26/2025] Open
Abstract
The 'Body Mass Index' (BMI) is an anachronistic and outdated ratio that is used as an internationally accepted diagnostic criterion for obesity, and to prioritise, stratify, and outcome-assess its management options. On an individual level, the BMI has the potential to mislead, including inaccuracies in cardiovascular risk assessment. Furthermore, the BMI places excessive emphasis on a reduction in overall body weight (rather than optimised body composition) and contributes towards a misunderstanding of the quiddity of obesity and a dispassionate societal perspective and response to the global obesity problem. The overall objective of this review is to provide an overview of obesity that transitions away from the BMI and towards a novel vista: viewing obesity from the perspective of the skeletal muscle (SM). We resurrect the SM as a tissue hidden in plain sight and provide an overview of the key role that the SM plays in influencing metabolic health and efficiency. We discuss the complex interlinks between the SM and the adipose tissue (AT) through key myokines and adipokines, and argue that rather than two separate tissues, the SM and AT should be considered as a single entity: the 'Adipo-Muscle Axis'. We discuss the vicious circle of sarcopenic obesity, in which aging- and obesity-related decline in SM mass contributes to a worsened metabolic status and insulin resistance, which in turn further compounds SM mass and function. We provide an overview of the approaches that can mitigate against the decline in SM mass in the context of negative energy balance, including the optimisation of dietary protein intake and resistance physical exercises, and of novel molecules in development that target the SM, which will play an important role in the future management of obesity. Finally, we argue that the Adipo-Muscle Ratio (AMR) would provide a more clinically meaningful descriptor and definition of obesity than the BMI and would help to shift our focus regarding its effective management away from merely inducing weight loss and towards optimising the AMR with proper attention to the maintenance and augmentation of SM mass and function.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV1 5FB, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
| | - Stefan Kabisch
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV1 5FB, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
8
|
Davies IG. Exploring high-protein diets in the context of cardiac rehabilitation. Proc Nutr Soc 2025; 84:75-86. [PMID: 37877360 DOI: 10.1017/s0029665123004779] [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: 10/26/2023]
Abstract
The review aims to explore the potential benefit and risk of high-protein diets (HPD) regarding the comorbidity of sarcopoenia and CVD in the setting of cardiac rehabilitation (CR). CR is standard care for individuals who have experienced a cardiac event, but the current practice of predominantly aerobic exercise, a lower-fat diet and weight loss poorly addresses the issue of sarcopoenia. HPD, especially when combined with resistance exercise (RE), may be valuable adjuncts to current CR practice and benefit both muscle and cardiovascular health. Meta-analyses and randomised controlled trials of HPD and CVD risk show beneficial but variable effects regarding weight loss, the lipid profile, insulin resistance and lean body mass in those living with or high risk of CVD. Meta-analyses of prospective cohort studies on hard CVD endpoints favour lower- and plant-protein diets over higher animal protein, but the evidence is inconsistent. HPD augment the strength and muscle gaining benefits of RE in older populations, but there are no published data in those living with CVD providing promising opportunities for CR research. HPD raise concern regarding renal and bone health, the microbiome, branched chain amino acids and environmental sustainability and findings suggest that plant-based HPD may confer ecological and overall health advantages compared to animal-based HPD. However, incorporating RE with HPD might alleviate certain health risks. In conclusion, a largely plant-based HPD is deemed favourable for CR when combined with RE, but further research regarding efficacy and safety in CR populations is needed.
Collapse
Affiliation(s)
- Ian G Davies
- Research Institute of Sports and Exercise Sciences, Student Life Building, Liverpool John Moores University, Copperas Hill, Liverpool L3 5LJ, UK
| |
Collapse
|
9
|
Rajewski P, Cieściński J, Rajewski P, Suwała S, Rajewska A, Potasz M. Dietary Interventions and Physical Activity as Crucial Factors in the Prevention and Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease. Biomedicines 2025; 13:217. [PMID: 39857800 PMCID: PMC11760440 DOI: 10.3390/biomedicines13010217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide and affects nearly 30% of the adult population and 10% of the pediatric population. It is estimated that this number will double by 2030. MASLD is one of the leading causes of hepatocellular carcinoma, cirrhosis, and liver transplantation, as well as a significant risk factor for cardiovascular disease and mortality. Due to the ever-increasing number of patients, the long-term asymptomatic course of the disease, serious complications, and lack of preventive programs, as well as insufficient awareness of the disease among patients and doctors themselves, MASLD is a growing interdisciplinary problem and a real challenge for modern medicine. The main cause of MASLD is an inappropriate lifestyle-inadequate nutrition and insufficient physical activity, which lead to various components of metabolic syndrome. Lifestyle changes-appropriate diet, weight reduction, and systematic physical activity-are also the basis for the prevention and treatment of MASLD. Hence, in recent years, so much importance has been attached to lifestyle medicine, to non-pharmacological treatment as prevention of lifestyle diseases. The narrative review presents possible therapeutic options for non-pharmacological management in the prevention and treatment of MASLD. The best documented and available diets used in MASLD were discussed, focusing on the benefits and drawbacks of the Mediterranean, high-protein, ketogenic, and intermittent fasting diets. In addition, the most recent recommendations regarding physical activity are summarized.
Collapse
Affiliation(s)
- Paweł Rajewski
- Department of Internal and Infectious Diseases, Provincial Infectious Disease Hospital, 85-030 Bydgoszcz, Poland
- Faculty of Health Sciences, University of Health Sciences in Bydgoszcz, 85-067 Bydgoszcz, Poland
| | - Jakub Cieściński
- Department of Radiology, Provincial Infectious Disease Hospital, 85-030 Bydgoszcz, Poland;
| | - Piotr Rajewski
- Department of Neurology, Collegium Medicum—Faculty of Medicine, Nicolaus Copernicus University in Toruń, 85-094 Bygoszcz, Poland;
| | - Szymon Suwała
- Department of Endocrinology and Diabetology, Collegium Medicum—Faculty of Medicine, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland;
| | - Alicja Rajewska
- University Clinical Hospital, 60-355 Poznań, Poland; (A.R.); (M.P.)
| | - Maciej Potasz
- University Clinical Hospital, 60-355 Poznań, Poland; (A.R.); (M.P.)
| |
Collapse
|
10
|
Reid-McCann RJ, Brennan SF, Ward NA, Logan D, McKinley MC, McEvoy CT. Effect of Plant Versus Animal Protein on Muscle Mass, Strength, Physical Performance, and Sarcopenia: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Nutr Rev 2025:nuae200. [PMID: 39813010 DOI: 10.1093/nutrit/nuae200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
CONTEXT Dietary protein is recommended for sarcopenia-a debilitating condition of age-related loss of muscle mass and strength that affects 27% of older adults. The effects of protein on muscle health may depend on protein quality. OBJECTIVE The aim was to synthesize randomized controlled trial (RCT) data comparing plant with animal protein for muscle health. DATA SOURCES Forty-three eligible RCTs were sourced from Medline, Embase, Scopus, Web of Science, and CENTRAL databases. DATA EXTRACTION Four reviewers (R.J.R.-M., S.F.B., N.A.W., D.L.) extracted data from RCTs (study setting, population, intervention characteristics, outcomes, summary statistics) and conducted quality assessment using the Cochrane Risk of Bias 2.0. DATA ANALYSIS Standardized mean differences (SMDs) (95% CIs) were combined using a random-effects meta-analysis and forest plots were generated. I2 statistics were calculated to test for statistical heterogeneity. CONCLUSION Thirty RCTs (70%) were eligible for meta-analysis and all examined muscle mass outcomes. Compared with animal protein, plant protein resulted in lower muscle mass following the intervention (SMD = -0.20; 95% CI: -0.37, -0.03; P = .02), with stronger effects in younger (<60 years; SMD = -0.20; 95% CI: -0.37, -0.03; P = .02) than in older (≥60 years; SMD = -0.05; 95% CI: -0.32, 0.23; P = .74) adults. There was no pooled effect difference between soy and milk protein for muscle mass (SMD = -0.02; 95% CI: -0.20, 0.16; P = .80) (n = 17 RCTs), yet animal protein improved muscle mass compared with non-soy plant proteins (rice, chia, oat, and potato; SMD = -0.58; 95% CI: -1.06, -0.09; P = .02) (n = 5 RCTs) and plant-based diets (SMD = -0.51; 95% CI: -0.91, -0.11; P = .01) (n = 7 RCTs). No significant difference was found between plant or animal protein for muscle strength (n = 14 RCTs) or physical performance (n = 5 RCTs). No trials examined sarcopenia as an outcome. Animal protein may have a small beneficial effect over non-soy plant protein for muscle mass; however, research into a wider range of plant proteins and diets is needed. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42020188658.
Collapse
Affiliation(s)
- Rachel J Reid-McCann
- Nutrition and Metabolism Research Group, Centre for Public Health, Queen's University Belfast Royal Victoria Hospital, Belfast BT12 6BJ, United Kingdom
| | - Sarah F Brennan
- Nutrition and Metabolism Research Group, Centre for Public Health, Queen's University Belfast Royal Victoria Hospital, Belfast BT12 6BJ, United Kingdom
| | - Nicola A Ward
- Nutrition and Metabolism Research Group, Centre for Public Health, Queen's University Belfast Royal Victoria Hospital, Belfast BT12 6BJ, United Kingdom
| | - Danielle Logan
- Nutrition and Metabolism Research Group, Centre for Public Health, Queen's University Belfast Royal Victoria Hospital, Belfast BT12 6BJ, United Kingdom
| | - Michelle C McKinley
- Nutrition and Metabolism Research Group, Centre for Public Health, Queen's University Belfast Royal Victoria Hospital, Belfast BT12 6BJ, United Kingdom
| | - Claire T McEvoy
- Nutrition and Metabolism Research Group, Centre for Public Health, Queen's University Belfast Royal Victoria Hospital, Belfast BT12 6BJ, United Kingdom
| |
Collapse
|
11
|
Eckart AC, Sharma Ghimire P. Exploring Predictors of Type 2 Diabetes Within Animal-Sourced and Plant-Based Dietary Patterns with the XGBoost Machine Learning Classifier: NHANES 2013-2016. J Clin Med 2025; 14:458. [PMID: 39860464 PMCID: PMC11766419 DOI: 10.3390/jcm14020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Understanding the relationship between dietary patterns, nutrient intake, and chronic disease risk is critical for public health strategies. However, confounding from lifestyle and individual factors complicates the assessment of diet-disease associations. Emerging machine learning (ML) techniques offer novel approaches to clarifying the importance of multifactorial predictors. This study investigated the associations between animal-sourced and plant-based dietary patterns and Type 2 diabetes (T2D) history, accounting for diet-lifestyle patterns employing the XGBoost algorithm. Methods: Using data from the National Health and Nutrition Examination Survey (NHANES) from 2013 to 2016, individuals consuming animal-sourced foods (ASF) and plant-based foods (PBF) were propensity score-matched on key confounders, including age, gender, body mass index, energy intake, and physical activity levels. Predictors of T2D history were analyzed using the XGBoost classifier, with feature importance derived from Shapley plots. Lifestyle and dietary patterns derived from principal component analysis (PCA) were incorporated as predictors, and high multicollinearity among predictors was examined. Results: A total of 2746 respondents were included in the analysis. Among the top predictors of T2D were age, BMI, unhealthy lifestyle, and the ω6: ω3 fatty acid ratio. Higher intakes of protein from ASFs and fats from PBFs were associated with lower T2D risk. The XGBoost model achieved an accuracy of 83.4% and an AUROC of 68%. Conclusions: This study underscores the complex interactions between diet, lifestyle, and body composition in T2D risk. Machine learning techniques like XGBoost provide valuable insights into these multifactorial relationships by mitigating confounding and identifying key predictors. Future research should focus on prospective studies incorporating detailed nutrient analyses and ML approaches to refine prevention strategies and dietary recommendations for T2D.
Collapse
Affiliation(s)
- Adam C. Eckart
- Department of Health and Human Performance, Kean University, Union, NJ 07083, USA;
| | | |
Collapse
|
12
|
Hamamah S, Iatcu OC, Covasa M. Dietary Influences on Gut Microbiota and Their Role in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Nutrients 2024; 17:143. [PMID: 39796579 PMCID: PMC11722922 DOI: 10.3390/nu17010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major contributor to liver-related morbidity, cardiovascular disease, and metabolic complications. Lifestyle interventions, including diet and exercise, are first line in treating MASLD. Dietary approaches such as the low-glycemic-index Mediterranean diet, the ketogenic diet, intermittent fasting, and high fiber diets have demonstrated potential in addressing the metabolic dysfunction underlying this condition. The development and progression of MASLD are closely associated with taxonomic shifts in gut microbial communities, a relationship well-documented in the literature. Given the importance of diet as a primary treatment for MASLD, it is important to understand how gut microbiota and their metabolic byproducts mediate favorable outcomes induced by healthy dietary patterns. Conversely, microbiota changes conferred by unhealthy dietary patterns such as the Western diet may induce dysbiosis and influence steatotic liver disease through promoting hepatic inflammation, up-regulating lipogenesis, dysregulating bile acid metabolism, increasing insulin resistance, and causing oxidative damage in hepatocytes. Although emerging evidence has identified links between diet, microbiota, and development of MASLD, significant gaps remain in understanding specific microbial roles, metabolite pathways, host interactions, and causal relationships. Therefore, this review aims to provide mechanistic insights into the role of microbiota-mediated processes through the analysis of both healthy and unhealthy dietary patterns and their contribution to MASLD pathophysiology. By better elucidating the interplay between dietary nutrients, microbiota-mediated processes, and the onset and progression of steatotic liver disease, this work aims to identify new opportunities for targeted dietary interventions to treat MASLD efficiently.
Collapse
Affiliation(s)
- Sevag Hamamah
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA;
| | - Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| |
Collapse
|
13
|
Pan L, Wang L, Ma H, Ding F. Relevance of combined influence of nutritional and inflammatory status on non-alcoholic fatty liver disease and advanced fibrosis: A mediation analysis of lipid biomarkers. J Gastroenterol Hepatol 2024; 39:2853-2862. [PMID: 39392197 DOI: 10.1111/jgh.16760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/27/2024] [Accepted: 09/22/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND AIM This study aimed to investigate the relationship between advanced lung cancer inflammation index (ALI) and non-alcoholic fatty liver disease (NAFLD) and advanced liver fibrosis (AF). METHODS A total of 5642 individuals from the National Health and Nutrition Examination Survey (NHANES) between 2017 and 2020 were examined. Limited cubic spline regression model, and weighted logistic regression were employed to determine if ALI levels were related to the prevalence of NAFLD and AF. Additionally, a mediating analysis was conducted to investigate the role of lipid biomarkers, such as total cholesterol (TC) and high-density lipoprotein cholesterol (HDL-C), in the effects of ALI on the prevalence of NAFLD and AF. RESULTS After adjusting for potential confounders, a significant positive association was found between ALI with NAFLD and AF prevalence. Compared with those in ALI Tertile 1, participants in Tertile 3 had higher odds of NAFLD prevalence (odds ratio [OR]: 3.16; 95% confidence interval [CI]: 2.52-3.97) and AF (OR: 3.17; 95% CI: 2.30-4.36). Participants in both Tertile 2 and Tertile 3 had lower odds of developing AF (P for trend = 0.005). Moreover, we discovered a nonlinear association between ALI and NAFLD. An inflection point of 74.25 for NAFLD was identified through a two-segment linear regression model. Moreover, TC and HDL-C levels mediated the association between ALI and NAFLD by 10.2% and 4.2%, respectively (both P < 0.001). CONCLUSION Our findings suggest that higher ALI levels are positively associated with an increased prevalence of NAFLD and AF, partly mediated by lipid biomarkers.
Collapse
Affiliation(s)
- Lei Pan
- Department of Histology and embryology, Hebei Medical University, Shijiazhuang, China
| | - Lixuan Wang
- Department of Histology and embryology, Hebei Medical University, Shijiazhuang, China
| | - Huijuan Ma
- Department of physiology, Hebei Medical University, Shijiazhuang, China
| | - Fan Ding
- Hubei Jingmen Maternal and Child Health Hospital, Jingmen, China
| |
Collapse
|
14
|
Lépine G, Mariotti F, Tremblay-Franco M, Courrent M, Verny MA, David J, Mathé V, Jame P, Anchisi A, Lefranc-Millot C, Perreau C, Guérin-Deremaux L, Chollet C, Castelli F, Chu-Van E, Huneau JF, Rémond D, Pickering G, Fouillet H, Polakof S. Increasing plant protein in the diet induces changes in the plasma metabolome that may be beneficial for metabolic health. A randomized crossover study in males. Clin Nutr 2024; 43:146-157. [PMID: 39454458 DOI: 10.1016/j.clnu.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND & AIM Dietary shifts replacing animal protein (AP) with plant protein (PP) sources have been associated with lowering cardiometabolic risk (CMR), but underlying mechanisms are poorly characterized. This nutritional intervention aims to characterize the metabolic changes induced by diets containing different proportions of AP and PP sources in males at CMR. DESIGN This study is a 4-week, crossover, randomized, controlled-feeding trial in which 19 males with CMR followed two diets providing either 36 % for the control diet (CON-D) or 64 % for the flexitarian diet (FLEX-D) of total protein intake from PP sources. Plasma nontargeted metabolomes (LC-MS method) were measured in the fasted state and after a high-fat challenge meal at the end of each intervention arm. Lipogenesis and protein synthesis fluxes, flow-mediated dilatation (FMD) and gluco-lipidic responses were assessed after the challenge meal. Data were analyzed with mixed models, and univariate and multivariate models for metabolomics data. RESULTS In both arms CMR improved with time, with decreased body weight (-0.9 %), insulin resistant (-34 %, HOMA-IR, Homeostatic Model Assessment for Insulin Resistance) and low-density lipoproteins (LDL)-cholesterol (-11 %). Diet had no effect on FMD or metabolic fluxes, but a trend (0.05 CONCLUSIONS Despite little changes in risk factors after 4 wk, this study evidenced subtle metabolic adaptations in amino acids and lipid metabolism and gut microbiota activity occurring after higher PP source intake that may be beneficial to CMR. CLINICALTRIALS GOV STUDY IDENTIFIER NCT04236518. CLINICAL TRIAL REGISTRY NCT04236518 on ClinicalTrials.gov.
Collapse
Affiliation(s)
- Gaïa Lépine
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France; Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - François Mariotti
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Marie Tremblay-Franco
- Toxalim - Research Centre in Food Toxicology, Toulouse University, INRAE, ENVT, INP-Purpan, UT3, F-31300, Toulouse, France; MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, 31077, France
| | | | - Marie-Anne Verny
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France
| | - Jérémie David
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France
| | - Véronique Mathé
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Patrick Jame
- Universite Claude Bernard Lyon 1, CNRS, ISA, UMR5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | - Anthony Anchisi
- Universite Claude Bernard Lyon 1, CNRS, ISA, UMR5280, 5 rue de la Doua, F-69100 Villeurbanne, France
| | | | | | | | - Céline Chollet
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Florence Castelli
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Emeline Chu-Van
- CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France
| | - Jean-François Huneau
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France
| | - Didier Rémond
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France
| | | | - Hélène Fouillet
- Université Paris-Saclay, AgroParisTech, INRAE, UMR PNCA, 91120, Palaiseau, France.
| | - Sergio Polakof
- Université Clermont Auvergne, INRAE, UNH, 63000, Clermont-Ferrand, France.
| |
Collapse
|
15
|
Pletsch-Borba L, Wernicke C, Machann J, Meyer NM, Huong Nguyen T, Pohrt A, Hornemann S, Gerbracht C, Pfeiffer AF, Spranger J, Mai K. Increase in PUFA and protein, and decrease in carbohydrate intake improves liver fat in 12 months and the role of weight loss as a mediator: A randomized controlled trial. Clin Nutr 2024; 43:361-369. [PMID: 39577067 DOI: 10.1016/j.clnu.2024.11.010] [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: 02/14/2023] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND & AIMS Recently, a beneficial effect of high intake of unsaturated fatty acids (UFA) and protein on intrahepatic lipids (IHL) was demonstrated over 12 months within a randomized controlled trial (the NutriAct trial). We now aimed to explore the specific macronutrient components driving this IHL improvement within this trial in middle-aged and elderly subjects (50-80 y) at risk for age-related diseases. METHODS The NutriAct trial (n = 502) analyzed the effect of a high-protein and high-UFA diet on age related diseases including fatty liver disease. Individuals who completed 3-day food records with available IHL data both at baseline and at month 12 were included in this analysis. The impact of each macronutrient (E%) on IHL (measured by magnetic resonance spectroscopy) was analyzed by linear regression analyses and mediation analysis. Adherence in the intervention group was defined as intake at month 12 of ≥1 g protein/kg bodyweight or ≥25%E UFA intake; in the control group it was defined as intake of ≥15%E protein or ≥17%E UFA. RESULTS 248 participants were included in the analyses (34 % male, median age 66 y). Although BMI changed similarly in both groups within 12 months (mean change -0.41 kg/m2 in the control and -0.70 kg/m2 in the intervention group, p within groups <0.001, p between groups = 0.09), IHL improved more strongly in the compliant intervention participants than in compliant controls (estimate of relative change 0.21 % (95 % CI 0.01, 0.40), p = 0.03). Participants with stronger increase in protein and PUFA intake and a greater decrease in carbohydrate intake showed a stronger improvement in IHL (estimate for linear relative change -0.04 % (95%CI -0.06, -0.02), estimate 4th quartile vs. 1st quartile -0.40 % (95%CI -0.65, -0.16), and 0.32 % (95%CI 0.05, 0.59), respectively). These associations were partially mediated by BMI changes. Increase in PUFA intake was also directly associated with IHL improvement independently of BMI changes (estimate for linear relative change -0.03 % (95%CI -0.05, -0.01)). CONCLUSIONS Beneficial effects of increased protein and decreased carbohydrate intake on IHL are mediated by BMI changes in middle-aged and elderly subjects. The effect of high PUFA intake on IHL improvement was partly independent of weight loss. These results give insight into the understanding of a macronutrient specific effect on IHL changes in a long-term dietary intervention. CLINICAL TRIAL REGISTRATION The trial was registered at German Clinical Trials Register (drks.de) as DRKS00010049.
Collapse
Affiliation(s)
- Laura Pletsch-Borba
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Charlotte Wernicke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Germany; Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Germany
| | - Nina Mt Meyer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany
| | - Thu Huong Nguyen
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany
| | - Anne Pohrt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biometry and Clinical Epidemiology, Germany
| | - Silke Hornemann
- Human Study Center, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
| | - Christiana Gerbracht
- Human Study Center, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Andreas Fh Pfeiffer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Human Study Center, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
| | - Joachim Spranger
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany; Max Rubner Center for Cardiovascular Metabolic Renal Research, 10115, Berlin, Germany
| | - Knut Mai
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany; Max Rubner Center for Cardiovascular Metabolic Renal Research, 10115, Berlin, Germany.
| |
Collapse
|
16
|
Mansouri F, de Simone G, Bordoni L, Gabbianelli R. The effects of nonsoy legumes consumption on serum levels of inflammatory biomarkers and Adiponectin in overweight/obese adults: A systematic review and meta-analysis of randomized controlled trials. J Nutr Biochem 2024; 133:109718. [PMID: 39103105 DOI: 10.1016/j.jnutbio.2024.109718] [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: 03/15/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
Nonsoy legumes offer many health benefits, including improved arterial function, reduced cholesterol levels, and better management of cardiovascular diseases and type 2 diabetes. This systematic review and meta-analysis aim to clarify the inconclusive findings from randomized controlled trials (RCTs) by comprehensively evaluating the effects of nonsoy legumes consumption on serum levels of inflammatory biomarkers and Adiponectin. The search encompassed databases up to January 2024, including PubMed, EMBASE, MEDLINE, Scopus, Web of Science, and Cochrane CENTRAL to retrieve all RCTs examining the effects of nonsoy legumes on inflammatory biomarkers or Adiponectin. The effect sizes quantified as mean differences (MD) and standard deviations (SD) of outcomes, and an overall effect estimate was derived using a random-effects model. RCTs examining serum levels of C-reactive protein (CRP), Interleukin-6 (IL-6), Tumor Necrosis Factor-alpha (TNF-α), Interleukin-1β (IL-1β), and Adiponectin were included in the final meta-analysis. Results revealed that consumption of nonsoy legumes increased Adiponectin serum levels (P=.0017) and reduced IL-1β serum levels (P<.0001). However, it may not significantly affect CRP (P=.2951), IL-6 (P=.2286), and TNF-α (P=.6661) levels. Subgroup analyses showed that nonsoy legumes consumption significantly decreased TNF-α serum levels in studies involving healthy participants. Additionally, sensitivity analysis using the leave-one-out method suggested a potential significant reduction in serum levels of IL-6. This study indicates that consuming nonsoy legumes can increase levels of Adiponectin and decrease serum levels of IL-1β in overweight or obese adults.
Collapse
Affiliation(s)
- Fatemeh Mansouri
- School of Advanced Studies, University of Camerino, Camerino, Macerata, Italy; Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, Camerino, Macerata, Italy
| | - Gaia de Simone
- School of Advanced Studies, University of Camerino, Camerino, Macerata, Italy; Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, Camerino, Macerata, Italy
| | - Laura Bordoni
- Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, Camerino, Macerata, Italy.
| | - Rosita Gabbianelli
- Unit of Molecular Biology and Nutrigenomics, School of Pharmacy and Health Products, University of Camerino, Camerino, Macerata, Italy.
| |
Collapse
|
17
|
Jian H, Li R, Huang X, Li J, Li Y, Ma J, Zhu M, Dong X, Yang H, Zou X. Branched-chain amino acids alleviate NAFLD via inhibiting de novo lipogenesis and activating fatty acid β-oxidation in laying hens. Redox Biol 2024; 77:103385. [PMID: 39426289 PMCID: PMC11536022 DOI: 10.1016/j.redox.2024.103385] [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: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
The adverse metabolic impacts of branched-chain amino acids (BCAA) have been elucidated are mediated by isoleucine and valine. Dietary restriction of isoleucine promotes metabolic health and increases lifespan. However, a high protein diet enriched in BCAA is presently the most useful therapeutic strategy for nonalcoholic fatty liver disease (NAFLD), yet, its underlying mechanism remains largely unknown. Fatty liver hemorrhagic syndrome (FLHS), a specialized laying hen NAFLD model, can spontaneously develop fatty liver and hepatic steatosis under a high-energy and high-protein dietary background that the pathogenesis of FLHS is similar to human NAFLD. The mechanism underlying dietary BCAA control of NAFLD development in laying hens remains unclear. Herein, we demonstrate that dietary supplementation with 67 % High BCAA has unique mitigative impacts on NAFLD in laying hens. A High BCAA diet alleviates NAFLD, by inhibiting the tryptophan-ILA-AHR axis and MAPK9-mediated de novo lipogenesis (DNL), promoting ketogenesis and energy metabolism, and activating PPAR-RXR and pexophagy to promote fatty acid β-oxidation. Furthermore, we uncover that High BCAA strongly activates ubiquitin-proteasome autophagy via downregulating UFMylation to trigger MAPK9-mediated DNL, fatty acid elongation and lipid droplet formation-related proteins ubiquitination degradation, activating PPAR-RXR and pexophagy mediated fatty acid β-oxidation and lipolysis. Together, our data highlight moderating intake of high BCAA by inhibiting the AHR/MAPK9 are promising new strategies in NAFLD and FLHS treatment.
Collapse
Affiliation(s)
- Huafeng Jian
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China; Xianghu Laboratory, Hangzhou, 311231, China
| | - Ru Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Xuan Huang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Jiankui Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Yan Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | | | - Mingkun Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Xinyang Dong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Hua Yang
- Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Xiaoting Zou
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Flores-Hernández MN, Martínez-Coria H, López-Valdés HE, Arteaga-Silva M, Arrieta-Cruz I, Gutiérrez-Juárez R. Efficacy of a High-Protein Diet to Lower Glycemic Levels in Type 2 Diabetes Mellitus: A Systematic Review. Int J Mol Sci 2024; 25:10959. [PMID: 39456742 PMCID: PMC11507302 DOI: 10.3390/ijms252010959] [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: 07/20/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Diabetes is a metabolic disease with a high worldwide prevalence and an important factor in mortality and disability in the population. Complications can be reduced or prevented with lifestyle changes in physical activity, dietary habits, and smoking cessation. High-protein diets (HPDs, >30% or >1.0 g/Kg/day) decrease hyperglycemia in part due to their content of branched-chain amino acids (BCAAs), mainly leucine. Leucine (and other BCAAs) improve glucose metabolism by directly signaling in the medio-basal hypothalamus (MBH), increasing liver insulin sensitivity. To determine the effectiveness of an HPD to lower hyperglycemia, we analyzed the results of published clinical studies focusing on the levels of fasting plasma glucose and/or glycosylated hemoglobin (HbA1c) in patients with type 2 diabetes mellitus (T2DM). We carried out a systematic search for clinical studies using HPDs. We searched five databases (Scopus, Web of Science, PubMed, Epistemonikos, and Cochrane), collecting 179 articles and finally selecting 8 articles to analyze their results. In conclusion, HPDs are an effective alternative to reduce hyperglycemia in patients with T2DM, especially so-called Paleolithic diets, due to their higher-quality protein from animal and vegetal sources and their exclusion of grains, dairy products, salt, refined fats, and added sugars.
Collapse
Affiliation(s)
- María Nelly Flores-Hernández
- Departamento de Ciencias Biomédicas, Escuela de Medicina, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico;
| | - Hilda Martínez-Coria
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (H.M.-C.); (H.E.L.-V.)
| | - Héctor E. López-Valdés
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (H.M.-C.); (H.E.L.-V.)
| | - Marcela Arteaga-Silva
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico;
| | - Isabel Arrieta-Cruz
- Departamento de Investigación Básica, División de Investigación, Instituto Nacional de Geriatría, Secretaría de Salud, Mexico City 10200, Mexico;
| | - Roger Gutiérrez-Juárez
- Departamento de Ciencias Biomédicas, Escuela de Medicina, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico;
| |
Collapse
|
19
|
Foster C, Gagnon CA, Ashraf AP. Altered lipid metabolism and the development of metabolic-associated fatty liver disease. Curr Opin Lipidol 2024; 35:200-207. [PMID: 38484227 DOI: 10.1097/mol.0000000000000933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
PURPOSE OF REVIEW An increasing amount of research has underscored the significant role of lipoproteins in the pathogenesis of metabolic-associated fatty liver disease (MAFLD). This comprehensive review examines the intricate relationship between lipoprotein abnormalities and the development of MAFLD. RECENT FINDINGS Atherogenic dyslipidemia seen in insulin resistance states play a significant role in initiating and exacerbating hepatic lipid accumulation. There are also specific genetic factors ( PNPLA3 , TM6SF2 , MBOAT7 , HSD17B13 , GCKR- P446L) and transcription factors (SREBP-2, FXR, and LXR9) that increase susceptibility to both lipoprotein disorders and MAFLD. Most monogenic primary lipid disorders do not cause hepatic steatosis unless accompanied by metabolic stress. Hepatic steatosis occurs in the presence of secondary systemic metabolic stress in conjunction with predisposing environmental factors that lead to insulin resistance. Identifying specific aberrant lipoprotein metabolic factors promoting hepatic fat accumulation and subsequently exacerbating steatohepatitis will shed light on potential targets for therapeutic interventions. SUMMARY The clinical implications of interconnection between genetic factors and an insulin resistant environment that predisposes MAFLD is many fold. Potential therapeutic strategies in preventing or mitigating MAFLD progression include lifestyle modifications, pharmacological interventions, and emerging therapies targeting aberrant lipoprotein metabolism.
Collapse
Affiliation(s)
- Christy Foster
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Alabama at Birmingham
| | - Charles A Gagnon
- University of Alabama at Birmingham Marnix E. Heersink School of Medicine, Birmingham, Alabama, USA
| | - Ambika P Ashraf
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Alabama at Birmingham
| |
Collapse
|
20
|
Seifi N, Bahari H, Foroumandi E, Hasanpour E, Nikoumanesh M, Ferns GA, Esmaily H, Ghayour‐Mobarhan M. The association of dietary indices for hyperinsulinemia and insulin resistance with the risk of metabolic syndrome: a population-based cross-sectional study. J Clin Hypertens (Greenwich) 2024; 26:832-841. [PMID: 38980195 PMCID: PMC11232453 DOI: 10.1111/jch.14832] [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: 12/30/2023] [Revised: 04/07/2024] [Accepted: 04/16/2024] [Indexed: 07/10/2024]
Abstract
We aimed to investigate the association between an empirical dietary index for hyperinsulinemia (EDIH), empirical dietary index for insulin resistance (EDIR), and MetS and its components in an adult Iranian population. In this cross-sectional study, a total of 6482 participants aged 35-65 years were recruited as part of the MASHAD cohort study. Dietary intakes were assessed using a validated food frequency questionnaire (FFQ). The International Diabetes Federation (IDF) criteria were used to define MetS. Multivariable logistic regression models were applied to determine the association between EDIH, EDIR, and MetS and its components. The mean age and BMI of participants were 48.44±8.20 years, and 27.98±4.73 kg/m2, respectively. Around 59% of the population was female. Of the total population, 35.4% had MetS. According to the full-adjusted model, there was no significant association between higher quartiles of EDIH and EDIR and odds of MetS (Q4 EDIH; OR (95%CI):0.93 (0.74-1.18), Q4 EDIR; OR (95%CI):1.14 (0.92-1.40). Regarding MetS components, EDIR was associated with increased odds of hypertension and diabetes (Q4 EDIR; OR (95%CI):1.22 (1.04-1.44) and 1.22 (1.01-1.47), respectively). EDIH was also associated with decreased odds of hypertriglyceridemia (Q4 EDIH; OR (95%CI): 0.72 (0.60-0.87)). This study showed no significant association between hyperinsulinemia and insulin resistance potential of diet and odds of MetS among Iranian adults. However, EDIR was significantly associated with increased odds of hypertension and diabetes as MetS components.
Collapse
Affiliation(s)
- Najmeh Seifi
- International UNESCO Center for Health‐Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIran
| | - Hossein Bahari
- Transplant Research CenterClinical Research InstituteMashhad University of Medical SciencesMashhadIran
| | - Elaheh Foroumandi
- Non‐Communicable Diseases Research CenterDepartment of Nutrition & BiochemistrySchool of MedicineSabzevar University of Medical SciencesSabzevarIran
| | - Elahe Hasanpour
- International UNESCO Center for Health‐Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIran
| | - Mahya Nikoumanesh
- International UNESCO Center for Health‐Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIran
| | - Gordon A. Ferns
- Division of Medical EducationBrighton & Sussex Medical School, FalmerBrightonSussexUK
| | - Habibollah Esmaily
- Department of BiostatisticsSchool of HealthMashhad University of Medical SciencesMashhadIran
- Social Determinants of Health Research CenterMashhad University of Medical SciencesMashhadIran
| | - Majid Ghayour‐Mobarhan
- International UNESCO Center for Health‐Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
21
|
Lara-Romero C, Romero-Gómez M. Treatment Options and Continuity of Care in Metabolic-associated Fatty Liver Disease: A Multidisciplinary Approach. Eur Cardiol 2024; 19:e06. [PMID: 38983581 PMCID: PMC11231815 DOI: 10.15420/ecr.2023.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/14/2024] [Indexed: 07/11/2024] Open
Abstract
The terms non-alcoholic fatty liver disease and non-alcoholic steatohepatitis have some limitations as they use exclusionary confounder terms and the use of potentially stigmatising language. Recently, a study with content experts and patients has been set to change this nomenclature. The term chosen to replace non-alcoholic fatty liver disease was metabolic dysfunction-associated steatotic liver disease (MASLD), which avoids stigmatising and helps improve awareness and patient identification. MASLD is the most common cause of chronic liver disease with an increasing prevalence, accounting for 25% of the global population. It is considered the hepatic manifestation of the metabolic syndrome with lifestyle playing a fundamental role in its physiopathology. Diet change and physical activity are the cornerstones of treatment, encompassing weight loss and healthier behaviours and a holistic approach. In Europe, there is no approved drug for MASLD to date and there is a substantial unmet medical need for effective treatments for patients with MASLD. This review not only provides an update on advances in evidence for nutrition and physical activity interventions but also explores the different therapeutic options that are being investigated and whose development focuses on the restitution of metabolic derangements and halting inflammatory and fibrogenic pathways.
Collapse
Affiliation(s)
- Carmen Lara-Romero
- Gastroenterology and Hepatology Department, Virgen del Rocío University Hospital Seville, Spain
- Clinical and Translational Research in Digestive Diseases, Institute of Biomedicine of Seville, University of Seville Seville, Spain
| | - Manuel Romero-Gómez
- Gastroenterology and Hepatology Department, Virgen del Rocío University Hospital Seville, Spain
- Clinical and Translational Research in Digestive Diseases, Institute of Biomedicine of Seville, University of Seville Seville, Spain
| |
Collapse
|
22
|
Lv H, Liu Y. Management of non-alcoholic fatty liver disease: Lifestyle changes. World J Gastroenterol 2024; 30:2829-2833. [PMID: 38947294 PMCID: PMC11212717 DOI: 10.3748/wjg.v30.i22.2829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
In this editorial, we commented on a recently released manuscript by Zeng et al in the World Journal of Gastroenterology. We focused specifically on lifestyle changes in patients with non-alcoholic fatty liver disease (NAFLD). NAFLD is a hepatic manifestation of the metabolic syndrome, which ultimately leads to advanced hepatic fibrosis, cirrhosis, and hepatocellular carcinoma and affects more than 25% of the population globally. Existing therapeutic strategies against NAFLD such as pharmacologic therapies focus on liver protection, anti-inflammation, and regulating disease-related metabolic disorder symptoms. Although several drugs are in late-stage development, potent drugs against the diseases are lacking. Additionally, existing surgical approaches such as bariatric surgery are not routinely used to treat NAFLD. Intervening in patients' unhealthy lifestyles, such as weight loss through dietary changes and exercises to ameliorate patient-associated metabolic disorders and metabolic syndrome, is the first-line treatment for patients with NAFLD. With sufficient intrinsic motivation and adherence, the management of unhealthy lifestyles can reduce the severity of the disease, improve the quality of life, and increase the survival expectancy of patients with NAFLD.
Collapse
Affiliation(s)
- Hao Lv
- Department of General Surgery, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, China
| | - Yang Liu
- Department of General Surgery, Xi'an Jiaotong University Second Affiliated Hospital, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
23
|
Liu Z, Jin P, Liu Y, Zhang Z, Wu X, Weng M, Cao S, Wang Y, Zeng C, Yang R, Liu C, Sun P, Tian C, Li N, Zeng Q. A comprehensive approach to lifestyle intervention based on a calorie-restricted diet ameliorates liver fat in overweight/obese patients with NAFLD: a multicenter randomized controlled trial in China. Nutr J 2024; 23:64. [PMID: 38872173 PMCID: PMC11170812 DOI: 10.1186/s12937-024-00968-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a globally increasing health epidemic. Lifestyle intervention is recommended as the main therapy for NAFLD. However, the optimal approach is still unclear. This study aimed to evaluate the effects of a comprehensive approach of intensive lifestyle intervention (ILI) concerning enhanced control of calorie-restricted diet (CRD), exercise, and personalized nutrition counseling on liver steatosis and extrahepatic metabolic status in Chinese overweight and obese patients with NAFLD. METHODS This study was a multicenter randomized controlled trial (RCT) conducted across seven hospitals in China. It involved 226 participants with a body mass index (BMI) above 25. These participants were randomly assigned to two groups: the ILI group, which followed a low carbohydrate, high protein CRD combined with exercise and intensive counseling from a dietitian, and a control group, which adhered to a balanced CRD along with exercise and standard counseling. The main measure of the study was the change in the fat attenuation parameter (FAP) from the start of the study to week 12, analyzed within the per-protocol set. Secondary measures included changes in BMI, liver stiffness measurement (LSM), and the improvement of various metabolic indexes. Additionally, predetermined subgroup analyses of the FAP were conducted based on variables like gender, age, BMI, ethnicity, hyperlipidemia, and hypertension. RESULTS A total of 167 participants completed the whole study. Compared to the control group, ILI participants achieved a significant reduction in FAP (LS mean difference, 16.07 [95% CI: 8.90-23.25] dB/m) and BMI (LS mean difference, 1.46 [95% CI: 1.09-1.82] kg/m2) but not in LSM improvement (LS mean difference, 0.20 [95% CI: -0.19-0.59] kPa). The ILI also substantially improved other secondary outcomes (including ALT, AST, GGT, body fat mass, muscle mass and skeletal muscle mass, triglyceride, fasting blood glucose, fasting insulin, HbA1c, HOMA-IR, HOMA-β, blood pressure, and homocysteine). Further subgroup analyses showed that ILI, rather than control intervention, led to more significant FAP reduction, especially in patients with concurrent hypertension (p < 0.001). CONCLUSION In this RCT, a 12-week intensive lifestyle intervention program led to significant improvements in liver steatosis and other metabolic indicators in overweight and obese Chinese patients suffering from nonalcoholic fatty liver disease. Further research is required to confirm the long-term advantages and practicality of this approach. TRIAL REGISTRATION This clinical trial was registered on ClinicalTrials.gov (registration number: NCT03972631) in June 2019.
Collapse
Affiliation(s)
- Zhong Liu
- Health Management Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Piaopiao Jin
- Health Management Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuping Liu
- Department of Health Management, Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Zhimian Zhang
- Health Management Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiangming Wu
- Zhejiang Nutriease Health Technology Company Limited, Hangzhou, 311121, China
| | - Min Weng
- Department of Nutrition, The First Affiliated Hospital, Kunming Medical University, Kunming, 650034, China
| | - Suyan Cao
- Health Management Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yan Wang
- Health Management Center, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Chang Zeng
- Health Management Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Rui Yang
- Healthcare Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chenbing Liu
- Health Management Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ping Sun
- Department of Health Management, Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Cuihuan Tian
- Health Management Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Nan Li
- Health Management Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qiang Zeng
- Health Management Institute, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
24
|
Wang H, Ma Q, Chen Y, Luo L, Ye J, Zhong B. Optimized strategy among diet, exercise, and pharmacological interventions for nonalcoholic fatty liver disease: A network meta-analysis of randomized controlled trials. Obes Rev 2024; 25:e13727. [PMID: 38509775 DOI: 10.1111/obr.13727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Emerging treatment methods, including exercise, diet, and drugs, for nonalcoholic fatty liver disease have been proposed. However, the differences in their efficacy have not been determined. We aimed to compare the effects of these treatments excluding surgery via a systematic review and network meta-analysis of randomized controlled trials. DATA SOURCE The data sources included PubMed, Embase, Web of Science and Cochrane up to February 1st, 2023. The endpoints consisted of body mass index (BMI), serum markers of metabolism and liver injury markers, liver fat content, and stiffness. RESULTS A total of 174 studies with 10,183 patients were included in this meta-analysis. In terms of improving BMI, Pan-agonist of peroxisome proliferator-activated receptors (PPAR) is the best treatment with the highest SUCRA (surface under the cumulative ranking) of 84.8% (mean = -3.40, 95% CI -5.55, -1.24) by the comparative effectiveness ranking. GLP-1 (glucagon-like peptide-1) has the best effect in improving the liver fat content based on the MRI-PDFF, steatosis score (SUCRA 99.7%, mean = -2.19, 95% CI -2.90, -1.48) and ballooning score (SUCRA 61.2%, mean = -0.82, 95% CI -4.46, 2.83). CONCLUSIONS Pan-agonist of PPAR was the most efficacious regimen in lowering BMIs, whereas GLP-1R agonists achieved the highest efficacy of steatosis improvement in this network meta-analysis.
Collapse
Affiliation(s)
- Hao Wang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Infectious Diseases, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qianqian Ma
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Infectious Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Youpeng Chen
- Department of Infectious Diseases, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ling Luo
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junzhao Ye
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bihui Zhong
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
25
|
Portincasa P, Khalil M, Mahdi L, Perniola V, Idone V, Graziani A, Baffy G, Di Ciaula A. Metabolic Dysfunction-Associated Steatotic Liver Disease: From Pathogenesis to Current Therapeutic Options. Int J Mol Sci 2024; 25:5640. [PMID: 38891828 PMCID: PMC11172019 DOI: 10.3390/ijms25115640] [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: 04/02/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
The epidemiological burden of liver steatosis associated with metabolic diseases is continuously growing worldwide and in all age classes. This condition generates possible progression of liver damage (i.e., inflammation, fibrosis, cirrhosis, hepatocellular carcinoma) but also independently increases the risk of cardio-metabolic diseases and cancer. In recent years, the terminological evolution from "nonalcoholic fatty liver disease" (NAFLD) to "metabolic dysfunction-associated fatty liver disease" (MAFLD) and, finally, "metabolic dysfunction-associated steatotic liver disease" (MASLD) has been paralleled by increased knowledge of mechanisms linking local (i.e., hepatic) and systemic pathogenic pathways. As a consequence, the need for an appropriate classification of individual phenotypes has been oriented to the investigation of innovative therapeutic tools. Besides the well-known role for lifestyle change, a number of pharmacological approaches have been explored, ranging from antidiabetic drugs to agonists acting on the gut-liver axis and at a systemic level (mainly farnesoid X receptor (FXR) agonists, PPAR agonists, thyroid hormone receptor agonists), anti-fibrotic and anti-inflammatory agents. The intrinsically complex pathophysiological history of MASLD makes the selection of a single effective treatment a major challenge, so far. In this evolving scenario, the cooperation between different stakeholders (including subjects at risk, health professionals, and pharmaceutical industries) could significantly improve the management of disease and the implementation of primary and secondary prevention measures. The high healthcare burden associated with MASLD makes the search for new, effective, and safe drugs a major pressing need, together with an accurate characterization of individual phenotypes. Recent and promising advances indicate that we may soon enter the era of precise and personalized therapy for MASLD/MASH.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.K.); (L.M.); (V.P.); (V.I.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.K.); (L.M.); (V.P.); (V.I.); (A.D.C.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.K.); (L.M.); (V.P.); (V.I.); (A.D.C.)
| | - Valeria Perniola
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.K.); (L.M.); (V.P.); (V.I.); (A.D.C.)
| | - Valeria Idone
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.K.); (L.M.); (V.P.); (V.I.); (A.D.C.)
- Aboca S.p.a. Società Agricola, 52037 Sansepolcro, Italy
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.K.); (L.M.); (V.P.); (V.I.); (A.D.C.)
| |
Collapse
|
26
|
Vidal-Cevallos P, Sorroza-Martínez AP, Chávez-Tapia NC, Uribe M, Montalvo-Javé EE, Nuño-Lámbarri N. The Relationship between Pathogenesis and Possible Treatments for the MASLD-Cirrhosis Spectrum. Int J Mol Sci 2024; 25:4397. [PMID: 38673981 PMCID: PMC11050641 DOI: 10.3390/ijms25084397] [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: 03/12/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a term that entails a broad spectrum of conditions that vary in severity. Its development is influenced by multiple factors such as environment, microbiome, comorbidities, and genetic factors. MASLD is closely related to metabolic syndrome as it is caused by an alteration in the metabolism of fatty acids due to the accumulation of lipids because of an imbalance between its absorption and elimination in the liver. Its progression to fibrosis is due to a constant flow of fatty acids through the mitochondria and the inability of the liver to slow down this metabolic load, which generates oxidative stress and lipid peroxidation, triggering cell death. The development and progression of MASLD are closely related to unhealthy lifestyle habits, and nutritional epigenetic and genetic mechanisms have also been implicated. Currently, lifestyle modification is the first-line treatment for MASLD and nonalcoholic steatohepatitis; weight loss of ≥10% produces resolution of steatohepatitis and fibrosis regression. In many patients, body weight reduction cannot be achieved; therefore, pharmacological treatment should be offered in particular populations.
Collapse
Affiliation(s)
- Paulina Vidal-Cevallos
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (P.V.-C.); (N.C.C.-T.); (M.U.); (E.E.M.-J.)
| | | | - Norberto C. Chávez-Tapia
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (P.V.-C.); (N.C.C.-T.); (M.U.); (E.E.M.-J.)
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
| | - Misael Uribe
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (P.V.-C.); (N.C.C.-T.); (M.U.); (E.E.M.-J.)
| | - Eduardo E. Montalvo-Javé
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (P.V.-C.); (N.C.C.-T.); (M.U.); (E.E.M.-J.)
- Department of Surgery, Faculty of Medicine, Universidad Nacional Autónoma de Mexico, Mexico City 04360, Mexico
- Hepatopancreatobiliary Clinic, Department of Surgery, Hospital General de Mexico “Dr. Eduardo Liceaga”, Mexico City 06720, Mexico
| | - Natalia Nuño-Lámbarri
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
- Department of Surgery, Faculty of Medicine, Universidad Nacional Autónoma de Mexico, Mexico City 04360, Mexico
| |
Collapse
|
27
|
Schepp M, Freuer D, Wawro N, Peters A, Heier M, Teupser D, Meisinger C, Linseisen J. Association of the habitual dietary intake with the fatty liver index and effect modification by metabotypes in the population-based KORA-Fit study. Lipids Health Dis 2024; 23:99. [PMID: 38575962 PMCID: PMC10993479 DOI: 10.1186/s12944-024-02094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is an emerging threat for public health with diet being a major risk factor in disease development and progression. However, the effects of habitual food consumption on fatty liver are still inconclusive as well as the proposed role of the individuals' metabolic profiles. Therefore, the aim of our study is to examine the associations between diet and NAFLD with an emphasis on the influence of specific metabotypes in the general population. METHODS A total of 689 participants (304 men and 385 women) of the KORA-Fit (S4) survey, a follow-up study of the population-based KORA cohort study running in the Region of Augsburg, Germany, were included in this analysis. Dietary information was derived from repeated 24-h food lists and a food frequency questionnaire. The intake of energy and energy-providing nutrients were calculated using the national food composition database. The presence of fatty liver was quantified by the fatty liver index (FLI), and metabotypes were calculated using K-means clustering. Multivariable linear regression models were used for the analysis of habitual food groups and FLI; for the evaluation of macronutrients, energy substitution models were applied. RESULTS A higher consumption of nuts and whole grains, and a better diet quality (according to Alternate Healthy Eating Index and Mediterranean Diet Score) were associated with lower FLI values, while the intake of soft drinks, meat, fish and eggs were associated with a higher FLI. The isocaloric substitution of carbohydrates with polyunsaturated fatty acids was associated with a decreased FLI, while substitution with monounsaturated fatty acids and protein showed increased FLI. Statistically significant interactions with the metabotype were observed for most food groups. CONCLUSION The consumption of plant-based food groups, including nuts and whole grains, and diet quality, were associated with lower FLI values, whereas the intake of soft drinks and products of animal origin (meat, fish, eggs) were associated with a higher FLI. The observed statistically significant interactions with the metabotype for most food groups could help to develop targeted prevention strategies on a population-based level if confirmed in independent prospective studies.
Collapse
Affiliation(s)
- M Schepp
- University of Augsburg, University Hospital Augsburg, EpidemiologyAugsburg, Germany.
| | - D Freuer
- University of Augsburg, University Hospital Augsburg, EpidemiologyAugsburg, Germany
| | - N Wawro
- University of Augsburg, University Hospital Augsburg, EpidemiologyAugsburg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - A Peters
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
| | - M Heier
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- KORA Study Centre, University Hospital Augsburg, Augsburg, Germany
| | - D Teupser
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - C Meisinger
- University of Augsburg, University Hospital Augsburg, EpidemiologyAugsburg, Germany
| | - J Linseisen
- University of Augsburg, University Hospital Augsburg, EpidemiologyAugsburg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
28
|
Meyer NMT, Pohrt A, Wernicke C, Pletsch-Borba L, Apostolopoulou K, Haberbosch L, Machann J, Pfeiffer AFH, Spranger J, Mai K. Improvement in Visceral Adipose Tissue and LDL Cholesterol by High PUFA Intake: 1-Year Results of the NutriAct Trial. Nutrients 2024; 16:1057. [PMID: 38613089 PMCID: PMC11013849 DOI: 10.3390/nu16071057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/14/2024] Open
Abstract
We assessed the effect of a dietary pattern rich in unsaturated fatty acids (UFA), protein and fibers, without emphasizing energy restriction, on visceral adipose tissue (VAT) and cardiometabolic risk profile. Within the 36-months randomized controlled NutriAct trial, we randomly assigned 502 participants (50-80 years) to an intervention or control group (IG, CG). The dietary pattern of the IG includes high intake of mono-/polyunsaturated fatty acids (MUFA/PUFA 15-20% E/10-15% E), predominantly plant protein (15-25% E) and fiber (≥30 g/day). The CG followed usual care with intake of 30% E fat, 55% E carbohydrates and 15% E protein. Here, we analyzed VAT in a subgroup of 300 participants via MRI at baseline and after 12 months, and performed further metabolic phenotyping. A small but comparable BMI reduction was seen in both groups (mean difference IG vs. CG: -0.216 kg/m2 [-0.477; 0.045], partial η2 = 0.009, p = 0.105). VAT significantly decreased in the IG but remained unchanged in the CG (mean difference IG vs. CG: -0.162 L [-0.314; -0.011], partial η2 = 0.015, p = 0.036). Change in VAT was mediated by an increase in PUFA intake (ß = -0.03, p = 0.005) and induced a decline in LDL cholesterol (ß = 0.11, p = 0.038). The NutriAct dietary pattern, particularly due to high PUFA content, effectively reduces VAT and cardiometabolic risk markers, independent of body weight loss.
Collapse
Affiliation(s)
- Nina Marie Tosca Meyer
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Anne Pohrt
- Institute of Biometry and Clinical Epidemiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Charlotte Wernicke
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Laura Pletsch-Borba
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- BIH Charité Junior Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Konstantina Apostolopoulou
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Linus Haberbosch
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- BIH Charité Junior Digital Clinician Scientist Program, BIH Biomedical Innovation Academy, Berlin Institute of Health, Charité—Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jürgen Machann
- Institute for Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, University of Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany;
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Otfried-Müller-Straße 12/1, 72076 Tübingen, Germany
- German Center for Diabetes Research, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
- Department of Human Nutrition, German Institute of Human Nutrition, Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, 10785 Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Hessische Str. 3-4, 10115 Berlin, Germany
| | - Knut Mai
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (N.M.T.M.)
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany
- Department of Human Nutrition, German Institute of Human Nutrition, Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Potsdamer Str. 58, 10785 Berlin, Germany
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Hessische Str. 3-4, 10115 Berlin, Germany
| |
Collapse
|
29
|
Clemente-Suárez VJ, Peris-Ramos HC, Redondo-Flórez L, Beltrán-Velasco AI, Martín-Rodríguez A, David-Fernandez S, Yáñez-Sepúlveda R, Tornero-Aguilera JF. Personalizing Nutrition Strategies: Bridging Research and Public Health. J Pers Med 2024; 14:305. [PMID: 38541047 PMCID: PMC10970995 DOI: 10.3390/jpm14030305] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 11/11/2024] Open
Abstract
In recent years, although life expectancy has increased significantly, non-communicable diseases (NCDs) continue to pose a significant threat to the health of the global population. Therefore, eating habits have been recognized as key modifiable factors that influence people's health and well-being. For this reason, it is interesting to study dietary patterns, since the human diet is a complex mixture of macronutrients, micronutrients, and bioactive compounds, and can modulate multiple physiological processes, including immune function, the metabolism, and inflammation. To ensure that the data we acquired were current and relevant, we searched primary and secondary sources, including scientific journals, bibliographic indexes, and databases in the last 15 years with the most relevant articles. After this search, we observed that all the recent research on NCDs suggests that diet is a critical factor in shaping an individual's health outcomes. Thus, cardiovascular, metabolic, mental, dental, and visual health depends largely on the intake, habits and patterns, and nutritional behaviors. A diet high in processed and refined foods, added sugars, and saturated fats can increase the risk of developing chronic diseases. On the other hand, a diet rich in whole, nutrient-dense foods, such as vegetables, fruits, nuts, legumes, and a high adherence to Mediterranean diet can improve health's people.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | - Helia Carmen Peris-Ramos
- Faculty of Biomedical and Health Sciences, Clinical Odontology Department, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (H.C.P.-R.); (S.D.-F.)
| | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Tajo Street, s/n, Villaviciosa de Odón, 28670 Madrid, Spain;
| | | | - Alexandra Martín-Rodríguez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (V.J.C.-S.); (J.F.T.-A.)
| | - Susana David-Fernandez
- Faculty of Biomedical and Health Sciences, Clinical Odontology Department, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain; (H.C.P.-R.); (S.D.-F.)
| | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile;
| | | |
Collapse
|
30
|
Chen G, Fan L, Yang T, Xu T, Wang Z, Wang Y, Kong L, Sun X, Chen K, Xie Q, Zhao H. Prognostic nutritional index (PNI) and risk of non-alcoholic fatty liver disease and advanced liver fibrosis in US adults: Evidence from NHANES 2017-2020. Heliyon 2024; 10:e25660. [PMID: 38390093 PMCID: PMC10881309 DOI: 10.1016/j.heliyon.2024.e25660] [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: 09/29/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Objective This study explored the potential association between the Prognostic Nutritional Index (PNI) and the incidence of non-alcoholic fatty liver disease (NAFLD) and advanced liver fibrosis (AF) in the adult population of the United States. Methods Information on 6409 participants ≥18 years old was downloaded from the U.S. National Health and Nutrition Examination Survey (NHANES) from 2017 to 2020. Multivariate analysis was combined with demographic factors to assess the relationships between PNI, NAFLD, and AF. A restricted cubic spline (RCS) was used to characterise the nonlinear association between the PNI and NAFLD and AF. Results Patients without NAFLD had substantially lower mean values for parameters such as age, lymphocyte count, neutrophil count, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammatory index (SII), total cholesterol, triglycerides, HbA1c, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) than patients with NAFLD. Interestingly, non-NAFLD patients showed a pronounced increase in serum albumin levels compared to their NAFLD counterparts. In the subset without AF, there were discernibly lower measures of NLR, age, AST, ALT, γ-glutamyl transferase, triglycerides, neutrophil count, and body mass index (BMI) than in patients with AF. It was evident that those without AF had markedly elevated mean albumin and PNI levels in comparison to AF-affected individuals. In the comprehensive multivariable framework, a direct correlation was observed between PNI and NAFLD (adjusted odds ratio[aOR] = 1.07, 95% confidence interval [CI]: 1.05-1.09; p < 0.001), whereas PNI and AF were inversely correlated (aOR = 0.92; 95% CI: 0.88-0.96; p < 0.001). Within the RCS model, a swift ascendancy was noted in the relationship between the PNI and NAFLD, peaking at approximately 52. Conversely, a non-linear inverse association was observed between PNI and AF. Conclusion Our analytical results indicate that elevated PNI levels are positively associated with an increased risk of NAFLD, but inversely related to the risk of AF. For robust validation of these observations, further research is required.
Collapse
Affiliation(s)
- Ge Chen
- Qingdao Medical College of Qingdao University, Qingdao, Shandong, China
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Liqing Fan
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Ting Yang
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Tingting Xu
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Zixuan Wang
- Qingdao Medical College of Qingdao University, Qingdao, Shandong, China
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Yan Wang
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Lingling Kong
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Xutong Sun
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| | - Kan Chen
- Laizhou Maternity and Child Healthcare Hospital, Laizhou, Shandong, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Zhao
- The First Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Shandong, China
| |
Collapse
|
31
|
Habumugisha T, Engebretsen IMS, Måren IE, Kaiser CWM, Dierkes J. Reducing meat and/or dairy consumption in adults: a systematic review and meta-analysis of effects on protein intake, anthropometric values, and body composition. Nutr Rev 2024; 82:277-301. [PMID: 37236631 PMCID: PMC10859689 DOI: 10.1093/nutrit/nuad055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023] Open
Abstract
CONTEXT Consumers are increasingly encouraged to reduce meat and dairy consumption. However, few meta-analyses of randomized controlled trials (RCTs) on the effect of reducing meat and/or dairy on (absolute) protein intake, anthropometric values, and body composition are available. OBJECTIVE The aim of this systematic review and meta-analysis was to evaluate the effect of reducing meat and/or dairy consumption on (absolute) protein intake, anthropometric values, and body composition in adults aged ≥ 45 years. DATA SOURCES The MEDLINE, Cochrane CENTRAL, Embase, ClinicalTrials.gov, and International Clinical Trials Registry Platform databases were searched up to November 24, 2021. DATA EXTRACTION Randomized controlled trials reporting protein intake, anthropometric values, and body composition were included. DATA ANALYSIS Data were pooled using random-effects models and expressed as the mean difference (MD) with 95%CI. Heterogeneity was assessed and quantified using Cochran's Q and I2 statistics. In total, 19 RCTs with a median duration of 12 weeks (range, 4-24 weeks) and a total enrollment of 1475 participants were included. Participants who consumed meat- and/or dairy-reduced diets had a significantly lower protein intake than those who consumed control diets (9 RCTs; MD, -14 g/d; 95%CI, -20 to -8; I2 = 81%). Reducing meat and/or dairy consumption had no significant effect on body weight (14 RCTs; MD, -1.2 kg; 95%CI, -3 to 0.7; I2 = 12%), body mass index (13 RCTs; MD, -0.3 kg/m2; 95%CI, -1 to 0.4; I2 = 34%), waist circumference (9 RCTs; MD, -0.5 cm; 95%CI, -2.1 to 1.1; I2 = 26%), amount of body fat (8 RCTs; MD, -1.0 kg; 95%CI, -3.0 to 1.0; I2 = 48%), or lean body mass (9 RCTs; MD, -0.4 kg; 95%CI, -1.5 to 0.7; I2 = 0%). CONCLUSION Reduction of meat and/or dairy appears to reduce protein intake. There is no evidence of a significant impact on anthropometric values or body composition. More long-term intervention studies with defined amounts of meat and dairy are needed to investigate the long-term effects on nutrient intakes and health outcomes. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42020207325.
Collapse
Affiliation(s)
- Theogene Habumugisha
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Centre for Nutrition, Mohn Nutrition Research Laboratory, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | | | | | - Jutta Dierkes
- Centre for Nutrition, Mohn Nutrition Research Laboratory, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
32
|
Guo L, Xiao X. Guideline for the Management of Diabetes Mellitus in the Elderly in China (2024 Edition). Aging Med (Milton) 2024; 7:5-51. [PMID: 38571669 PMCID: PMC10985780 DOI: 10.1002/agm2.12294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 04/05/2024] Open
Abstract
With the deepening of aging in China, the prevalence of diabetes in older people has increased noticeably, and standardized diabetes management is critical for improving clinical outcomes of diabetes in older people. In 2021, the National Center of Gerontology, Chinese Society of Geriatrics, and Diabetes Professional Committee of Chinese Aging Well Association organized experts to write the first guideline for diabetes diagnosis and treatment in older people in China, the Guideline for the Management of Diabetes Mellitus in the Elderly in China (2021 Edition). The guideline emphasizes that older patients with diabetes are a highly heterogeneous group requiring comprehensive assessment and stratified and individualized management strategies. The guideline proposes simple treatments and de-intensified treatment strategies for older patients with diabetes. This edition of the guideline provides clinicians with practical and operable clinical guidance, thus greatly contributing to the comprehensive and full-cycle standardized management of older patients with diabetes in China and promoting the extensive development of clinical and basic research on diabetes in older people and related fields. In the past 3 years, evidence-based medicine for older patients with diabetes and related fields has further advanced, and new treatment concepts, drugs, and technologies have been developed. The guideline editorial committee promptly updated the first edition of the guideline and compiled the Guideline for the Management of Diabetes Mellitus in the Elderly in China (2024 Edition). More precise management paths for older patients with diabetes are proposed, for achieving continued standardization of the management of older Chinese patients with diabetes and improving their clinical outcomes.
Collapse
Affiliation(s)
- Lixin Guo
- National Center of Gerontology, Chinese Society of Geriatrics, Diabetes Professional Committee of Chinese Aging Well AssociationBeijingChina
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Xinhua Xiao
- National Center of Gerontology, Chinese Society of Geriatrics, Diabetes Professional Committee of Chinese Aging Well AssociationBeijingChina
- Department of EndocrinologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
33
|
Roth-Walter F, Berni Canani R, O'Mahony L, Peroni D, Sokolowska M, Vassilopoulou E, Venter C. Nutrition in chronic inflammatory conditions: Bypassing the mucosal block for micronutrients. Allergy 2024; 79:353-383. [PMID: 38084827 DOI: 10.1111/all.15972] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024]
Abstract
Nutritional Immunity is one of the most ancient innate immune responses, during which the body can restrict nutrients availability to pathogens and restricts their uptake by the gut mucosa (mucosal block). Though this can be a beneficial strategy during infection, it also is associated with non-communicable diseases-where the pathogen is missing; leading to increased morbidity and mortality as micronutritional uptake and distribution in the body is hindered. Here, we discuss the acute immune response in respect to nutrients, the opposing nutritional demands of regulatory and inflammatory cells and particularly focus on some nutrients linked with inflammation such as iron, vitamins A, Bs, C, and other antioxidants. We propose that while the absorption of certain micronutrients is hindered during inflammation, the dietary lymph path remains available. As such, several clinical trials investigated the role of the lymphatic system during protein absorption, following a ketogenic diet and an increased intake of antioxidants, vitamins, and minerals, in reducing inflammation and ameliorating disease.
Collapse
Affiliation(s)
- Franziska Roth-Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine Vienna, Medical University Vienna and University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Liam O'Mahony
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Diego Peroni
- Section of Paediatrics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Emilia Vassilopoulou
- Pediatric Area, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carina Venter
- Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
34
|
Engin A. Nonalcoholic Fatty Liver Disease and Staging of Hepatic Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:539-574. [PMID: 39287864 DOI: 10.1007/978-3-031-63657-8_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is in parallel with the obesity epidemic, and it is the most common cause of liver diseases. The patients with severe insulin-resistant diabetes having high body mass index (BMI), high-grade adipose tissue insulin resistance, and high hepatocellular triacylglycerols (triglycerides; TAG) content develop hepatic fibrosis within a 5-year follow-up. Insulin resistance with the deficiency of insulin receptor substrate-2 (IRS-2)-associated phosphatidylinositol 3-kinase (PI3K) activity causes an increase in intracellular fatty acid-derived metabolites such as diacylglycerol (DAG), fatty acyl CoA, or ceramides. Lipotoxicity-related mechanism of NAFLD could be explained still best by the "double-hit" hypothesis. Insulin resistance is the major mechanism in the development and progression of NAFLD/nonalcoholic steatohepatitis (NASH). Metabolic oxidative stress, autophagy, and inflammation induce NASH progression. In the "first hit" the hepatic concentrations of diacylglycerol increase with an increase in saturated liver fat content in human NAFLD. Activities of mitochondrial respiratory chain complexes are decreased in the liver tissue of patients with NASH. Hepatocyte lipoapoptosis is a critical feature of NASH. In the "second hit," reduced glutathione levels due to oxidative stress lead to the overactivation of c-Jun N-terminal kinase (JNK)/c-Jun signaling that induces cell death in the steatotic liver. Accumulation of toxic levels of reactive oxygen species (ROS) is caused at least by two ineffectual cyclical pathways. First is the endoplasmic reticulum (ER) oxidoreductin (Ero1)-protein disulfide isomerase oxidation cycle through the downstream of the inner membrane mitochondrial oxidative metabolism and the second is the Kelch like-ECH-associated protein 1 (Keap1)-nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathways. In clinical practice, on ultrasonographic examination, the elevation of transaminases, γ-glutamyltransferase, and the aspartate transaminase to platelet ratio index indicates NAFLD. Fibrosis-4 index, NAFLD fibrosis score, and cytokeratin18 are used for grading steatosis, staging fibrosis, and discriminating the NASH from simple steatosis, respectively. In addition to ultrasonography, "controlled attenuation parameter," "magnetic resonance imaging proton-density fat fraction," "ultrasound-based elastography," "magnetic resonance elastography," "acoustic radiation force impulse elastography imaging," "two-dimensional shear-wave elastography with supersonic imagine," and "vibration-controlled transient elastography" are recommended as combined tests with serum markers in the clinical evaluation of NAFLD. However, to confirm the diagnosis of NAFLD, a liver biopsy is the gold standard. Insulin resistance-associated hyperinsulinemia directly accelerates fibrogenesis during NAFLD development. Although hepatocyte lipoapoptosis is a key driving force of fibrosis progression, hepatic stellate cells and extracellular matrix cells are major fibrogenic effectors. Thereby, these are pharmacological targets of therapies in developing hepatic fibrosis. Nonpharmacological management of NAFLD mainly consists of two alternatives: lifestyle modification and metabolic surgery. Many pharmacological agents that are thought to be effective in the treatment of NAFLD have been tried, but due to lack of ability to attenuate NAFLD, or adverse effects during the phase trials, the vast majority could not be licensed.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
35
|
Ren Z, Bosma H, Wesselius A, Eussen SJ, Kooi ME, van der Kallen CJ, Koster A, van Greevenbroek MM, Dagnelie P, Stehouwer CD, Brouwers MC. Traditional lifestyle factors partly mediate the association of socioeconomic position with intrahepatic lipid content: The Maastricht study. JHEP Rep 2023; 5:100855. [PMID: 37771365 PMCID: PMC10522893 DOI: 10.1016/j.jhepr.2023.100855] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/02/2023] [Accepted: 07/07/2023] [Indexed: 09/30/2023] Open
Abstract
Background & Aims Recent studies have unveiled an association between socioeconomic position (SEP) and intrahepatic lipid (IHL) content. The aim of this study was to examine to what extent traditional lifestyle factors mediate the relationship between SEP and IHL content, independent of aetiology, and non-alcoholic fatty liver disease (NAFLD). Methods We used cross-sectional data derived from The Maastricht Study (N = 4,001; mean age: 60 years, 49% women, 32% low education level, 21% diabetes, 21% NAFLD). Education, income, and occupation were used as indicators of SEP. Physical activity (accelerometer), intake of total energy, alcohol, saturated fat, protein, vitamin E, dietary fibre, and fructose from sugar-sweetened beverages (SSBs) and fruit juice (food frequency questionnaires) were potential mediators. IHL content was quantified by magnetic resonance imaging. Age, sex, and type 2 diabetes were covariates. Multiple parallel mediation analyses (bootstraps = 10,000) were performed. Results Individuals with a low education level had a 1.056-fold higher IHL content (95% CI: 1.03-1.08) and a 44% greater NAFLD risk (OR:1.44; 95% CI:1.18-1.77) compared with those with higher education levels. Approximately 8.9% of educational disparity in risk of IHL content was attributable to moderate-to-vigorous physical activity; 6.3% to fructose intake from SSBs; 5.5% to dietary fibre; and -23% to alcohol. Approximately 8.7% of educational disparity in risk of NAFLD was attributable to moderate-to-vigorous physical activity; and 7.7% to fructose intake from SSBs. However, the indirect effect of these mediators was small (0.998 for IHL content and 1.045 for NAFLD) in comparison to the total effect. Similar results were found when income and occupation were used as SEP indicators. Conclusions Societal measures may alleviate the burden of NAFLD and further studies that identify mediators other than traditional lifestyle factors are warranted to define the relationship underlying SEP and IHL content. Impact and implications Individuals with a low or medium level of education, income, or occupational status had more fat accumulation in their livers than individuals with a higher education, income, or occupational status. This difference may be attributed to the influence of unhealthy lifestyle factors, such as reduced physical activity and a higher intake of sugar-sweetened beverages among individuals with lower socioeconomic position. Nevertheless, other yet unknown factors may also play a role.
Collapse
Affiliation(s)
- Zhewen Ren
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Laboratory for Metabolism and Vascular Medicine, Maastricht University, Maastricht, The Netherlands
| | - Hans Bosma
- Department of Social Medicine, Maastricht University, Maastricht, The Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Anke Wesselius
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
- NUTRIM School for Nutrition and Translational Research in Metabolism Maastricht University, Maastricht, The Netherlands
| | - Simone J.P.M. Eussen
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
- NUTRIM School for Nutrition and Translational Research in Metabolism Maastricht University, Maastricht, The Netherlands
| | - M. Eline Kooi
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Carla J.H. van der Kallen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Annemarie Koster
- Department of Social Medicine, Maastricht University, Maastricht, The Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Marleen M.J. van Greevenbroek
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Laboratory for Metabolism and Vascular Medicine, Maastricht University, Maastricht, The Netherlands
| | - Pieter Dagnelie
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Coen D.A. Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Martijn C.G.J. Brouwers
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
36
|
Reddy A, Gatta PD, Mason S, Nicoll AJ, Ryan M, Itsiopoulos C, Abbott G, Johnson NA, Sood S, Roberts SK, George ES, Tierney AC. Adherence to a Mediterranean diet may improve serum adiponectin in adults with nonalcoholic fatty liver disease: The MEDINA randomized controlled trial. Nutr Res 2023; 119:98-108. [PMID: 37801761 DOI: 10.1016/j.nutres.2023.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 10/08/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects approximately 30% of adults worldwide, with chronic low-grade inflammation being a key pathophysiological feature of progression. The Mediterranean diet (MedDiet) is recognized for improving metabolic and hepatic outcomes in people with diabetes and NAFLD, in part, via anti-inflammatory properties. The aim of this study was to determine the effect of an ad libitum MedDiet versus low-fat diet (LFD) on inflammatory markers in adults with NAFLD. It was hypothesized that the MedDiet, and its individual components, would improve inflammation. This multicenter, randomized controlled trial, randomized participants to a MedDiet or LFD intervention for 12 weeks. Primary outcomes included change from baseline to 12 weeks for serum high-sensitivity C-reactive protein, interleukin-6, tumor necrosis factor-α, adiponectin, leptin, and resistin. Forty-two participants (60% female; age 52.3 ± 12.6 years; body mass index, 32.2 ± 6.2 kg/m²) were randomized to the MedDiet (n = 19) or low-fat diet (n = 23). At 12 weeks, the LFD showed a greater decrease in leptin compared with the MedDiet (-1.20 ± 3.9 ng/mL vs 0.64 ± 3.5 ng/mL, P = .010). Adiponectin significantly improved within the MedDiet (13.7 ± 9.2 µg/mL to 17.0 ± 12.5 µg/mL, P = .016), but not within the LFD group. No statistically significant changes were observed for other inflammatory markers following the MedDiet or LFD. Adherence to the MedDiet significantly improved in both study arms, although greater improvements were seen in the MedDiet group. Adiponectin significantly improved following a Mediterranean diet intervention, in the absence of weight loss. The low-fat diet did not elicit improvements in inflammatory markers. High-quality clinical trials appropriately powered to inflammatory markers are required in this population.
Collapse
Affiliation(s)
- Anjana Reddy
- School of Allied Health, Human Services and Sport, La Trobe University, Australia; Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Melbourne, VIC, Australia
| | - Paul Della Gatta
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Shaun Mason
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Amanda J Nicoll
- Department of Gastroenterology, Eastern Health, Box Hill, Australia
| | - Marno Ryan
- Department of Gastroenterology and Hepatology, St Vincent's Hospital, Fitzroy, Australia
| | - Catherine Itsiopoulos
- School of Allied Health, Human Services and Sport, La Trobe University, Australia; School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Gavin Abbott
- Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Melbourne, VIC, Australia
| | - Nathan A Johnson
- The Boden Collaboration for Obesity, Nutrition, Exercise and Eating Disorders, The University of Sydney, Sydney, New South Wales, Australia
| | - Siddharth Sood
- Department of Gastroenterology, Melbourne Health, Melbourne, Australia; Department of Medicine, University of Melbourne, Parkville, Australia
| | - Stuart K Roberts
- Department of Gastroenterology, Alfred Health, Prahran, Australia; Central Clinical School, Monash University, Clayton, Australia
| | - Elena S George
- School of Allied Health, Human Services and Sport, La Trobe University, Australia; Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| | - Audrey C Tierney
- School of Allied Health, Human Services and Sport, La Trobe University, Australia; School of Allied Health, Health Implementation Science and Technology Research Cluster, Health Research Institute, University of Limerick, Ireland
| |
Collapse
|
37
|
Lundsgaard AM, Bojsen-Møller KN, Kiens B. Dietary Regulation of Hepatic Triacylglycerol Content-the Role of Eucaloric Carbohydrate Restriction with Fat or Protein Replacement. Adv Nutr 2023; 14:1359-1373. [PMID: 37591342 PMCID: PMC10721463 DOI: 10.1016/j.advnut.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 07/21/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
Accumulation of hepatic triacylglycerol (TG) is highly associated with impaired whole-body insulin-glucose homeostasis and dyslipidemia. The summarized findings from human intervention studies investigating the effect of reduced dietary carbohydrate and increased fat intake (and in studies also increased protein) while maintaining energy intake at eucaloric requirements reveal a beneficial effect of carbohydrate reduction on hepatic TG content in obese individuals with steatosis and indices of insulin resistance. Evidence suggests that the reduction of hepatic TG content after reduced intake of carbohydrates and increased fat/protein intake in humans, results from regulation of fatty acid (FA) metabolism within the liver, with an increase in hepatic FA oxidation and ketogenesis, together with a concomitant downregulation of FA synthesis from de novo lipogenesis. The adaptations in hepatic metabolism may result from reduced intrahepatic monosaccharide and insulin availability, reduced glycolysis and increased FA availability when carbohydrate intake is reduced.
Collapse
Affiliation(s)
- Anne-Marie Lundsgaard
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| | | | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
38
|
Younossi ZM, Zelber-Sagi S, Henry L, Gerber LH. Lifestyle interventions in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2023; 20:708-722. [PMID: 37402873 DOI: 10.1038/s41575-023-00800-4] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 07/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a dynamic chronic liver disease that develops in close association with metabolic irregularities. Between 2016 and 2019, the global prevalence among adults was reported as 38% and among children and adolescents it was about 10%. NAFLD can be progressive and is associated with increased mortality from cardiovascular disease, extrahepatic cancers and liver complications. Despite these numerous adverse outcomes, no pharmacological treatments currently exist to treat nonalcoholic steatohepatitis, the progressive form of NAFLD. Therefore, the main treatment is the pursuit of a healthy lifestyle for both children and adults, which includes a diet rich in fruits, nuts, seeds, whole grains, fish and chicken and avoiding overconsumption of ultra-processed food, red meat, sugar-sweetened beverages and foods cooked at high heat. Physical activity at a level where one can talk but not sing is also recommended, including leisure-time activities and structured exercise. Avoidance of smoking and alcohol is also recommended. Policy-makers, community and school leaders need to work together to make their environments healthy by developing walkable and safe spaces with food stores stocked with culturally appropriate and healthy food items at affordable prices as well as providing age-appropriate and safe play areas in both schools and neighbourhoods.
Collapse
Affiliation(s)
- Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA.
- Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, USA.
- Inova Medicine, Inova Health System, Falls Church, VA, USA.
| | | | - Linda Henry
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
- Inova Medicine, Inova Health System, Falls Church, VA, USA
| | - Lynn H Gerber
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, USA
- Inova Medicine, Inova Health System, Falls Church, VA, USA
| |
Collapse
|
39
|
Pappe CL, Peters B, Dommisch H, Woelber JP, Pivovarova-Ramich O. Effects of reducing free sugars on 24-hour glucose profiles and glycemic variability in subjects without diabetes. Front Nutr 2023; 10:1213661. [PMID: 37850088 PMCID: PMC10577299 DOI: 10.3389/fnut.2023.1213661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/13/2023] [Indexed: 10/19/2023] Open
Abstract
Background The Western diet, especially beverages and high processed food products, is high in sugars which are associated with the development of obesity and diabetes. The reduction of refined carbohydrates including free and added sugars improves glycemic control in individuals with diabetes, but the data regarding effects in subjects without diabetes are limited. Objective This study aimed to evaluate the effects of reducing free sugar intake on 24-h glucose profiles and glycemic variability using continuous glucose monitoring (CGM). Methods In the randomized controlled study, 21 normal weight and overweight/obese subjects (BMI 18-40 kg/m2) without diabetes were assigned to a 4-week reduced-sugar (RS) diet or control diet after a 2-week baseline phase. During the baseline phase, all participants were advised not to change their habitual diet. During the intervention phase, RS participants were asked to avoid added sugar and white flour products, whereas participants of the control group were requested to proceed their habitual diet. Anthropometric parameters and HbA1c were assessed before and at the end of the intervention phase. Interstitial glucose was measured using continuous glucose monitoring (CGM), and the food intake was documented by dietary records for 14 consecutive days during the baseline phase and for the first 14 consecutive days during the intervention phase. Mean 24-h glucose as well as intra- and inter-day indices of glucose variability, i.e., standard deviation (SD) around the sensor glucose level, coefficient of variation in percent (CV), mean amplitude of glucose excursions (MAGE), continuous overlapping net glycemic action (CONGA), and mean absolute glucose (MAG), were calculated for the baseline and intervention phases. Results During the intervention, the RS group decreased the daily intake of sugar (i.e., -22.4 ± 20.2 g, -3.28 ± 3.61 EN %), total carbohydrates (-6.22 ± 6.92 EN %), and total energy intake (-216 ± 108 kcal) and increased the protein intake (+2.51 ± 1.56 EN %) compared to the baseline values, whereby this intervention-induced dietary changes differed from the control group. The RS group slightly reduced body weight (-1.58 ± 1.33 kg), BMI, total fat, and visceral fat content and increased muscle mass compared to the baseline phase, but these intervention-induced changes showed no differences in comparison with the control group. The RS diet affected neither the 24-h mean glucose levels nor intra- and inter-day indices of glucose variability, HbA1c, or diurnal glucose pattern in the within- and between-group comparisons. Conclusion The dietary reduction of free sugars decreases body weight and body fat which may be associated with reduced total energy intake but does not affect the daily mean glucose and glycemic variability in individuals without diabetes. Clinical trial registration German Clinical Trials Register (DRKS); identifier: DRKS00026699.
Collapse
Affiliation(s)
- Christina Laeticia Pappe
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Beeke Peters
- Research Group Molecular Nutritional Medicine and Department of Human Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Oberschleißheim, Germany
| | - Henrik Dommisch
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Department of Periodontology, Health Science Center, University of Washington, Seattle, WA, United States
| | - Johan Peter Woelber
- Policlinic of Operative Dentistry, Periodontology, and Pediatric Dentistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Olga Pivovarova-Ramich
- Research Group Molecular Nutritional Medicine and Department of Human Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), Oberschleißheim, Germany
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
40
|
Lv Y, Rong S, Deng Y, Bao W, Xia Y, Chen L. Plant-based diets, genetic predisposition and risk of non-alcoholic fatty liver disease. BMC Med 2023; 21:351. [PMID: 37697334 PMCID: PMC10496397 DOI: 10.1186/s12916-023-03028-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/09/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND Diets rich in plant-based foods are associated with lower risks of non-alcoholic fatty liver disease (NAFLD), while the prospective evidence is limited. We aimed to examine longitudinal associations of plant-based diets and genetic susceptibility with NAFLD risk. METHODS This longitudinal cohort study included 159,222 participants (58.0 ± 8.0 years old, 55.7% female) free of NAFLD in the UK Biobank. We calculated the overall plant-based diet index (PDI), the healthful plant-based diet index (hPDI), and the unhealthful plant-based diet index (uPDI). New-onset NAFLD was the primary outcome. The weighted polygenic risk score was calculated based on risk variants associated with NAFLD. Hazard ratios (HR) and 95% confidential intervals (CI) were estimated by Cox proportional hazards model. Magnetic resonance imaging-derived proton density fat fraction (MRI-PDFF) measured liver fat content in a subsample of 20,692 participants (57.5 ± 7.4 years old, 52.6% female) was the secondary outcome. The associations between plant-based diet indices and MRI-PDFF were evaluated using generalized linear models. RESULTS During a median follow-up of 9.5 years, 1541 new-onset NAFLD cases were documented. Compared to the lowest quintile, multivariable-adjusted hazard ratios (HRs) of NAFLD in the highest quintile were 0.78 (95% confidential intervals [CI], 0.66-0.93, p-trend =0.02), 0.74 (95% CI, 0.62-0.87, p-trend <0.0001), and 1.24 (95% CI, 1.05-1.46, p-trend = 0.02) for overall PDI, hPDI, and uPDI, respectively. For liver fat content, higher overall PDI and hPDI were associated with lower MRI-PDFF, while higher uPDI was associated with higher liver fat content. We observed a significant interaction between hPDI and PRS (p-interaction =0.03), and the NAFLD risk was lowest among participants with the highest hPDI and low genetic risk. CONCLUSIONS Higher intake of plant-based diets especially healthful plant-based diets was associated with lower NAFLD risk and liver fat content regardless of genetic susceptibility, whereas an unhealthful plant-based diet was associated with higher NAFLD risk and intrahepatic steatosis. These results suggest that the quality of plant-based foods should be highlighted when adopting a plant-based diet.
Collapse
Affiliation(s)
- Yanling Lv
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China
- Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuang Rong
- Department of Preventive Medicine, School of Public Health, Wuhan University, No.115 Donghu Road, Wuhan, 430071, Hubei, China.
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China.
| | - Yan Deng
- Department of Preventive Medicine, School of Public Health, Wuhan University, No.115 Donghu Road, Wuhan, 430071, Hubei, China
- Department of Nutrition, Hygiene and Toxicology, Academy of Nutrition and Health, School of Public Health, Medical College, Wuhan University of Science and Technology, Wuhan, 430065, China
| | - Wei Bao
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Yang Xia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
- Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
41
|
Zhang J, Zheng Y, Martens L, Pfeiffer AFH. The Regulation and Secretion of Glucagon in Response to Nutrient Composition: Unraveling Their Intricate Mechanisms. Nutrients 2023; 15:3913. [PMID: 37764697 PMCID: PMC10536047 DOI: 10.3390/nu15183913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Glucagon was initially regarded as a hyperglycemic substance; however, recent research has revealed its broader role in metabolism, encompassing effects on glucose, amino acids (AAs), and lipid metabolism. Notably, the interplay of glucagon with nutrient intake, particularly of AAs, and non-nutrient components is central to its secretion. Fasting and postprandial hyperglucagonemia have long been linked to the development and progression of type 2 diabetes (T2DM). However, recent studies have brought to light the positive impact of glucagon agonists on lipid metabolism and energy homeostasis. This review explores the multifaceted actions of glucagon, focusing on its regulation, signaling pathways, and effects on glucose, AAs, and lipid metabolism. The interplay between glucagon and other hormones, including insulin and incretins, is examined to provide a mechanistic understanding of its functions. Notably, the liver-α-cell axis, which involves glucagon and amino acids, emerges as a critical aspect of metabolic regulation. The dysregulation of glucagon secretion and its impact on conditions such as T2DM are discussed. The review highlights the potential therapeutic applications of targeting the glucagon pathway in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jiudan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| | - Yang Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Lisa Martens
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
- Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| |
Collapse
|
42
|
Cogorno L, Formisano E, Vignati A, Prigione A, Tramacere A, Borgarelli C, Sukkar SG, Pisciotta L. Non-alcoholic fatty liver disease: Dietary and nutraceutical approaches. LIVER RESEARCH 2023; 7:216-227. [PMID: 39958388 PMCID: PMC11791914 DOI: 10.1016/j.livres.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/20/2023] [Accepted: 08/24/2023] [Indexed: 02/18/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD), defined as the presence of fat accumulation in imaging or histology in more than 5% of hepatocytes and exclusion of other causes for secondary hepatic fat accumulation, is one of the major causes of chronic liver disease worldwide. Metabolic syndrome is associated with an increased risk of progression from NAFLD to non-alcoholic steatohepatitis (NASH), fibrosis, and forthcoming liver failure. Also, genetic predisposition contributes to the risk of NAFLD development. This review explores the role of diets and nutraceuticals in delaying the development and the evolution of NAFLD to chronic liver disease. The Mediterranean diet, high-protein diet, low-carbohydrate/high-fat diet, high-carbohydrate/low-fat diet, and intermittent fasting are the dietary approaches investigated given the presence of relevant literature data. Moreover, this review focused on nutraceuticals with proven efficacy in ameliorating NAFLD and grouped them into four different categories: plant-based nutraceuticals (Ascophyllum nodosum and Fucus vesiculosus, Silymarin, Berberine, Curcumin, Resveratrol, Nigella sativa, Quercetin), vitamin-like substances (vitamin E, vitamin D, vitamin C, coenzyme Q10, inositol), fatty acids (omega-3), and microbiota-management tools (probiotics).
Collapse
Affiliation(s)
- Ludovica Cogorno
- Department of Experimental Medicine-Medical Pathophysiology, Food Science and Endocrinology Section, Sapienza University of Rome, Rome, Italy
| | - Elena Formisano
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, Genoa, Italy
| | - Andrea Vignati
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Amalia Prigione
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | | | | | - Samir Giuseppe Sukkar
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, Genoa, Italy
| | - Livia Pisciotta
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- Dietetics and Clinical Nutrition Unit, IRCCS Policlinic Hospital San Martino, Genoa, Italy
| |
Collapse
|
43
|
Sandel P, Ma L, Wang H, Pasman EA. You Are What You Eat: A Review on Dietary Interventions for Treating Pediatric Nonalcoholic Fatty Liver Disease. Nutrients 2023; 15:3350. [PMID: 37571287 PMCID: PMC10421125 DOI: 10.3390/nu15153350] [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: 06/26/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
As the obesity pandemic worsens, cases of pediatric nonalcoholic fatty liver disease (NAFLD) and complications of this disease, such as progressive liver failure, in young adults will continue to rise. Lifestyle changes in the form of dietary modifications and exercise are currently first-line treatments. Large pediatric-specific randomized controlled trials to support specific interventions are currently lacking. A variety of dietary modifications in children with NAFLD have been suggested and studied with mixed results, including low-sugar and high-protein diets, the Mediterranean diet, and the Dietary Approach to Stop Hypertension (DASH). The roles of dietary supplements such as Vitamin E, polyunsaturated fatty acids (PUFAs), ginger, and probiotics have also been investigated. A further understanding of specific dietary interventions and supplements is needed to provide both generalizable and sustainable dietary recommendations to reverse the progression of NAFLD in the pediatric population.
Collapse
Affiliation(s)
- Piper Sandel
- Section of Academic General Pediatrics, Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA; (L.M.); (H.W.)
| | - Lawrence Ma
- Section of Academic General Pediatrics, Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA; (L.M.); (H.W.)
| | - Helen Wang
- Section of Academic General Pediatrics, Department of Pediatrics, University of California San Diego, San Diego, CA 92123, USA; (L.M.); (H.W.)
| | - Eric A. Pasman
- Division of Pediatric Gastroenterology, Department of Pediatrics, Naval Medical Center San Diego, San Diego, CA 92134, USA;
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
44
|
Okekunle AP, Lee H, Provido SMP, Chung GH, Hong S, Yu SH, Lee CB, Lee JE. Dietary intakes of branched-chain amino acids and plasma lipid profiles among filipino women in Korea: the Filipino Women's Diet and Health Study (FiLWHEL). Nutr J 2023; 22:34. [PMID: 37430285 DOI: 10.1186/s12937-023-00861-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/24/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND The potential role of dietary branched-chain amino acids (BCAA) in metabolic health, including cardiovascular disease and diabetes, is evolving, and it is yet to be understood if dietary BCAA intakes are associated with plasma lipid profiles or dyslipidaemia. This study tested the association of dietary BCAA intakes with plasma lipid profiles and dyslipidaemia among Filipino women in Korea. METHODS Energy-adjusted dietary BCAA intakes (isoleucine, leucine, valine, and total BCAA) and fasting blood profiles of triglycerides (TG), total cholesterol (TC), high-density lipoprotein-cholesterol (HDL-C), and low-density lipoprotein-cholesterol (LDL-C) were determined in a sample of 423 women enrolled in the Filipino Women's Diet and Health Study (FiLWHEL). The generalized linear model was applied to estimate least-square (LS) means and 95% confidence intervals (CIs) and compare plasma TG, TC, HDL-C, and LDL-C across tertile distribution of energy-adjusted dietary BCAA intakes at P < 0.05. RESULTS Mean of energy-adjusted dietary total BCAA intake was 8.3 ± 3.9 g/d. Average plasma lipid profiles were 88.5 ± 47.4 mg/dl for TG, 179.7 ± 34.5 mg/dl for TC, 58.0 ± 13.7 mg/dl for HDL-C, and 104.0 ± 30.5 mg/dl for LDL-C. LS means, and 95% CIs across tertiles of energy-adjusted total BCAA intakes were 89.9 mg/dl, 88.8 mg/dl and 85.8 mg/dl (P-trend = 0.45) for TG, 179.1 mg/dl, 183.6 mg/dl and 176.5 mg/dl (P-trend = 0.48) for TC, 57.5 mg/dl, 59.6 mg/dl and 57.1 mg/dl (P-trend = 0.75) for HDL-C and 103.6 mg/dl, 106.2 mg/dl and 102.3 mg/dl (P-trend = 0.68) for LDL-C. Furthermore, the multivariable-adjusted prevalence ratios and 95% confidence intervals for dyslipidaemia across increasing tertile distribution of energy-adjusted total BCAA intake were; 1.00, 0.67 (0.40, 1.13) and 0.45 (0.16, 1.27; P-trend = 0.03) for the first, second and third tertile, respectively. CONCLUSIONS Higher dietary intakes of BCAA presented a statistically significant inverse trend with the prevalence of dyslipidaemia among Filipino women in this study and testing these associations in longitudinal studies may be necessary to confirm these findings.
Collapse
Grants
- 2020H1D3A1A04081265 National Research Foundation of Korea
- 2020H1D3A1A04081265 National Research Foundation of Korea
- 0448A-2021077 Seoul National University Asia Center
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201300000001270 Hanmi Pharmaceutical Co., Ltd. Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
- 201600000000225 Chong Kun Dang Pharm., Seoul, Korea
Collapse
Affiliation(s)
- Akinkunmi Paul Okekunle
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea
- Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Heejin Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea
| | - Sherlyn Mae P Provido
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea
| | - Grace H Chung
- Department of Child Development & Family Studies, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea
| | - Sangmo Hong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153 Gyeongchun-ro, Guri, 11923, Korea
| | - Sung Hoon Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153 Gyeongchun-ro, Guri, 11923, Korea
| | - Chang Beom Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Hanyang University Guri Hospital, Hanyang University College of Medicine, 153 Gyeongchun-ro, Guri, 11923, Korea
| | - Jung Eun Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak- ro, Gwanak-gu, Seoul, 08826, Korea.
- Research Institute of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Korea.
| |
Collapse
|
45
|
Shao Y, Chen S, Han L, Liu J. Pharmacotherapies of NAFLD: updated opportunities based on metabolic intervention. Nutr Metab (Lond) 2023; 20:30. [PMID: 37415199 DOI: 10.1186/s12986-023-00748-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/22/2023] [Indexed: 07/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease that is becoming increasingly prevalent, and it ranges from simple steatosis to cirrhosis. However, there is still a lack of pharmacotherapeutic strategies approved by the Food and Drug Administration, which results in a higher risk of death related to carcinoma and cardiovascular complications. Of note, it is well established that the pathogenesis of NAFLD is tightly associated with whole metabolic dysfunction. Thus, targeting interconnected metabolic conditions could present promising benefits to NAFLD, according to a number of clinical studies. Here, we summarize the metabolic characteristics of the development of NAFLD, including glucose metabolism, lipid metabolism and intestinal metabolism, and provide insight into pharmacological targets. In addition, we present updates on the progresses in the development of pharmacotherapeutic strategies based on metabolic intervention globally, which could lead to new opportunities for NAFLD drug development.
Collapse
Affiliation(s)
- Yaodi Shao
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Suzhen Chen
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liu Han
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junli Liu
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
46
|
Tore EC, Eussen SJPM, Bastani NE, Dagnelie PC, Elshorbagy AK, Grootswagers P, Kožich V, Olsen T, Refsum H, Retterstøl K, Stehouwer CDA, Stolt ETK, Vinknes KJ, van Greevenbroek MMJ. The Associations of Habitual Intake of Sulfur Amino Acids, Proteins and Diet Quality with Plasma Sulfur Amino Acid Concentrations: The Maastricht Study. J Nutr 2023; 153:2027-2040. [PMID: 37164267 DOI: 10.1016/j.tjnut.2023.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/12/2023] [Accepted: 05/04/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Plasma sulfur amino acids (SAAs), i.e., methionine, total cysteine (tCys), total homocysteine (tHcy), cystathionine, total glutathione (tGSH), and taurine, are potential risk factors for obesity and cardiometabolic disorders. However, except for plasma tHcy, little is known about how dietary intake modifies plasma SAA concentrations. OBJECTIVE To investigate whether the intake of SAAs and proteins or diet quality is associated with plasma SAAs. METHODS Data from a cross-sectional subset of The Maastricht Study (n = 1145, 50.5% men, 61 interquartile range: [55, 66] y, 22.5% with prediabetes and 34.3% with type 2 diabetes) were investigated. Dietary intake was assessed using a validated food frequency questionnaire. The intake of SAAs (total, methionine, and cysteine) and proteins (total, animal, and plant) was estimated from the Dutch and Danish food composition tables. Diet quality was assessed using the Dutch Healthy Diet Index, the Mediterranean Diet Score, and the Dietary Approaches to Stop Hypertension score. Fasting plasma SAAs were measured by liquid chromatography (LC) tandem mass spectrometry (MS) (LC/MS-MS). Associations were investigated with multiple linear regressions with tertiles of dietary intake measures (main exposures) and z-standardized plasma SAAs (outcomes). RESULTS Intake of total SAAs and total proteins was positively associated with plasma tCys and cystathionine. Associations were stronger in women and in those with normal body weight. Higher intake of cysteine and plant proteins was associated with lower plasma tHcy and higher cystathionine. Higher methionine intake was associated with lower plasma tGSH, whereas cysteine intake was positively associated with tGSH. Higher intake of methionine and animal proteins was associated with higher plasma taurine. Better diet quality was consistently related to lower plasma tHcy concentrations, but it was not associated with the other SAAs. CONCLUSION Targeted dietary modifications might be effective in modifying plasma concentrations of tCys, tHcy, and cystathionine, which have been associated with obesity and cardiometabolic disorders.
Collapse
Affiliation(s)
- Elena C Tore
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands.
| | - Simone J P M Eussen
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, Maastricht University, Maastricht, the Netherlands; CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Nasser E Bastani
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Pieter C Dagnelie
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Amany K Elshorbagy
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom; Department of Physiology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Pol Grootswagers
- Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Viktor Kožich
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, and General University Hospital in Prague, Czech Republic
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Kjetil Retterstøl
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Coen DA Stehouwer
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Emma T K Stolt
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kathrine J Vinknes
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, Maastricht University, Maastricht, the Netherlands; CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
47
|
Jafari-Rastegar N, Hosseininia HS, Jalilvand E, Naseroleslami M, Khakpai F, Mousavi-Niri N. Oral administration of nano-tyrosol reversed the diabetes-induced liver damage in streptozotocin-induced diabetic rats. J Diabetes Metab Disord 2023; 22:297-305. [PMID: 37255797 PMCID: PMC10225388 DOI: 10.1007/s40200-022-01133-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/31/2022] [Accepted: 09/19/2022] [Indexed: 06/01/2023]
Abstract
Objectives The present study was designed to evaluate the effects of Tyrosol and Nano-tyrosol on the cellular arrangement, collagen disposition, protein level of insulin receptor (INSR), and superoxide dismutase (SOD) activity in both control and streptozotocin-induced diabetic rats. Methods Type 2 Diabetes (T2D) was induced in rats by a single intraperitoneal injection of streptozotocin (50 mg/kg). Experimental rats were administered Tyrosol and Nano-tyrosol 1 ml intra-gastrically at a dose of 20 mg/kg once a day for 30 days. Then, rats were sacrificed according to ethical principles. Livers were removed and processed for histological studies using the paraffin technique. Furthermore, non-paraffin sections were used for the INSR-1 western blot technique. Results At the end of the experiments, the rats in diabetic control and plain niosome groups exhibited a significant increase in collagen disposition (p < 0.001), and apoptotic cells (p < 0.001), as well as decreased total protein levels of INSR (p < 0.001), and SOD activity (p < 0.001) in the hepatic cells. Oral administration of Tyrosol and Nano-tyrosol to diabetic rats reversed all the above-mentioned parameters to near normal levels (p < 0.001). Nano-tyrosol showed the highest significant effect rather than Tyrosol. Conclusion The results of the present study suggested the beneficial effects of Tyrosol and especially Nano-tyrosol on decreasing the adverse effects of diabetes.
Collapse
Affiliation(s)
- Nima Jafari-Rastegar
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Herbal pharmacology research center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Haniyeh-Sadat Hosseininia
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Herbal pharmacology research center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elahe Jalilvand
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Herbal pharmacology research center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Naseroleslami
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fateme Khakpai
- Cognitive and Neuroscience Research Center (CNRC), Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Neda Mousavi-Niri
- Department of Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, P.O.Box: 193951495, Tehran, Iran
| |
Collapse
|
48
|
Bischoff SC, Ockenga J, Eshraghian A, Barazzoni R, Busetto L, Campmans-Kuijpers M, Cardinale V, Chermesh I, Kani HT, Khannoussi W, Lacaze L, Léon-Sanz M, Mendive JM, Müller MW, Tacke F, Thorell A, Vranesic Bender D, Weimann A, Cuerda C. Practical guideline on obesity care in patients with gastrointestinal and liver diseases - Joint ESPEN/UEG guideline. Clin Nutr 2023; 42:987-1024. [PMID: 37146466 DOI: 10.1016/j.clnu.2023.03.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Patients with chronic gastrointestinal disease such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), celiac disease, gastroesophageal reflux disease (GERD), pancreatitis, and chronic liver disease (CLD) often suffer from obesity because of coincidence (IBD, IBS, celiac disease) or related pathophysiology (GERD, pancreatitis and CLD). It is unclear if such patients need a particular diagnostic and treatment that differs from the needs of lean gastrointestinal patients. The present guideline addresses this question according to current knowledge and evidence. OBJECTIVE The present practical guideline is intended for clinicians and practitioners in general medicine, gastroenterology, surgery and other obesity management, including dietitians and focuses on obesity care in patients with chronic gastrointestinal diseases. METHODS The present practical guideline is the shortened version of a previously published scientific guideline developed according to the standard operating procedure for ESPEN guidelines. The content has been re-structured and transformed into flow-charts that allow a quick navigation through the text. RESULTS In 100 recommendations (3× A, 33× B, 24 × 0, 40× GPP, all with a consensus grade of 90% or more) care of gastrointestinal patients with obesity - including sarcopenic obesity - is addressed in a multidisciplinary way. A particular emphasis is on CLD, especially metabolic associated liver disease, since such diseases are closely related to obesity, whereas liver cirrhosis is rather associated with sarcopenic obesity. A special chapter is dedicated to obesity care in patients undergoing bariatric surgery. The guideline focuses on adults, not on children, for whom data are scarce. Whether some of the recommendations apply to children must be left to the judgment of the experienced pediatrician. CONCLUSION The present practical guideline offers in a condensed way evidence-based advice how to care for patients with chronic gastrointestinal diseases and concomitant obesity, an increasingly frequent constellation in clinical practice.
Collapse
Affiliation(s)
- Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Johann Ockenga
- Medizinische Klinik II, Klinikum Bremen-Mitte, Bremen FRG, Bremen, Germany.
| | - Ahad Eshraghian
- Department of Gastroenterology and Hepatology, Avicenna Hospital, Shiraz, Iran.
| | - Rocco Barazzoni
- Department of Medical, Technological and Translational Sciences, University of Trieste, Ospedale di Cattinara, Trieste, Italy.
| | - Luca Busetto
- Department of Medicine, University of Padova, Padova, Italy.
| | - Marjo Campmans-Kuijpers
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, the Netherlands.
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Irit Chermesh
- Department of Gastroenterology, Rambam Health Care Campus, Affiliated with Technion-Israel Institute of Technology, Haifa, Israel.
| | - Haluk Tarik Kani
- Department of Gastroenterology, Marmara University, School of Medicine, Istanbul, Turkey.
| | - Wafaa Khannoussi
- Hepato-Gastroenterology Department, Mohammed VI University Hospital, Oujda, Morocco; and Laboratoire de Recherche des Maladies Digestives (LARMAD), Mohammed the First University, Oujda, Morocco.
| | - Laurence Lacaze
- Department of General Surgery, Mantes-la-Jolie Hospital, Mantes-la-Jolie, France.
| | - Miguel Léon-Sanz
- Department of Endocrinology and Nutrition, University Hospital Doce de Octubre, Medical School, University Complutense, Madrid, Spain.
| | - Juan M Mendive
- La Mina Primary Care Academic Health Centre, Catalan Institute of Health (ICS), University of Barcelona, Barcelona, Spain.
| | - Michael W Müller
- Department of General and Visceral Surgery, Regionale Kliniken Holding, Kliniken Ludwigsburg-Bietigheim gGmbH, Krankenhaus Bietigheim, Bietigheim-Bissingen, Germany.
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| | - Anders Thorell
- Department of Clinical Science, Danderyds Hospital, Karolinska Institutet & Department of Surgery, Ersta Hospital, Stockholm, Sweden.
| | - Darija Vranesic Bender
- Unit of Clinical Nutrition, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Arved Weimann
- Department of General, Visceral and Oncological Surgery, St. George Hospital, Leipzig, Germany.
| | - Cristina Cuerda
- Departamento de Medicina, Universidad Complutense de Madrid, Nutrition Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
49
|
Tsamos G, Vasdeki D, Koufakis T, Michou V, Makedou K, Tzimagiorgis G. Therapeutic Potentials of Reducing Liver Fat in Non-Alcoholic Fatty Liver Disease: Close Association with Type 2 Diabetes. Metabolites 2023; 13:metabo13040517. [PMID: 37110175 PMCID: PMC10141666 DOI: 10.3390/metabo13040517] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), the most widespread chronic liver disease worldwide, confers a significant burden on health systems and leads to increased mortality and morbidity through several extrahepatic complications. NAFLD comprises a broad spectrum of liver-related disorders, including steatosis, cirrhosis, and hepatocellular carcinoma. It affects almost 30% of adults in the general population and up to 70% of people with type 2 diabetes (T2DM), sharing common pathogenetic pathways with the latter. In addition, NAFLD is closely related to obesity, which acts in synergy with other predisposing conditions, including alcohol consumption, provoking progressive and insidious liver damage. Among the most potent risk factors for accelerating the progression of NAFLD to fibrosis or cirrhosis, diabetes stands out. Despite the rapid rise in NAFLD rates, identifying the optimal treatment remains a challenge. Interestingly, NAFLD amelioration or remission appears to be associated with a lower risk of T2DM, indicating that liver-centric therapies could reduce the risk of developing T2DM and vice versa. Consequently, assessing NAFLD requires a multidisciplinary approach to identify and manage this multisystemic clinical entity early. With the continuously emerging new evidence, innovative therapeutic strategies are being developed for the treatment of NAFLD, prioritizing a combination of lifestyle changes and glucose-lowering medications. Based on recent evidence, this review scrutinizes all practical and sustainable interventions to achieve a resolution of NAFLD through a multimodal approach.
Collapse
Affiliation(s)
- Georgios Tsamos
- Division of Gastroenterology, Norfolk and Norwich University Hospital, Norwich NR4 7UY, UK
| | - Dimitra Vasdeki
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theocharis Koufakis
- Division of Endocrinology and Metabolism and Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Vassiliki Michou
- Sports Medicine Laboratory, School of Physical Education & Sport Science, Aristotle University of Thessaloniki, 57001 Thessaloniki, Greece
| | - Kali Makedou
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Georgios Tzimagiorgis
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, 54636 Thessaloniki, Greece
| |
Collapse
|
50
|
Zhang J, Schäfer SM, Kabisch S, Csanalosi M, Schuppelius B, Kemper M, Markova M, Meyer NMT, Pivovarova-Ramich O, Keyhani-Nejad F, Rohn S, Pfeiffer AFH. Implication of sugar, protein and incretins in excessive glucagon secretion in type 2 diabetes after mixed meals. Clin Nutr 2023; 42:467-476. [PMID: 36857956 DOI: 10.1016/j.clnu.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 01/10/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023]
Abstract
AIMS Amino acids powerfully release glucagon but their contribution to postprandial hyperglucagonemia in type 2 diabetes remains unclear. Exogenously applied GIP stimulates, while GLP-1 inhibits, glucagon secretion in humans. However, their role in mixed meals is unclear, which we therefore characterized. METHODS In three experiments, participants with type 2 diabetes and obese controls randomly received different loads of sugars and/or proteins. In the first experiment, participants ingested the rapidly cleaved saccharose (SAC) or slowly cleaved isomaltulose (ISO) which is known to elicit opposite profiles of GIP and GLP-1 secretion. In the second one participants received test meals which contained saccharose or isomaltulose in combination with milk protein. The third set of participants underwent randomized oral protein tests with whey protein or casein. Incretins, glucagon, C-peptide, and insulin were profiled by specific immunological assays. RESULTS 50 g of the sugars alone suppressed glucagon in controls but slightly less in type 2 diabetes patients. Participants with type 2 diabetes showed excessive glucagon responses within 15 min and lasting over 3 h, while the obese controls showed small initial and delayed greater glucagon responses to mixed meals. The release of GIP was significantly faster and greater with SAC compared to ISO, while GLP-1 showed an inverse pattern. The glucagon responses to whey or casein were only moderately increased in type 2 diabetes patients without a left shift of the dose response curve. CONCLUSIONS The rapid hypersecretion of glucagon after mixed meals in type 2 diabetes patients compared to controls is unaffected by endogenous incretins. The defective suppression of glucagon by glucose combined with hypersecretion to protein is required for the exaggerated response. CLINICAL TRIALS NUMBERS NCT03806920, NCT02219295, NCT04564391.
Collapse
Affiliation(s)
- Jiudan Zhang
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Sylva Mareike Schäfer
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Institute of Nutritional Science, Justus-Liebig University of Giessen, Giessen, Germany
| | - Stefan Kabisch
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Marta Csanalosi
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Bettina Schuppelius
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Margrit Kemper
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Mariya Markova
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Nina Marie Tosca Meyer
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Olga Pivovarova-Ramich
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany; Reseach Group Molecular Nutritional Medicine, Dept. of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558, Nuthetal, Germany
| | - Farnaz Keyhani-Nejad
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Sascha Rohn
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Grindelallee 117, 20146, Hamburg, Germany; Institute of Food Technology and Food Chemistry, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Andreas F H Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| |
Collapse
|